Microbiota Effect on Trimethylamine N-Oxide Production: From Cancer to Fitness—A Practical Preventing Recommendation and Therapies
Abstract
:1. Introduction
2. TMAO in Physiological Conditions
3. TMAO in Pathological Conditions
3.1. TMAO in Atherosclerosis and Cardiovascular Disease
3.2. TMAO in Chronic Kidney Disease
3.3. TMAO and Type II Diabetes
3.4. TMAO, Microbiota Homeostasis and Cancer
4. Practical Recommendations for Prevention and Treatments Reducing TMAO Production
4.1. Diet
4.2. Drugs, Supplements and Physical Activity
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Oellgaard, J.; Winther, S.A.; Hansen, T.S.; Rossing, P.; von Scholten, B.J. Trimethylamine N-oxide (TMAO) as a New Potential Therapeutic Target for Insulin Resistance and Cancer. Curr. Pharm. Des. 2017, 23, 3699–3712. [Google Scholar] [CrossRef] [PubMed]
- Tang, W.H.W.; Hazen, S.L. Microbiome, trimethylamine N-oxide, and cardiometabolic disease. Transl. Res. 2017, 179, 108–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Janeiro, M.H.; Ramírez, M.J.; Milagro, F.I.; Martínez, J.A.; Solas, M. Implication of trimethylamine n-oxide (TMAO) in disease: Potential biomarker or new therapeutic target. Nutrients 2018, 10, 1398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bain, M.; Fornasini, G.; Evans, A. Trimethylamine: Metabolic, Pharmacokinetic and Safety Aspects. Curr. Drug Metab. 2005, 6, 227–240. [Google Scholar] [CrossRef]
- Ahmadmehrabi, S.; Tang, W.H.W. Gut microbiome and its role in cardiovascular diseases. Curr. Opin. Cardiol. 2017, 32, 761–766. [Google Scholar] [CrossRef] [Green Version]
- Jie, Z.; Xia, H.; Zhong, S.L.; Feng, Q.; Li, S.; Liang, S.; Zhong, H.; Liu, Z.; Gao, Y.; Zhao, H.; et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017, 8, 845. [Google Scholar] [CrossRef] [Green Version]
- Zeisel, S.H.; Warrier, M. Trimethylamine N-Oxide, the Microbiome, and Heart and Kidney Disease. Annu. Rev. Nutr. 2017, 37, 157–181. [Google Scholar] [CrossRef]
- Wiedeman, A.M.; Barr, S.I.; Green, T.J.; Xu, Z.; Innis, S.M.; Kitts, D.D. Dietary choline intake: Current state of knowledge across the life cycle. Nutrients 2018, 10, 1513. [Google Scholar] [CrossRef] [Green Version]
- Zeisel, S.H.; Da Costa, K.A. Choline: An essential nutrient for public health. Nutr. Rev. 2009, 67, 615–623. [Google Scholar] [CrossRef] [Green Version]
- Fennema, D.; Phillips, I.R.; Shephard, E.A. 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, 1839–1850. [Google Scholar] [CrossRef]
- Zhu, Y.; Jameson, E.; Crosatti, M.; Schäfer, H.; Rajakumar, K.; Bugg, T.D.H.; Chen, Y. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc. Natl. Acad. Sci. USA 2014, 111, 4268–4273. [Google Scholar] [CrossRef] [Green Version]
- Koeth, R.A.; Levison, B.S.; Culley, M.K.; Buffa, J.A.; Wang, Z.; Gregory, J.C.; Org, E.; Wu, Y.; Li, L.; Smith, J.D.; et al. γ-butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab. 2014, 20, 799–812. [Google Scholar] [CrossRef] [Green Version]
- Akerman, B.R.; Lemass, H.; Chow, L.M.L.; Lambert, D.M.; Greenberg, C.; Bibeau, C.; Mamer, O.A.; Treacy, E.P. Trimethylaminuria is caused by mutations of the FMO3 gene in a North American cohort. Mol. Genet. Metab. 1999, 68, 24–31. [Google Scholar] [CrossRef]
- Treacy, E.P.; Akerman, B.R.; Chow, L.M.L.; Youil, R.; Bibeau, C.; Lin, J.; Bruce, A.G.; Knight, M.; Danks, D.M.; Cashman, J.R.; et al. Mutations of the flavin-containing monooxygenase gene (FMO3) cause trimethylaminuria, a defect in detoxication. Hum. Mol. Genet. 1998, 7, 839–845. [Google Scholar] [CrossRef]
- Bennett, B.J.; Vallim, T.Q.D.A.; Wang, Z.; Shih, D.M.; Meng, Y.; Gregory, J.; Allayee, H.; Lee, R.; Graham, M.; Crooke, R.; et al. Trimethylamine-N-Oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013, 17, 49–60. [Google Scholar] [CrossRef] [Green Version]
- Busby, M.G.; Fischer, L.; Da Costa, K.A.; Thompson, D.; Mar, M.H.; Zeisel, S.H. Choline- and betaine-defined diets for use in clinical research and for the management of trimethylaminuria. J. Am. Diet. Assoc. 2004, 104, 1836–1845. [Google Scholar] [CrossRef]
- Brugère, J.F.; Borrel, G.; Gaci, N.; Tottey, W.; O’Toole, P.W.; Malpuech-Brugère, C. Archaebiotics: Proposed therapeutic use of archaea to prevent trimethylaminuria and cardiovascular disease. Gut Microbes 2013, 5, 5–10. [Google Scholar] [CrossRef] [Green Version]
- Sawicka, A.K.; Renzi, G.; Olek, R.A. The bright and the dark sides of L-carnitine supplementation: A systematic review. J. Int. Soc. Sports Nutr. 2020, 17, 1–10. [Google Scholar] [CrossRef]
- Heianza, Y.; Ma, W.; Manson, J.A.E.; Rexrode, K.M.; Qi, L. Gut microbiota metabolites and risk of major adverse cardiovascular disease events and death: A systematic review and meta-analysis of prospective studies. J. Am. Heart Assoc. 2017, 6, 4947. [Google Scholar] [CrossRef]
- Schiattarella, G.G.; Sannino, A.; Toscano, E.; Giugliano, G.; Gargiulo, G.; Franzone, A.; Trimarco, B.; Esposito, G.; Perrino, C. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: A systematic review and dose-response meta-analysis. Eur. Heart J. 2017, 38, 2948–2956. [Google Scholar] [CrossRef]
- Samulak, J.J.; Sawicka, A.K.; Hartmane, D.; Grinberga, S.; Pugovics, O.; Lysiak-Szydlowska, W.; Olek, R.A. L-Carnitine supplementation increases trimethylamine-N-oxide but not markers of atherosclerosis in healthy aged women. Ann. Nutr. Metab. 2019, 74, 11–17. [Google Scholar] [CrossRef] [PubMed]
- Olek, R.A.; Samulak, J.J.; Sawicka, A.K.; Hartmane, D.; Grinberga, S.; Pugovics, O.; Lysiak-Szydlowska, W. Increased Trimethylamine N-Oxide Is Not Associated with Oxidative Stress Markers in Healthy Aged Women. Oxid. Med. Cell. Longev. 2019, 2019, 6247169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bordoni, L.; Sawicka, A.K.; Szarmach, A.; Winklewski, P.J.; Olek, R.A.; Gabbianelli, R. A pilot study on the effects of L-carnitine and trimethylamine-N-oxide on platelet mitochondrial dna methylation and CVD biomarkers in aged women. Int. J. Mol. Sci. 2020, 21, 1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fukami, K.; Yamagishi, S.I.; Sakai, K.; Kaida, Y.; Yokoro, M.; Ueda, S.; Wada, Y.; Takeuchi, M.; Shimizu, M.; Yamazaki, H.; et al. Oral L-carnitine supplementation increases trimethylamine-N-oxide but reduces markers of vascular injury in hemodialysis patients. J. Cardiovasc. Pharmacol. 2015, 65, 289–295. [Google Scholar] [CrossRef] [PubMed]
- Vallance, H.D.; Koochin, A.; Branov, J.; Rosen-Heath, A.; Bosdet, T.; Wang, Z.; Hazen, S.L.; Horvath, G. Marked elevation in plasma trimethylamine-N-oxide (TMAO) in patients with mitochondrial disorders treated with oral L-carnitine. Mol. Genet. Metab. Rep. 2018, 15, 130–133. [Google Scholar] [CrossRef]
- Wallace, T.C.; Blusztajn, J.K.; Caudill, M.A.; Klatt, K.C.; Natker, E.; Zeisel, S.H.; Zelman, K.M. The underconsumed and underappreciated essential nutrient. Nutr. Today 2018, 53, 240–253. [Google Scholar] [CrossRef]
- Zeisel, S.H.; Mar, M.H.; Howe, J.C.; Holden, J.M. Concentrations of choline-containing compounds and betaine in common foods. J. Nutr. 2003, 133, 1302–1307. [Google Scholar] [CrossRef] [Green Version]
- Schugar, R.C.; Brown, J.M. Emerging roles of flavin monooxygenase 3 in cholesterol metabolism and atherosclerosis. Curr. Opin. Lipidol. 2015, 26, 426–431. [Google Scholar] [CrossRef] [Green Version]
