The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases
Abstract
:1. Introduction
2. Relationship between Microbiota and Cardiovascular Diseases
2.1. Diet, Gut Microbiota and Cardiovascular Diseases
2.2. Microbiota and Cardiovascular Diseases
2.3. Microbiota and Atherosclerosis
2.4. Other Microbiota Aspects Related to Cardiovascular Diseases
2.4.1. Microbiota, Choline and Homocysteine Cycle
2.4.2. Vitamin B-Complex and Microbiota
2.4.3. Low-Grade Inflammation
3. Microbiota-Targeted Therapeutics
3.1. Physical Activity, Microbiota and Cardiovascular Diseases
3.2. Probiotic Administration, Microbiota, Bile Acids and Cardiovascular Diseases
3.3. Fecal Microbiota Transplantation
3.4. Personalized Nutrition
4. Prevention of Cardiovascular Diseases
5. Further Directions and Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- World Health Organization. Cardiovascular Disease. Available online: https://www.who.int/cardiovascular_diseases/about_cvd/en/ (accessed on 13 November 2019).
- Benjamin, E.J.; Virani, S.S.; Callaway, C.W.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Chiuve, S.E.; Cushman, M.; Delling, F.N.; Deo, R.; et al. Heart disease and stroke statistics-2018 update: A report from the American heart association. Circulation 2018, 137, e67–e492. [Google Scholar] [CrossRef]
- The, L. GBD 2017: A fragile world. Lancet (Lond. Engl.) 2018, 392, 1683. [Google Scholar]
- Ross, R. Atherosclerosis—An inflammatory disease. N. Engl. J. Med. 1999, 340, 115–126. [Google Scholar] [CrossRef] [PubMed]
- Maguire, E.M.; Pearce, S.W.A.; Xiao, Q. Foam cell formation: A new target for fighting atherosclerosis and cardiovascular disease. Vasc. Pharmacol. 2019, 112, 54–71. [Google Scholar] [CrossRef] [PubMed]
- Nabel, E.G. Cardiovascular disease. N. Engl. J. Med. 2003, 349, 60–72. [Google Scholar] [CrossRef]
- Ardissino, D.; Berzuini, C.; Merlini, P.A.; Mannuccio Mannucci, P.; Surti, A.; Burtt, N.; Voight, B.; Tubaro, M.; Peyvandi, F.; Spreafico, M.; et al. Influence of 9p21.3 genetic variants on clinical and angiographic outcomes in early-onset myocardial infarction. J. Am. Coll. Cardiol. 2011, 58, 426–434. [Google Scholar] [CrossRef]
- Ripatti, S.; Tikkanen, E.; Orho-Melander, M.; Havulinna, A.S.; Silander, K.; Sharma, A.; Guiducci, C.; Perola, M.; Jula, A.; Sinisalo, J.; et al. A multilocus genetic risk score for coronary heart disease: Case-control and prospective cohort analyses. Lancet 2010, 376, 1393–1400. [Google Scholar] [CrossRef] [Green Version]
- Labarthe, D.R. Epidemiology and Prevention of Cardiovascular Diseases: A Global Challenge; Jones & Bartlett Publishers: Sudbury, MA, USA, 2010. [Google Scholar]
- 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]
- Karlsson, F.H.; Fåk, F.; Nookaew, I.; Tremaroli, V.; Fagerberg, B.; Petranovic, D.; Bäckhed, F.; Nielsen, J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat. Commun. 2012, 3, 1245. [Google Scholar] [CrossRef] [Green Version]
- Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of action of probiotics. Adv. Nutr. 2019, 10, S49–S66. [Google Scholar] [CrossRef] [Green Version]
- Mouzaki, M.; Comelli, E.M.; Arendt, B.M.; Bonengel, J.; Fung, S.K.; Fischer, S.E.; McGilvray, I.D.; Allard, J.P. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 2013, 58, 120–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, W.H.W.; Backhed, F.; Landmesser, U.; Hazen, S.L. Intestinal microbiota in cardiovascular health and disease: JACC state-of-the-art review. J. Am. Coll. Cardiol. 2019, 73, 2089–2105. [Google Scholar] [CrossRef] [PubMed]
- Tang, W.W.; Kitai, T.; Hazen, S.L. Gut microbiota in cardiovascular health and disease. Circ. Res. 2017, 120, 1183–1196. [Google Scholar] [CrossRef] [Green Version]
- Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
- Reinhardt, C. The gut microbiota as an influencing factor of arterial thrombosis. Hamostaseologie 2019, 39, 173–179. [Google Scholar] [CrossRef]
- Jackel, S.; Kiouptsi, K.; Lillich, M.; Hendrikx, T.; Khandagale, A.; Kollar, B.; Hormann, N.; Reiss, C.; Subramaniam, S.; Wilms, E.; et al. Gut microbiota regulate hepatic von Willebrand factor synthesis and arterial thrombus formation via Toll-like receptor-2. Blood 2017, 130, 542–553. [Google Scholar] [CrossRef] [Green Version]
- Álvarez-Mercado, A.I.; Navarro-Oliveros, M.; Robles-Sánchez, C.; Plaza-Díaz, J.; Sáez-Lara, M.J.; Muñoz-Quezada, S.; Fontana, L.; Abadía-Molina, F. Microbial population changes and their relationship with human health and disease. Microorganisms 2019, 7, 68. [Google Scholar] [CrossRef] [Green Version]
- Group, N.H.W.; Peterson, J.; Garges, S.; Giovanni, M.; McInnes, P.; Wang, L.; Schloss, J.A.; Bonazzi, V.; McEwen, J.E.; Wetterstrand, K.A.; et al. The NIH human microbiome project. Genome Res. 2009, 19, 2317–2323. [Google Scholar] [CrossRef] [Green Version]
- Roy, S.; Trinchieri, G. Microbiota