- Chhibber-Goel, J.; Gaur, A.; Singhal, V.; Parakh, N.; Bhargava, B.; Sharma, A. The complex metabolism of trimethylamine in humans: Endogenous and exogenous sources. Expert Rev. Mol. Med. 2016, 18, e8. [Google Scholar] [CrossRef]
- Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef]
- Tremaroli, V.; Bäckhed, F. Functional interactions between the gut microbiota and host metabolism. Nature 2012, 489, 242–249. [Google Scholar] [CrossRef]
- Kamada, N.; Seo, S.U.; Chen, G.Y.; Núñez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 2013, 13, 321–335. [Google Scholar] [CrossRef]
- Yang, S.; Dai, H.; Lu, Y.; Li, R.; Gao, C.; Pan, S. Trimethylamine N-Oxide Promotes Cell Proliferation and Angiogenesis in Colorectal Cancer. J. Immunol. Res. 2022, 2022, 7043856. [Google Scholar] [CrossRef]
- Craciun, S.; Balskus, E.P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl. Acad. Sci. USA 2012, 109, 21307–21312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartiala, J.; Bennett, B.J.; Tang, W.H.W.; Wang, Z.; Stewart, A.F.R.; Roberts, R.; McPherson, R.; Lusis, A.J.; Hazen, S.L.; Allayee, H. Comparative genome-wide association studies in mice and humans for trimethylamine N-Oxide, a proatherogenic metabolite of choline and L-carnitine. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1307–1313. [Google Scholar] [CrossRef] [Green Version]
- Obeid, R.; Awwad, H.M.; Rabagny, Y.; Graeber, S.; Herrmann, W.; Geisel, J. Plasma trimethylamine N-oxide concentration is associated with choline, phospholipids, and methyl metabolism. Am. J. Clin. Nutr. 2016, 103, 703–711. [Google Scholar] [CrossRef] [Green Version]
- Aslibekyan, S.; Irvin, M.R.; Hidalgo, B.A.; Perry, R.T.; Jeyarajah, E.J.; Garcia, E.; Shalaurova, I.; Hopkins, P.N.; Province, M.A.; Tiwari, H.K.; et al. Genome- and CD4+ T-cell methylome-wide association study of circulating trimethylamine-N-oxide in the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN). J. Nutr. Intermed. Metab. 2017, 8, 1–7. [Google Scholar] [CrossRef]
- Senthong, V.; Wang, Z.; Fan, Y.; Wu, Y.; Hazen, S.L.; Tang, W.H.W. Trimethylamine N-oxide and mortality risk in patients with peripheral artery disease. J. Am. Heart Assoc. 2016, 5, e4237. [Google Scholar] [CrossRef]
- Randrianarisoa, E.; Lehn-Stefan, A.; Wang, X.; Hoene, M.; Peter, A.; Heinzmann, S.S.; Zhao, X.; Königsrainer, I.; Königsrainer, A.; Balletshofer, B.; et al. Relationship of serum trimethylamine N-oxide (TMAO) levels with early atherosclerosis in humans. Sci. Rep. 2016, 6, 26745. [Google Scholar] [CrossRef] [Green Version]
- Nie, J.; Xie, L.; Zhao, B.X.; Li, Y.; Qiu, B.; Zhu, F.; Li, G.F.; He, M.; Wang, Y.; Wang, B.; et al. Serum trimethylamine N-oxide concentration is positively associated with first stroke in hypertensive patients. Stroke 2018, 49, 2021–2028. [Google Scholar] [CrossRef]
- Esposito, T.; Varriale, B.; D’Angelo, R.; Amato, A.; Sidoti, A. Regulation of flavin-containing mono-oxygenase (Fmo3) gene expression by steroids in mice and humans. Horm. Mol. Biol. Clin. Investig. 2014, 20, 99–109. [Google Scholar] [CrossRef] [PubMed]
- Manor, O.; Zubair, N.; Conomos, M.P.; Xu, X.; Rohwer, J.E.; Krafft, C.E.; Lovejoy, J.C.; Magis, A.T. A Multi-omic Association Study of Trimethylamine N-Oxide. Cell Rep. 2018, 24, 935–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kühn, T.; Rohrmann, S.; Sookthai, D.; Johnson, T.; Katzke, V.; Kaaks, R.; Von Eckardstein, A.; Müller, D. Intra-individual variation of plasma trimethylamine-N-oxide (TMAO), betaine and choline over 1 year. Clin. Chem. Lab. Med. 2017, 55, 261–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rohrmann, S.; Linseisen, J.; Allenspach, M.; Von Eckardstein, A.; Müller, D. Plasma concentrations of trimethylamine- n-oxide are directly associated with dairy food consumption and low-grade inflammation in a german adult population. J. Nutr. 2016, 146, 283–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krüger, R.; Merz, B.; Rist, M.J.; Ferrario, P.G.; Bub, A.; Kulling, S.E.; Watzl, B. Associations of current diet with plasma and urine TMAO in the KarMeN study: Direct and indirect contributions. Mol. Nutr. Food Res. 2017, 61, 1700363. [Google Scholar] [CrossRef]
- Romano, K.A.; Vivas, E.I.; Amador-Noguez, D.; Rey, F.E. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio 2015, 6, e02481-14. [Google Scholar] [CrossRef] [Green Version]
- Falony, G.; Vieira-Silva, S.; Raes, J. Microbiology Meets Big Data: The Case of Gut Microbiota-Derived Trimethylamine. Annu. Rev. Microbiol. 2015, 69, 305–321. [Google Scholar] [CrossRef]
- Tang, W.H.W.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [Green Version]
- Cho, C.E.; Taesuwan, S.; Malysheva, O.V.; Bender, E.; Tulchinsky, N.F.; Yan, J.; Sutter, J.L.; Caudill, M.A. 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. Mol. Nutr. Food Res. 2017, 61, 324. [Google Scholar] [CrossRef]
- Biagi, E.; Franceschi, C.; Rampelli, S.; Severgnini, M.; Ostan, R.; Turroni, S.; Consolandi, C.; Quercia, S.; Scurti, M.; Monti, D.; et al. Gut Microbiota and Extreme Longevity. Curr. Biol. 2016, 26, 1480–1485. [Google Scholar] [CrossRef]
- Santoro, A.; Ostan, R.; Candela, M.; Biagi, E.; Brigidi, P.; Capri, M.; Franceschi, C. Gut microbiota changes in the extreme decades of human life: A focus on centenarians. Cell. Mol. Life Sci. 2018, 75, 129–148. [Google Scholar] [CrossRef] [Green Version]
- Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
- Ley, R.E. Gut microbiota in 2015: Prevotella in the gut: Choose carefully. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 69–70. [Google Scholar] [CrossRef]
- Velasquez, M.T.; Ramezani, A.; Manal, A.; Raj, D.S. Trimethylamine N-oxide: The good, the bad and the unknown. Toxins 2016, 8, 326. [Google Scholar] [CrossRef] [Green Version]
- Landfald, B.; Valeur, J.; Berstad, A.; Raa, J. Microbial trimethylamine- N -oxide as a disease marker: Something fishy? Microb. Ecol. Health Dis. 2017, 28, 1327309. [Google Scholar] [CrossRef] [Green Version]
- Cheung, W.; Keski-Rahkonen, P.; Assi, N.; Ferrari, P.; Freisling, H.; Rinaldi, S.; Slimani, N.; Zamora-Ros, R.; Rundle, M.; Frost, G.; et al. A metabolomic study of biomarkers of meat and fish intake. Am. J. Clin. Nutr. 2017, 105, 600–608. [Google Scholar] [CrossRef] [Green Version]
- Barton, W.; Penney, N.C.; Cronin, O.; Garcia-Perez, I.; Molloy, M.G.; Holmes, E.; Shanahan, F.; Cotter, P.D.; O’Sullivan, O. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut 2018, 67, 625–633. [Google Scholar] [CrossRef]
- Penry, J.T.; Manore, M.M. Choline: An important micronutrient for maximal endurance-exercise performance? Int. J. Sport Nutr. Exerc. Metab. 2008, 18, 191–203. [Google Scholar] [CrossRef] [Green Version]
- Rogerson, D. Vegan diets: Practical advice for athletes and exercisers. J. Int. Soc. Sports Nutr. 2017, 14, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Hamaker, B.R.; Tuncil, Y.E. A perspective on the complexity of dietary fiber structures and their potential effect on the gut microbiota. J. Mol. Biol. 2014, 426, 3838–3850. [Google Scholar] [CrossRef]
- Sharma, P.; Bhandari, C.; Kumar, S.; Sharma, B.; Bhadwal, P.; Agnihotri, N. Dietary fibers: A way to a healthy microbiome. In Diet, Microbiome and Health; Elsever: Amsterdam, The Netherlands, 2018; ISBN 9780128114407. [Google Scholar]
- Wołyniec, W.; Kasprowicz, K.; Giebułtowicz, J.; Korytowska, N.; Zorena, K.; Bartoszewicz, M.; Rita-Tkachenko, P.; Renke, M.; Ratkowski, W. Changes in water soluble uremic toxins and urinary acute kidney injury biomarkers after 10-and 100-km runs. Int. J. Environ. Res. Public Health 2019, 16, 4153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schranner, D.; Kastenmüller, G.; Schönfelder, M.; Römisch-Margl, W.; Wackerhage, H. Metabolite Concentration Changes in Humans After a Bout of Exercise: A Systematic Review of Exercise Metabolomics Studies. Sport. Med.-Open 2020, 6, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kerksick, C.M.; Wilborn, C.D.; Roberts, M.D.; Smith-Ryan, A.; Kleiner, S.M.; Jäger, R.; Collins, R.; Cooke, M.; Davis, J.N.; Galvan, E.; et al. ISSN exercise & sports nutrition review update: Research & recommendations. J. Int. Soc. Sports Nutr. 2018, 5, 38. [Google Scholar]
- Hughes, R.L. A Review of the Role of the Gut Microbiome in Personalized Sports Nutrition. Front. Nutr. 2020, 6, 191. [Google Scholar] [CrossRef]
- Ekelund, U.; Ward, H.A.; Norat, T.; Luan, J.; May, A.M.; Weiderpass, E.; Sharp, S.J.; Overvad, K.; Østergaard, J.N.; Tjønneland, A.; et al. Physical activity and all-cause mortality across levels of overall and abdominal adiposity in European men and women: The European prospective investigation into cancer and nutrition study (EPIC). Am. J. Clin. Nutr. 2015, 101, 613–621. [Google Scholar] [CrossRef] [Green Version]
- Kyu, H.H.; Bachman, V.F.; Alexander, L.T.; Mumford, J.E.; Afshin, A.; Estep, K.; Veerman, J.L.; Delwiche, K.; Iannarone, M.L.; Moyer, M.L.; et al. Physical activity and risk of breast cancer, colon cancer, diabetes, ischemic heart disease, and ischemic stroke events: Systematic review and dose-response meta-analysis for the Global Burden of Disease Study 2013. BMJ 2016, 354, i3857. [Google Scholar] [CrossRef] [Green Version]
- Wolin, K.Y.; Yan, Y.; Colditz, G.A.; Lee, I.M. Physical activity and colon cancer prevention: A meta-analysis. Br. J. Cancer 2009, 100, 611–616. [Google Scholar] [CrossRef]
- World Cancer Research Fund/American Institute for Cancer Research. Diet, Nutrition, Physical Activity and Cancer: A Global Perspective; World Cancer Research Fund/American Institute for Cancer Research: Washington, DC, USA, 2018; ISBN 9781912259465. [Google Scholar]
- Fuchs, R. Physical Activity and Health. In International Encyclopedia of the Social & Behavioral Sciences, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2015; ISBN 9780080970875. [Google Scholar]
- Saint-Maurice, P.F.; Coughlan, D.; Kelly, S.P.; Keadle, S.K.; Cook, M.B.; Carlson, S.A.; Fulton, J.E.; Matthews, C.E. Association of Leisure-Time Physical Activity Across the Adult Life Course with All-Cause and Cause-Specific Mortality. JAMA Netw. Open 2019, 2, e190355. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Wang, Y.; Ke, B.; Du, J. TMAO: How gut microbiota contributes to heart failure. Transl. Res. 2021, 228, 109–125. [Google Scholar] [CrossRef]
- Mueller, D.M.; Allenspach, M.; Othman, A.; Saely, C.H.; Muendlein, A.; Vonbank, A.; Drexel, H.; von Eckardstein, A. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis 2015, 243, 638–644. [Google Scholar] [CrossRef]
- Hansen, T.H.; Gøbel, R.J.; Hansen, T.; Pedersen, O. The gut microbiome in cardio-metabolic health. Genome Med. 2015, 7, 33. [Google Scholar] [CrossRef]
- Wu, S.; Rhee, K.J.; Albesiano, E.; Rabizadeh, S.; Wu, X.; Yen, H.R.; Huso, D.L.; Brancati, F.L.; Wick, E.; McAllister, F.; et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat. Med. 2009, 15, 1016–1022. [Google Scholar] [CrossRef] [PubMed]
- Bae, S.; Ulrich, C.M.; Neuhouser, M.L.; Malysheva, O.; Bailey, L.B.; Xiao, L.; Brown, E.C.; Cushing-Haugen, K.L.; Zheng, Y.; Cheng, T.Y.D.; et al. Plasma choline metabolites and colorectal cancer risk in the women’s health initiative observational study. Cancer Res. 2014, 74, 7442–7452. [Google Scholar] [CrossRef] [Green Version]
- Xu, R.; Wang, Q.Q.; Li, L. A genome-wide systems analysis reveals strong link between colorectal cancer and trimethylamine N-oxide (TMAO), a gut microbial metabolite of dietary meat and fat. BMC Genom. 2015, 16, S4. [Google Scholar] [CrossRef] [Green Version]
- Chan, C.W.H.; Law, B.M.H.; Waye, M.M.Y.; Chan, J.Y.W.; Wei So, W.K.; Chow, K.M. Trimethylamine-N-oxide as one hypothetical link for the relationship between intestinal microbiota and cancer-Where we are and where shall we go? J. Cancer 2019, 10, 5874. [Google Scholar] [CrossRef]
- Li, X.S.; Obeid, S.; Klingenberg, R.; Gencer, B.; Mach, F.; Räber, L.; Windecker, S.; Rodondi, N.; Nanchen, D.; Muller, O.; et al. Gutmicrobiota-dependent trimethylamine N-oxide in acute coronary syndromes: A prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur. Heart J. 2017, 38, 814–824. [Google Scholar] [CrossRef] [Green Version]
- Senthong, V.; Wang, Z.; Li, X.S.; Fan, Y.; Wu, Y.; Tang, W.H.W.; Hazen, S.L. Intestinal microbiota-generated metabolite Trimethylamine-N-oxide and 5-year mortality risk in stable coronary artery disease: The contributory role of intestinal microbiota in a COURAGE-like patient cohort. J. Am. Heart Assoc. 2016, 5, e002816. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [Green Version]
- Chistiakov, D.A.; Melnichenko, A.A.; Myasoedova, V.A.; Grechko, A.V.; Orekhov, A.N. Mechanisms of foam cell formation in atherosclerosis. J. Mol. Med. 2017, 95, 1153–1165. [Google Scholar] [CrossRef]
- Thon, M.P.; Hemmler, A.; Glinzer, A.; Mayr, M.; Wildgruber, M.; Zernecke-Madsen, A.; Gee, M.W. A multiphysics approach for modeling early atherosclerosis. Biomech. Model. Mechanobiol. 2018, 17, 617–644. [Google Scholar] [CrossRef] [Green Version]
- Geng, J.; Yang, C.; Wang, B.; Zhang, X.; Hu, T.; Gu, Y.; Li, J. Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomed. Pharmacother. 2018, 97, 941–947. [Google Scholar] [CrossRef] [PubMed]
- Mohammadi, A.; Najar, A.G.; Yaghoobi, M.M.; Jahani, Y.; Vahabzadeh, Z. Trimethylamine-N-Oxide Treatment Induces Changes in the ATP-Binding Cassette Transporter A1 and Scavenger Receptor A1 in Murine Macrophage J774A.1 cells. Inflammation 2016, 39, 393–404. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Li, Q.; Jiang, H. Gut microbiota in atherosclerosis: Focus on trimethylamine N-oxide. Apmis 2020, 128, 353–366. [Google Scholar] [CrossRef] [Green Version]
- Sun, X.; Jiao, X.; Ma, Y.; Liu, Y.; Zhang, L.; He, Y.; Chen, Y. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem. Biophys. Res. Commun. 2016, 481, 63–70. [Google Scholar] [CrossRef]
- Ren, D.; Liu, Y.; Zhao, Y.; Yang, X. Hepatotoxicity and endothelial dysfunction induced by high choline diet and the protective effects of phloretin in mice. Food Chem. Toxicol. 2016, 94, 203–212. [Google Scholar] [CrossRef]
- Chou, R.H.; Chen, C.Y.; Chen, I.C.; Huang, H.L.; Lu, Y.W.; Kuo, C.S.; Chang, C.C.; Huang, P.H.; Chen, J.W.; Lin, S.J. Trimethylamine N-Oxide, Circulating Endothelial Progenitor Cells, and Endothelial Function in Patients with Stable Angina. Sci. Rep. 2019, 9, 4249. [Google Scholar] [CrossRef] [Green Version]
- Ingram, D.A.; Mead, L.E.; Tanaka, H.; Meade, V.; Fenoglio, A.; Mortell, K.; Pollok, K.; Ferkowicz, M.J.; Gilley, D.; Yoder, M.C. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood 2004, 104, 2752–2760. [Google Scholar] [CrossRef]
- Basile, D.P.; Yoder, M.C. Circulating and Tissue Resident Endothelial Progenitor Cells. J. Cell. Physiol. 2014, 229, 10–16. [Google Scholar] [CrossRef] [Green Version]
- Chopra, H.; Hung, M.K.; Kwong, D.L.; Zhang, C.F.; Pow, E.H.N. Insights into endothelial progenitor cells: Origin, classification, potentials, and prospects. Stem Cells Int. 2018, 2018, 9847015. [Google Scholar] [CrossRef] [Green Version]
- Prater, D.N.; Case, J.; Ingram, D.A.; Yoder, M.C. Working hypothesis to redefine endothelial progenitor cells. Leukemia 2007, 21, 1141–1149. [Google Scholar] [CrossRef] [Green Version]
- Hur, J.; Yoon, C.H.; Kim, H.S.; Choi, J.H.; Kang, H.J.; Hwang, K.K.; Oh, B.H.; Lee, M.M.; Park, Y.B. Characterization of Two Types of Endothelial Progenitor Cells and Their Different Contributions to Neovasculogenesis. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 288–293. [Google Scholar] [CrossRef] [PubMed]
- Davignon, J.; Ganz, P. Role of endothelial dysfunction in atherosclerosis. Circulation 2004, 109, III27–III32. [Google Scholar] [CrossRef] [Green Version]
- Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.F.; Wang, Z.; Hazen, S.L.; Lusis, A.J.; Shih, D.M. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κb. J. Am. Heart Assoc. 2016, 5, e002767. [Google Scholar] [CrossRef] [Green Version]
- Chen, H.; Li, J.; Li, N.; Liu, H.; Tang, J. Increased circulating trimethylamine N-oxide plays a contributory role in the development of endothelial dysfunction and hypertension in the RUPP rat model of preeclampsia. Hypertens. Pregnancy 2019, 38, 96–104. [Google Scholar] [CrossRef]
- Mostafa Mtairag, E.; Chollet-Martin, S.; Oudghiri, M.; Laquay, N.; Jacob, M.P.; Michel, J.B.; Feldman, L.J. Effects of interleukin-10 on monocyte/endothelial cell adhesion and MMP-9/TIMP-1 secretion. Cardiovasc. Res. 2001, 49, 882–890. [Google Scholar] [CrossRef] [Green Version]
- Li, T.; Chen, Y.; Gua, C.; Li, X. Elevated circulating trimethylamine N-oxide levels contribute to endothelial dysfunction in aged rats through vascular inflammation and oxidative stress. Front. Physiol. 2017, 8, 350. [Google Scholar] [CrossRef] [Green Version]
- Herrera, M.D.; Mingorance, C.; Rodríguez-Rodríguez, R.; Alvarez de Sotomayor, M. Endothelial dysfunction and aging: An update. Ageing Res. Rev. 2010, 9, 142–152. [Google Scholar] [CrossRef]
- Donato, A.J.; Morgan, R.G.; Walker, A.E.; Lesniewski, L.A. Cellular and molecular biology of aging endothelial cells. J. Mol. Cell. Cardiol. 2015, 89, 122–135. [Google Scholar] [CrossRef] [Green Version]
- Seals, D.R.; Jablonski, K.L.; Donato, A.J. Aging and vascular endothelial function in humans. Clin. Sci. 2011, 120, 357–375. [Google Scholar] [CrossRef] [Green Version]
- Ma, G.H.; Pan, B.; Chen, Y.; Guo, C.X.; Zhao, M.M.; Zheng, L.M.; Chen, B.X. Trimethylamine N-oxide in atherogenesis: Impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci. Rep. 2017, 37, BSR20160244. [Google Scholar] [CrossRef] [Green Version]
- Hai, X.; Landeras, V.; Dobre, M.A.; DeOreo, P.; Meyer, T.W.; Hostetter, T.H. Mechanism of prominent trimethylamine oxide (TMAO) accumulation in hemodialysis patients. PLoS ONE 2015, 10, e0143731. [Google Scholar] [CrossRef] [PubMed]
- Missailidis, C.; Hällqvist, J.; Qureshi, A.R.; Barany, P.; Heimbürger, O.; Lindholm, B.; Stenvinkel, P.; Bergman, P. Serum trimethylamine-N-Oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS ONE 2016, 11, e0141738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, R.B.; Morse, B.L.; Djurdjev, O.; Tang, M.; Muirhead, N.; Barrett, B.; Holmes, D.T.; Madore, F.; Clase, C.M.; Rigatto, C.; et al. Advanced chronic kidney disease populations have elevated trimethylamine N-oxide levels associated with increased cardiovascular events. Kidney Int. 2016, 89, 1144–1152. [Google Scholar] [CrossRef]
- Robbiano, L.; Mereto, E.; Corbu, C.; Brambilla, G. DNA damage induced by seven N-nitroso compounds in primary cultures of human and rat kidney cells. Mutat. Res. 1996, 368, 41–47. [Google Scholar] [CrossRef] [PubMed]
- Teft, W.A.; Morse, B.L.; Leake, B.F.; Wilson, A.; Mansell, S.E.; Hegele, R.A.; Ho, R.H.; Kim, R.B. Identification and Characterization of Trimethylamine-N-oxide Uptake and Efflux Transporters. Mol. Pharm. 2017, 14, 310–318. [Google Scholar] [CrossRef]
- Zixin, Y.; Lulu, C.; Xiangchang, Z.; Qing, F.; Binjie, Z.; Chunyang, L.; Tai, R.; Dongsheng, O. TMAO as a potential biomarker and therapeutic target for chronic kidney disease: A review. Front. Pharmacol. 2022, 12, 929262. [Google Scholar] [CrossRef]
- Kalagi, N.A.; Thota, R.N.; Stojanovski, E.; Alburikan, K.A.; Garg, M.L. Association between Plasma Trimethylamine N-Oxide Levels and Type 2 Diabetes: A Case Control Study. Nutrients 2022, 14, 2093. [Google Scholar] [CrossRef]
- Zhuang, R.; Ge, X.; Han, L.; Yu, P.; Gong, X.; Meng, Q.; Zhang, Y.; Fan, H.; Zheng, L.; Liu, Z.; et al. Gut microbe-generated metabolite trimethylamine N-oxide and the risk of diabetes: A systematic review and dose-response meta-analysis. Obes. Rev. 2019, 20, 883–894. [Google Scholar] [CrossRef]
- Tang, W.H.W.; Wang, Z.; Li, X.S.; Fan, Y.; Li, D.S.; Wu, Y.; Hazen, S.L. Increased Trimethylamine N-Oxide Portends High Mortality Risk Independent of Glycemic Control in Patients with Type 2 Diabetes Mellitus. Clin. Chem. 2017, 63, 297–306. [Google Scholar] [CrossRef] [Green Version]
- Dambrova, M.; Latkovskis, G.; Kuka, J.; Strele, I.; Konrade, I.; Grinberga, S.; Hartmane, D.; Pugovics, O.; Erglis, A.; Liepinsh, E. Diabetes is Associated with Higher Trimethylamine N-oxide Plasma Levels. Exp. Clin. Endocrinol. Diabetes 2016, 124, 251–256. [Google Scholar] [CrossRef] [Green Version]
- Gao, X.; Liu, X.; Xu, J.; Xue, C.; Xue, Y.; Wang, Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J. Biosci. Bioeng. 2014, 118, 476–481. [Google Scholar] [CrossRef]
- Jia, J.; Dou, P.; Gao, M.; Kong, X.; Li, C.; Liu, Z.; Huang, T. Assessment of Causal Direction Between Gut Microbiota-Dependent Metabolites and Cardiometabolic Health: A Bidirectional Mendelian Randomization Analysis. Diabetes 2019, 68, 1747–1755. [Google Scholar] [CrossRef]
- Rea, D.; Coppola, G.; Palma, G.; Barbieri, A.; Luciano, A.; Del Prete, P.; Rossetti, S.; Berretta, M.; Facchini, G.; Perdonà, S.; et al. Microbiota effects on cancer: From risks to therapies. Oncotarget 2018, 9, 17915–17927. [Google Scholar] [CrossRef] [Green Version]
- Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D. Metabolism in 2013: The gut microbiota manages host metabolism. Nat. Rev. Endocrinol. 2014, 10, 74–76. [Google Scholar] [CrossRef]
- Ianiro, G.; Bruno, G.; Lopetuso, L.; Beghella, F.; Laterza, L.; D’Aversa, F.; Gigante, G.; Cammarota, G.; Gasbarrini, A. Role of Yeasts in Healthy and Impaired Gut Microbiota: The Gut Mycome. Curr. Pharm. Des. 2014, 20, 4565–4569. [Google Scholar] [CrossRef]
- Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [Green Version]
- Columpsi, P.; Sacchi, P.; Zuccaro, V.; Cima, S.; Sarda, C.; Mariani, M.; Gori, A.; Bruno, R. Beyond the gut bacterial microbiota: The gut virome. J. Med. Virol. 2016, 88, 1467–1472. [Google Scholar] [CrossRef]
- Mills, S.; Stanton, C.; Lane, J.A.; Smith, G.J.; Ross, R.P. Precision nutrition and the microbiome, part I: Current state of the science. Nutrients 2019, 11, 923. [Google Scholar] [CrossRef] [Green Version]
- Cenit, M.C.; Sanz, Y.; Codoñer-Franch, P. Influence of gut microbiota on neuropsychiatric disorders. World J. Gastroenterol. 2017, 23, 5486–5498. [Google Scholar] [CrossRef] [Green Version]
- Slyepchenko, A.; Maes, M.; Jacka, F.N.; Köhler, C.A.; Barichello, T.; McIntyre, R.S.; Berk, M.; Grande, I.; Foster, J.A.; Vieta, E.; et al. Gut Microbiota, Bacterial Translocation, and Interactions with Diet: Pathophysiological Links between Major Depressive Disorder and Non-Communicable Medical Comorbidities. Psychother. Psychosom. 2016, 86, 31–46. [Google Scholar] [CrossRef]
- Hold, G.L.; Smith, M.; Grange, C.; Watt, E.R.; El-Omar, E.M.; Mukhopadhya, I. Role of the gut microbiota in inflammatory bowel disease pathogenesis: What have we learnt in the past 10 years? World J. Gastroenterol. 2014, 20, 1192–1210. [Google Scholar] [CrossRef] [PubMed]
- Llorente, C.; Schnabl, B. The Gut Microbiota and Liver Disease. Cell. Mol. Gastroenterol. Hepatol. 2 2015, 1, 275–284. [Google Scholar] [CrossRef] [Green Version]
- Vindigni, S.M.; Zisman, T.L.; Suskind, D.L.; Damman, C.J. The intestinal microbiome, barrier function, and immune system in inflammatory bowel disease: A tripartite pathophysiological circuit with implications for new therapeutic directions. Therap. Adv. Gastroenterol. 2016, 9, 606–625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fukui, H. Increased Intestinal Permeability and Decreased Barrier Function: Does It Really Influence the Risk of Inflammation? Inflamm. Intest. Dis. 2016, 1, 135–145. [Google Scholar] [CrossRef]
- Meng, C.; Bai, C.; Brown, T.D.; Hood, L.E.; Tian, Q. Human Gut Microbiota and Gastrointestinal Cancer. Genom. Proteom. Bioinform. 2018, 16, 33–49. [Google Scholar] [CrossRef]
- Mima, K.; Ogino, S.; Nakagawa, S.; Sawayama, H.; Kinoshita, K.; Krashima, R.; Ishimoto, T.; Imai, K.; Iwatsuki, M.; Hashimoto, D.; et al. The role of intestinal bacteria in the development and progression of gastrointestinal tract neoplasms. Surg. Oncol. 2017, 26, 368–376. [Google Scholar] [CrossRef] [PubMed]
- Bhandari, A.; Woodhouse, M.; Gupta, S. Colorectal cancer is a leading cause of cancer incidence and mortality among adults younger than 50 years in the USA: A SEER-based analysis with comparison to other young-onset cancers. J. Investig. Med. 2017, 65, 311–315. [Google Scholar] [CrossRef] [Green Version]
- Zackular, J.P.; Baxter, N.T.; Iverson, K.D.; Sadler, W.D.; Petrosino, J.F.; Chen, G.Y.; Schloss, P.D. The gut microbiome modulates colon tumorigenesis. MBio 2013, 4, e00692-13. [Google Scholar] [CrossRef] [Green Version]
- Zackular, J.P.; Baxter, N.T.; Chen, G.Y.; Schloss, P.D. Manipulation of the Gut Microbiota Reveals Role in Colon Tumorigenesis. mSphere 2016, 1, e00001-15. [Google Scholar] [CrossRef] [Green Version]
- Bullman, S.; Pedamallu, C.S.; Sicinska, E.; Clancy, T.E.; Zhang, X.; Cai, D.; Neuberg, D.; Huang, K.; Guevara, F.; Nelson, T.; et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 1443–1448. [Google Scholar] [CrossRef]
- Drewes, J.L.; Housseau, F.; Sears, C.L. Sporadic colorectal cancer: Microbial contributors to disease prevention, development and therapy. Br. J. Cancer 2016, 115, 273–280. [Google Scholar] [CrossRef]
- Gagnière, J.; Raisch, J.; Veziant, J.; Barnich, N.; Bonnet, R.; Buc, E.; Bringer, M.A.; Pezet, D.; Bonnet, M. Gut microbiota imbalance and colorectal cancer. World J. Gastroenterol. 2016, 22, 501–518. [Google Scholar] [CrossRef]
- Sun, J.; Kato, I. Gut microbiota, inflammation and colorectal cancer. Genes Dis. 2016, 3, 130–143. [Google Scholar] [CrossRef] [Green Version]
- Youssef, O.; Lahti, L.; Kokkola, A.; Karla, T.; Tikkanen, M.; Ehsan, H.; Carpelan-Holmström, M.; Koskensalo, S.; Böhling, T.; Rautelin, H.; et al. Stool Microbiota Composition Differs in Patients with Stomach, Colon, and Rectal Neoplasms. Dig. Dis. Sci. 2018, 63, 2950–2958. [Google Scholar] [CrossRef] [Green Version]
- Sears, C.L.; Garrett, W.S. Microbes, microbiota, and colon cancer. Cell Host Microbe 2014, 15, 317–328. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Domingue, J.C.; Sears, C.L. Microbiota dysbiosis in select human cancers: Evidence of association and causality. Semin. Immunol. 2017, 32, 25–34. [Google Scholar] [CrossRef]
- Arthur, J.C.; Perez-Chanona, E.; Mühlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123. [Google Scholar] [CrossRef] [Green Version]
- Huycke, M.M.; Abrams, V.; Moore, D.R. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 2002, 23, 529–536. [Google Scholar] [CrossRef]
- Sze, M.A.; Baxter, N.T.; Ruffin, M.T.; Rogers, M.A.M.; Schloss, P.D. Normalization of the microbiota in patients after treatment for colonic lesions. Microbiome 2017, 5, 150. [Google Scholar] [CrossRef]
- Deng, X.; Li, Z.; Li, G.; Li, B.; Jin, X.; Lyu, G. Comparison of microbiota in patients treated by surgery or chemotherapy by 16S rRNA sequencing reveals potential biomarkers for colorectal cancer therapy. Front. Microbiol. 2018, 9, 1607. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Alcoholado, L.; Ordóñez, R.; Otero, A.; Plaza-Andrade, I.; Laborda-Illanes, A.; Medina, J.A.; Ramos-Molina, B.; Gómez-Millán, J.; Queipo-Ortuño, M.I. Gut Microbiota-Mediated Inflammation and Gut Permeability in Patients with Obesity and Colorectal Cancer. Int. J. Mol. Sci. 2020, 21, 6782. [Google Scholar] [CrossRef]
- Liu, X.; Liu, H.; Yuan, C.; Zhang, Y.; Wang, W.; Hu, S.; Liu, L.; Wang, Y. Preoperative serum TMAO level is a new prognostic marker for colorectal cancer. Biomarkers Med. 2017, 11, 443–447. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.L.; Zhu, X.H.; Ran, L.; Lang, H.-D.; Yi, L.; Mi, M.-T. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J. Am. Heart Assoc. 2017, 6, e006347. [Google Scholar] [CrossRef] [PubMed]
- Yue, C.; Yang, X.; Li, J.; Chen, X.; Zhao, X.; Chen, Y.; Wen, Y. Trimethylamine N-oxide prime NLRP3 inflammasome via inhibiting ATG16L1-induced autophagy in colonicepithelial cells. Biochem. Biophys. Res. Commun. 2017, 490, 541–551. [Google Scholar] [CrossRef]
- Huang, C.-F.; Chen, L.; Li, Y.-C.; Wu, L.; Yu, G.-T.; Zhang, W.-F.; Sun, Z.-J. NLRP3 inflammasome activation promotes inflammation-induced carcinogenesis in head and neck squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2017, 36, 116. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Luo, Q.; Feng, X.; Zhang, R.; Li, J.; Chen, F. NLRP3 promotes tumor growth and metastasis in human oral squamous cell carcinoma. BMC Cancer 2018, 18, 500. [Google Scholar] [CrossRef] [Green Version]
- He, Q.; Fu, Y.; Tian, D.; Yan, W. The contrasting roles of inflammasomes in cancer. Am. J. Cancer Res. 2018, 8, 566–583. [Google Scholar]
- Chen, J.; Pitmon, E.; Wnag, K. Micorbiome, inflammation and colorec5toal cancer. Semin. Immunol. 2017, 32, 43–53. [Google Scholar] [CrossRef]
- Wu, D.; Cao, M.; Peng, J.; Li, N.; Yi, S.; Song, L.; Wang, X.; Zhang, M.; Zhao, J. The effect of trimethylamine N-oxide on Helicobacter pylori-induced changes of immunoinflammatory genes expression in gastric epithelial cells. Int. Immunopharmacol. 2017, 43, 172–178. [Google Scholar] [CrossRef]
- Kumari, N.; Dwarakanath, B.S.; Das, A.; Bhatt, A.N. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumor Biol. 2016, 37, 11553–11572. [Google Scholar] [CrossRef]
- Tilg, H.; Adolph, T.E.; Gerner, R.R.; Moschen, A.R. The Intestinal Microbiota in Colorectal Cancer. Cancer Cell 2018, 33, 954–964. [Google Scholar] [CrossRef]
- McEntyre, C.J.; Lever, M.; Chambers, S.T.; George, P.M.; Slow, S.; Elmslie, J.L.; Florkowski, C.M.; Lunt, H.; Krebs, J.D. Variation of betaine, N,N-dimethylglycine, choline, glycerophosphorylcholine, taurine and trimethylamine-N-oxide in the plasma and urine of overweight people with type 2 diabetes over a two-year period. Ann. Clin. Biochem. 2015, 52 Pt 3, 352–360. [Google Scholar] [CrossRef] [Green Version]
- Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [Green Version]
- Federico, A.; Morgillo, F.; Tuccillo, C.; Ciardiello, F.; Loguercio, C. Chronic inflammation and oxidative stress in human carcinogenesis. Int. J. Cancer 2007, 121, 2381–2386. [Google Scholar] [CrossRef]
- Liu, Y.; Dai, M. Trimethylamine N-Oxide Generated by the Gut Microbiota Is Associated with Vascular Inflammation: New Insights into Atherosclerosis. Mediat. Inflamm. 2020, 2020, 4634172. [Google Scholar] [CrossRef] [Green Version]
- Gao, X.; Xu, J.; Jiang, C.; Zhang, Y.; Xue, Y.; Li, Z.; Wang, J.; Xue, C.; Wang, Y. Fish oil ameliorates trimethylamine N-oxide-exacerbated glucose intolerance in high-fat diet-fed mice. Food Funct. 2015, 6, 1117–1125. [Google Scholar] [CrossRef]
- Hernandez-Alonso, P.; Canueto, D.; Giardina, S.; Salas-Salvado, J.; Canellas, N.; Correig, X.; Bullo, M. Pistachio intake modulates urinary gut microbiota-related metabolites in pre-diabetic subjects. Obes. Facts 2017, 10, 198. [Google Scholar] [CrossRef] [Green Version]
- Fiber Intervention on Gut Microbiota in Children with Prader-Willi Syndrome. Case Med. Res. 2019, 4, 991. [CrossRef]
- Velazquez, R.; Ferreira, E.; Knowles, S.; Fux, C.; Rodin, A.; Winslow, W.; Oddo, S. Lifelong choline supplementation ameliorates Alzheimer’s disease pathology and associated cognitive deficits by attenuating microglia activation. Aging Cell 2019, 18, e13037. [Google Scholar] [CrossRef] [Green Version]
- Leermakers, E.T.M.; Moreira, E.M.; Kiefte-de Jong, J.C.; Darweesh, S.K.L.; Visser, T.; Voortman, T.; Bautista, P.K.; Chowdhury, R.; Gorman, D.; Bramer, W.M.; et al. Effects of choline on health across the life course: A systematic review. Nutr. Rev. 2015, 73, 500–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarter, M.; Parikh, V. Choline transporters, cholinergic transmission and cognition. Nat. Rev. Neurosci. 2005, 6, 48–56. [Google Scholar] [CrossRef] [PubMed]
- Zeisel, S.H. The fetal origins of memory: The role of dietary choline in optimal brain development. J. Pediatr. 2006, 149, S131–S136. [Google Scholar] [CrossRef] [Green Version]
- Wessler, I.; Kirkpatrick, C.J. Acetylcholine beyond neurons: The non-neuronal cholinergic system in humans. Br. J. Pharmacol. 2008, 154, 1558–1571. [Google Scholar] [CrossRef] [Green Version]
- Vance, D.E.; Ridgway, N.D. The methylation of phosphatidylethanolamine. Prog. Lipid Res. 1988, 27, 61–79. [Google Scholar] [CrossRef]
- Jiang, X.; West, A.A.; Caudill, M.A. Maternal choline supplementation: A nutritional approach for improving offspring health? Trends Endocrinol. Metab. 2014, 25, 263–273. [Google Scholar] [CrossRef]
- Shronts, E.P. Essential nature of choline with implications for total parenteral nutrition. J. Am. Diet. Assoc. 1997, 97, 639–649. [Google Scholar] [CrossRef]
- European Food Safety Authority (EFSA). Panel on Dietetic Products Nutrition and Allergies (NDA) Overview on Dietary Reference Values for the EU Population as Derived by the EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA); EFSA: Parma, Italy, 2017. [Google Scholar]
- Zaina, S.; Heyn, H.; Carmona, F.J.; Varol, N.; Sayols, S.; Condom, E.; Ramírez-Ruz, J.; Gomez, A.; Gonçalves, I.; Moran, S.; et al. DNA methylation map of human atherosclerosis. Circ. Cardiovasc. Genet. 2014, 7, 692–700. [Google Scholar] [CrossRef] [Green Version]
- Zaina, S.; Lindholm, M.W.; Lund, G. Nutrition and aberrant DNA methylation patterns in atherosclerosis: More than just hyperhomocysteinemia? J. Nutr. 2005, 135, 5–8. [Google Scholar] [CrossRef] [Green Version]
- Da Costa, K.A.; Gaffney, C.E.; Fischer, L.M.; Zeisel, S.H. Choline deficiency in mice and humans is associated with increased plasma homocysteine concentration after a methionine load. Am. J. Clin. Nutr. 2005, 81, 440–444. [Google Scholar] [CrossRef] [Green Version]
- Clarke, R.; Collins, R.; Lewington, S.; Donald, A.; Alfthan, G.; Tuomilehto, J.; Arnesen, E.; Bonaa, K.; Blacher, J.; Boers, G.H.J.; et al. Homocysteine and risk of ischemic heart disease and stroke: A meta-analysis. J. Am. Med. Assoc. 2002, 288, 2015–2022. [Google Scholar]
- Wu, L.L.; Wu, J.T. Hyperhomocysteinemia is a risk factor for cancer and a new potential tumor marker. Clin. Chim. Acta 2002, 322, 21–28. [Google Scholar] [CrossRef] [PubMed]
- Seshadri, S.; Beiser, A.; Selhub, J.; Jacques, P.F.; Rosenberg, I.H.; D’Agostino, R.B.; Wilson, P.W.F.; Wolf, P.A. Plasma Homocysteine as a Risk Factor for Dementia and Alzheimer’s Disease. N. Engl. J. Med. 2002, 346, 476–483. [Google Scholar] [CrossRef] [PubMed]
- Van Meurs, J.B.J.; Dhonukshe-Rutten, R.A.M.; Pluijm, S.M.F.; van der Klift, M.; de Jonge, R.; Lindemans, J.; de Groot, L.C.P.G.M.; Hofman, A.; Witteman, J.C.M.; van Leeuwen, J.P.T.M.; et al. Homocysteine Levels and the Risk of Osteoporotic Fracture. N. Engl. J. Med. 2004, 350, 2033–2041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zeisel, S.H. Metabolic crosstalk between choline/1-carbon metabolism and energy homeostasis. Clin. Chem. Lab. Med. 2013, 51, 467–475. [Google Scholar] [CrossRef]
- Gao, X.; Wang, Y.; Randell, E.; Pedram, P.; Yi, Y.; Gulliver, W.; Sun, G. Higher Dietary Choline and Betaine Intakes are Associated with Better Body Composition in the Adult Population of Newfoundland, Canada. PLoS ONE 2016, 11, e0155403. [Google Scholar] [CrossRef] [Green Version]
- Craig, S.A.S. Betaine in human nutrition. Am. J. Clin. Nutr. 2004, 80, 539–549. [Google Scholar] [CrossRef] [Green Version]
- Tsuge, H.; Nakano, Y.; Onishi, H.; Futamura, Y.; Ohashi, K. A novel purification and some properties of rat liver mitochondrial choline dehydrogenase. Biochim. Biophys. Acta 1980, 614, 274–284. [Google Scholar] [CrossRef]
- Mödinger, Y.; Schön, C.; Wilhelm, M.; Hals, P.A. Plasma kinetics of choline and choline metabolites after a single dose of SuperbaBoostTM krill oil or choline bitartrate in healthy volunteers. Nutrients 2019, 11, 2548. [Google Scholar] [CrossRef] [Green Version]
- Cho, C.E.; Aardema, N.D.J.; Bunnell, M.L.; Larson, D.P.; Aguilar, S.S.; Bergeson, J.R.; Malysheva, O.V.; Caudill, M.A.; Lefevre, M. Effect of choline forms and gut microbiota composition on trimethylamine-n-oxide response in healthy men. Nutrients 2020, 12, 2220. [Google Scholar] [CrossRef]
- Lemos, B.S.; Medina-Vera, I.; Malysheva, O.V.; Caudill, M.A.; Fernandez, M.L. Effects of Egg Consumption and Choline Supplementation on Plasma Choline and Trimethylamine-N-Oxide in a Young Population. J. Am. Coll. Nutr. 2018, 37, 716–723. [Google Scholar] [CrossRef] [PubMed]
- Missimer, A.; Fernandez, M.L.; DiMarco, D.M.; Norris, G.H.; Blesso, C.N.; Murillo, A.G.; Vergara-Jimenez, M.; Lemos, B.S.; Medina-Vera, I.; Malysheva, O.V.; et al. Compared to an Oatmeal Breakfast, Two Eggs/Day Increased Plasma Carotenoids and Choline without Increasing Trimethyl Amine N-Oxide Concentrations. J. Am. Coll. Nutr. 2018, 37, 140–148. [Google Scholar] [CrossRef] [PubMed]
- Bremer, J. Carnitine. Metabolism and functions. Physiol. Rev. 1983, 63, 1420–1480. [Google Scholar] [CrossRef] [PubMed]
- Rebouche, C.J. Kinetics, pharmacokinetics, and regulation of L-Carnitine and acetyl-L-carnitine metabolism. Ann. N. Y. Acad. Sci. 2004, 1033, 30–41. [Google Scholar] [CrossRef]
- Koeth, R.A.; Lam-Galvez, B.R.; Kirsop, J.; Wang, Z.; Levison, B.S.; Gu, X.; Copeland, M.F.; Bartlett, D.; Cody, D.B.; Dai, H.J.; et al. L-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Investig. 2019, 129, 373–387. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Bergeron, N.; Levison, B.S.; Li, X.S.; Chiu, S.; Xun, J.; Koeth, R.A.; Lin, L.; Wu, Y.; Tang, W.H.W.; et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583–594. [Google Scholar] [CrossRef]
- Park, J.Y.; von Karsa, L.; Herrero, R. Prevention strategies for gastric cancer: A global perspective. Clin. Endosc. 2014, 47, 478–489. [Google Scholar] [CrossRef]
- Griffin, L.E.; Djuric, Z.; Angiletta, C.J.; Mitchell, C.M.; Baugh, M.E.; Davy, K.P.; Neilson, A.P. A Mediterranean diet does not alter plasma trimethylamine: N -oxide concentrations in healthy adults at risk for colon cancer. Food Funct. 2019, 10, 2138–2147. [Google Scholar] [CrossRef]
- Martin, F.P.J.; Wang, Y.; Sprenger, N.; Yap, I.K.S.; Lundstedt, T.; Lek, P.; Rezzi, S.; Ramadan, Z.; Van Bladeren, P.; Fay, L.B.; et al. Probiotic modulation of symbiotic gut microbial-host metabolic interactions in a humanized microbiome mouse model. Mol. Syst. Biol. 2008, 4, 157. [Google Scholar] [CrossRef] [Green Version]
- Tripolt, N.J.; Leber, B.; Triebl, A.; Köfeler, H.; Stadlbauer, V.; Sourij, H. Effect of Lactobacillus casei Shirota supplementation on trimethylamine-N-oxide levels in patients with metabolic syndrome: An open-label, randomized study. Atherosclerosis 2015, 242, 141–144. [Google Scholar] [CrossRef]
- Qiu, L.; Tao, X.; Xiong, H.; Yu, J.; Wei, H. Lactobacillus plantarum ZDY04 exhibits a strain-specific property of lowering TMAO via the modulation of gut microbiota in mice. Food Funct. 2018, 9, 4299–4309. [Google Scholar] [CrossRef]
- Qiu, L.; Yang, D.; Tao, X.; Yu, J.; Xiong, H.; Wei, H. Enterobacter aerogenes ZDY01 attenuates choline-induced trimethylamine N-oxide levels by remodeling gut microbiota in mice. J. Microbiol. Biotechnol. 2017, 27, 1491–1499. [Google Scholar] [CrossRef]
- Dridi, B.; Fardeau, M.L.; Ollivier, B.; Raoult, D.; Drancourt, M. Methanomassiliicoccus luminyensis gen. nov., sp. nov., a methanogenic archaeon isolated from human faeces. Int. J. Syst. Evol. Microbiol. 2012, 62, 1902–1907. [Google Scholar] [CrossRef]
- Poesen, R.; Evenepoel, P.; De Loor, H.; Delcour, J.A.; Courtin, C.M.; Kuypers, D.; Augustijns, P.; Verbeke, K.; Meijers, B. The influence of prebiotic arabinoxylan oligosaccharides on microbiota derived uremic retention solutes in patients with chronic kidney disease: A randomized controlled trial. PLoS ONE 2016, 11, e0153893. [Google Scholar] [CrossRef] [Green Version]
- Chaplin, A.; Carpéné, C.; Mercader, J. Resveratrol, metabolic syndrome, and gut microbiota. Nutrients 2018, 10, 1651. [Google Scholar] [CrossRef] [Green Version]
- Jung, C.M.; Heinze, T.M.; Schnackenberg, L.K.; Mullis, L.B.; Elkins, S.A.; Elkins, C.A.; Steele, R.S.; Sutherland, J.B. Interaction of dietary resveratrol with animal-associated bacteria. FEMS Microbiol. Lett. 2009, 297, 266–273. [Google Scholar] [CrossRef]
- Estaki, M.; Pither, J.; Baumeister, P.; Little, J.P.; Gill, S.K.; Ghosh, S.; Ahmadi-Vand, Z.; Marsden, K.R.; Gibson, D.L. Cardiorespiratory fitness as a predictor of intestinal microbial diversity and distinct metagenomic functions. Microbiome 2016, 4, 42. [Google Scholar] [CrossRef] [Green Version]
- Codella, R.; Luzi, L.; Terruzzi, I. Exercise has the guts: How physical activity may positively modulate gut microbiota in chronic and immune-based diseases. Dig. Liver Dis. 2018, 50, 331–341. [Google Scholar] [CrossRef] [Green Version]
- Munukka, E.; Ahtiainen, J.P.; Puigbó, P.; Jalkanen, S.; Pahkala, K.; Keskitalo, A.; Kujala, U.M.; Pietilä, S.; Hollmén, M.; Elo, L.; et al. Six-week endurance exercise alters gut metagenome that is not reflected in systemic metabolism in over-weight women. Front. Microbiol. 2018, 9, 2323. [Google Scholar] [CrossRef] [Green Version]
- Taniguchi, H.; Tanisawa, K.; Sun, X.; Kubo, T.; Hoshino, Y.; Hosokawa, M.; Takeyama, H.; Higuchi, M. Effects of short-term endurance exercise on gut microbiota in elderly men. Physiol. Rep. 2018, 6, e13935. [Google Scholar] [CrossRef]
- Allen, J.M.; Mailing, L.J.; Niemiro, G.M.; Moore, R.; Cook, M.D.; White, B.A.; Holscher, H.D.; Woods, J.A. Exercise Alters Gut Microbiota Composition and Function in Lean and Obese Humans. Med. Sci. Sports Exerc. 2018, 50, 747–757. [Google Scholar] [CrossRef] [PubMed]
- Jäger, R.; Mohr, A.E.; Carpenter, K.C.; Kerksick, C.M.; Purpura, M.; Moussa, A.; Townsend, J.R.; Lamprecht, M.; West, N.P.; Black, K.; et al. International Society of Sports Nutrition Position Stand: Probiotics. J. Int. Soc. Sports Nutr. 2019, 16, 1–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ticinesi, A.; Lauretani, F.; Tana, C.; Nouvenne, A.; Ridolo, E.; Meschi, T. Exercise and immune system as modulators of intestinal microbiome: Implications for the gut-muscle axis hypothesis. Exerc. Immunol. Rev. 2019, 25, 84–95. [Google Scholar] [PubMed]
- Karl, J.P.; Margolis, L.M.; Madslien, E.H.; Murphy, N.E.; Castellani, J.W.; Gundersen, Y.; Hoke, A.V.; Levangie, M.W.; Kumar, R.; Chakraborty, N.; et al. Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress. Am. J. Physiol.-Gastrointest. Liver Physiol. 2017, 312, G559–G571. [Google Scholar] [CrossRef] [Green Version]
- Mills, S.; Lane, J.A.; Smith, G.J.; Grimaldi, K.A.; Ross, R.P.; Stanton, C. Precision nutrition and the microbiome part ii: Potential opportunities and pathways to commercialisation. Nutrients 2019, 11, 1468. [Google Scholar] [CrossRef] [Green Version]
- Shin, N.R.; Lee, J.C.; Lee, H.Y.; Kim, M.S.; Whon, T.W.; Lee, M.S.; Bae, J.W. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014, 63, 727–735. [Google Scholar] [CrossRef] [Green Version]
- Effect of Metformin on Metabolic Improvement and Gut Microbiota. Appl. Environ. Microbiol. 2014, 80, 5935–5943. [CrossRef] [Green Version]
- Wu, M.; Yang, S.; Wang, S.; Cao, Y.; Zhao, R.; Li, X.; Xing, Y.; Liu, L. Effect of Berberine on Atherosclerosis and Gut Microbiota Modulation and Their Correlation in High-Fat Diet-Fed ApoE−/− Mice. Front. Pharmacol. 2020, 11, 223. [Google Scholar] [CrossRef] [Green Version]
- Zhu, L.; Zhang, D.; Zhu, H.; Zhu, J.; Weng, S.; Dong, L.; Liu, T.; Hu, Y.; Shen, X. Berberine treatment increases Akkermansia in the gut and improves high-fat diet-induced atherosclerosis in Apoe−/− mice. Atherosclerosis 2018, 268, 117–126. [Google Scholar] [CrossRef]
- Zhang, W.; Xu, J.H.; Yu, T.; Chen, Q.K. Effects of berberine and metformin on intestinal inflammation and gut microbiome composition in db/db mice. Biomed. Pharmacother. 2019, 118, 109131. [Google Scholar] [CrossRef]
- Wang, H.; Guan, L.; Li, J.; Lai, M.; Wen, X. The effects of berberine on the gut microbiota in APC min/+ mice fed with a high fat diet. Molecules 2018, 23, 2298. [Google Scholar] [CrossRef] [Green Version]
- Li, C.N.; Wang, X.; Lei, L.; Liu, M.Z.; Li, R.C.; Sun, S.J.; Liu, S.N.; Huan, Y.; Zhou, T.; Liu, Q.; et al. Berberine combined with stachyose induces better glycometabolism than berberine alone through modulating gut microbiota and fecal metabolomics in diabetic mice. Phyther. Res. 2020, 34, 1166–1174. [Google Scholar] [CrossRef] [Green Version]
- Zhang, T.; Li, Q.; Cheng, L.; Buch, H.; Zhang, F. Akkermansia muciniphila is a promising probiotic. Microb. Biotechnol. 2019, 12, 1109–1125. [Google Scholar] [CrossRef]
- Crovesy, L.; Masterson, D.; Rosado, E.L. Profile of the gut microbiota of adults with obesity: A systematic review. Eur. J. Clin. Nutr. 2020, 74, 1251–1262. [Google Scholar] [CrossRef]
- Wang, Z.; Roberts, A.B.; Buffa, J.A.; Levison, B.S.; Zhu, W.; Org, E.; Gu, X.; Huang, Y.; Zamanian-Daryoush, M.; Culley, M.K.; et al. Non-lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis. Cell 2015, 163, 1585–1595. [Google Scholar] [CrossRef] [Green Version]
- Roberts, A.B.; Gu, X.; Buffa, J.A.; Hurd, A.G.; Wang, Z.; Zhu, W.; Gupta, N.; Skye, S.M.; Cody, D.B.; Levison, B.S.; et al. Development of a gut microbe–targeted nonlethal therapeutic to inhibit thrombosis potential. Nat. Med. 2018, 24, 1407–1417. [Google Scholar] [CrossRef]
- Dambrova, M.; Skapare-makarova, E.; Konrade, I.; Pugovics, O.; Grinberga, S.; Tirzite, D.; Petrovska, R.; Kalvins, I.; Liepins, E. Meldonium decreases the diet-increased plasma levels of trimethylamine n-oxide, a metabolite associated with atherosclerosis. J. Clin. Pharmacol. 2013, 53, 1095–1098. [Google Scholar] [CrossRef]
- Kuka, J.; Liepinsh, E.; Makrecka-Kuka, M.; Liepins, J.; Cirule, H.; Gustina, D.; Loza, E.; Zharkova-Malkova, O.; Grinberga, S.; Pugovics, O.; et al. Suppression of intestinal microbiota-dependent production of pro-atherogenic trimethylamine N-oxide by shifting L-carnitine microbial degradation. Life Sci. 2014, 117, 84–92. [Google Scholar] [CrossRef]
- 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, 380–385. [Google Scholar] [CrossRef]
- Velebova, K.; Hoang, T.; Veleba, J.; Belinova, L.; Kopecky, J.; Kuda, O.; Segetova, M.; Kopecky, J.; Melenovsky, V.; Pelikanova, T. The effect of metformin on serum levels of Trimethylamine-N-oxide in patients with type 2 diabetes/prediabetes and chronic heart failure. Diabetologia 2016, 59, S533. [Google Scholar]
- Kuka, J.; Videja, M.; Makrecka-Kuka, M.; Liepins, J.; Grinberga, S.; Sevostjanovs, E.; Vilks, K.; Liepinsh, E.; Dambrova, M. Metformin decreases bacterial trimethylamine production and trimethylamine N-oxide levels in db/db mice. Sci. Rep. 2020, 10, 14555. [Google Scholar] [CrossRef] [PubMed]
- Kalagi, N.A.; Abbott, K.A.; Alburikan, K.A.; Alkofide, H.A.; Stojanovski, E.; Garg, M.L. Modulation of Circulating Trimethylamine N-Oxide Concentrations by Dietary Supplements and Pharmacological Agents: A Systematic Review. Adv. Nutr. 2019, 10, 876–887. [Google Scholar] [CrossRef] [PubMed]
- Jalandra, R.; Dalal, N.; Yadav, A.K.; Verma, D.; Sharma, M.; Singh, R.; Khosla, A.; Kumar, A.; Solanki, P.R. Emerging role of trimethylamine-N-oxide (TMAO) in colorectal cancer. Appl. Microbiol. Biotechnol. 2021, 105, 7651–7660. [Google Scholar] [CrossRef] [PubMed]
Betaine | Free Choline | Phosphocholine | |
---|---|---|---|
Egg yolk | 0.0 | 2.38 | 5.84 |
Milk | |||
Whole 3.25% Fat | 0.6 | 3.7 | 1.8 |
2% Fat | 0.9 | 2.8 | 1.6 |
Butter | 0.3 | 0.5 | 0.7 |
Cheese | |||
Cheddar | 0.7 | 1.6 | 0.6 |
Cottage 2% | 0.6 | 2.9 | 1.3 |
Mozzarella | 0.7 | 2.3 | 0.9 |
Swiss | 0.6 | 4.5 | 0.0 |
Chicken | |||
Meat and skin | 7.8 | 6.0 | 3.6 |
Liver | 16.9 | 49.2 | 4.1 |
Pork | |||
Sausage | 3.4 | 8.0 | 0.5 |
Bacon | 0.9 | 4.4 | 1.4 |
Loin, lean only | 2.4 | 1.6 | 2.2 |
Beef | |||
Ground 80% lean | 8.2 | 2.6 | 0.4 |
Liver | 4.4 | 56.2 | 11.8 |
Fish and seafoods (cooked) | |||
Cod (Altantic) | 9.7 | 17.7 | 1.6 |
Shrimp | 33.0 | 1.5 | 0.8 |
Salmon (Sockeye) | 2.1 | 8.6 | 1.1 |
Tilapia | 25.3 | 21.4 | 2.5 |
Tuna (canned in water) | 2.7 | 2.1 | 0.0 |
Vegetables | |||
Beets | 128.7 | 4.1 | 0.9 |
Broccoli | 0.1 | 18.1 | 0.4 |
Cabbage | 0.4 | 6.1 | 2.3 |
Carrots | 0.4 | 6.8 | 1.1 |
Lettuce iceberg | 0.1 | 4.8 | 1.5 |
Mushrooms | 10.7 | 5.9 | 1.3 |
Potato, white, flesh and skin | 0.2 | 7.9 | 0.3 |
Spinach boiled, drained | 726 | 1.7 | 1.1 |
Tomato paste | 0.4 | 26.2 | 4.3 |
Tomato | 0.1 | 4.4 | 1.8 |
Nuts (roasted and dried) | |||
Almonds | 0.5 | 9.4 | 1.9 |
Brazilnuts | 0.4 | 16.1 | 0.3 |
Cashews | 11.2 | 19.6 | 0.9 |
Hazelnuts | 0.4 | 15.2 | 0.9 |
Macadamia | 0.3 | 11.3 | 1.0 |
Pecans | 0.7 | 9.7 | 1.3 |
Pine | 0.4 | 8.4 | 2.1 |
Pistachio | 0.8 | 10.7 | 8.5 |
walnuts | 0.5 | 8.3 | 0.5 |
Legumes | |||
Beans, kidney, canned | 0.1 | 19.7 | 0.5 |
Peanut butter, smooth | 0.4 | 25.8 | 0.7 |
Soy milk | 0.8 | 13.1 | 3.4 |
Soy sauce (shoyu) | 39.6 | 31.0 | 0.0 |
Cereal grains and others | |||
Oat bran | 35.7 | 4.4 | 0.7 |
Rice, brown | 0.5 | 4.7 | 0.0 |
Pasta, dry | 460 | 9.7 | 0.0 |
Wheat flour, white | 124.4 | 5.7 | 0.1 |
Bread, wheat | 85.2 | 11.5 | 0.3 |
Kellogg’s all-bran | 360.0 | 25.5 | 1.7 |
Beef | |
Steak | 65.0 |
Ground | 87.5 |
Tenderloin | 78.6 |
T-Bone | 84.2 |
Loin | 64.6 |
Chicken | |
Liver | 94.0 |
Meat | 10.4 |
Wing Meat | 10.0 |
Turkey meat | 21.2 |
Lamb Chop | 40.5 |
Pork | |
Shoulder | 21.1 |
Ham | 53.5 |
White Ham | 33.5 |
Sausage | 7.1 |
Veal | |
Shoulder | 78.2 |
Sirloin | 132.8 |
Milk | |
2% fat | 2.9 |
4% fat | 2.3 |
Butter | 1.3 |
Cheese | |
Camembert | 14.4 |
Gruyere | 6.5 |
Feta | 1.8 |
Goat cheese | 15.3 |
Mozzarella | 0.3 |
Parmesan | 0.7 |
Yogurt | |
Regular | 12.2 |
0% fat | 12.5 |
Egg | |
White | 0.3 |
Yolk | 0.8 |
Fish and seafood | |
Anchovy | 1.8 |
Shrimp | 0.7 |
Cod (Antlantic) | 1.8 |
Hake (boiled) | 2.9 |
Mussels (cooked) | 2.6 |
Salmon (cooked) | 5.8 |
Smoked salmon | 1.0 |
Tuna | 1.5 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Tacconi, E.; Palma, G.; De Biase, D.; Luciano, A.; Barbieri, M.; de Nigris, F.; Bruzzese, F. Microbiota Effect on Trimethylamine N-Oxide Production: From Cancer to Fitness—A Practical Preventing Recommendation and Therapies. Nutrients 2023, 15, 563. https://doi.org/10.3390/nu15030563
Tacconi E, Palma G, De Biase D, Luciano A, Barbieri M, de Nigris F, Bruzzese F. Microbiota Effect on Trimethylamine N-Oxide Production: From Cancer to Fitness—A Practical Preventing Recommendation and Therapies. Nutrients. 2023; 15(3):563. https://doi.org/10.3390/nu15030563
Chicago/Turabian StyleTacconi, Edoardo, Giuseppe Palma, Davide De Biase, Antonio Luciano, Massimiliano Barbieri, Filomena de Nigris, and Francesca Bruzzese. 2023. "Microbiota Effect on Trimethylamine N-Oxide Production: From Cancer to Fitness—A Practical Preventing Recommendation and Therapies" Nutrients 15, no. 3: 563. https://doi.org/10.3390/nu15030563
APA StyleTacconi, E., Palma, G., De Biase, D., Luciano, A., Barbieri, M., de Nigris, F., & Bruzzese, F. (2023). Microbiota Effect on Trimethylamine N-Oxide Production: From Cancer to Fitness—A Practical Preventing Recommendation and Therapies. Nutrients, 15(3), 563. https://doi.org/10.3390/nu15030563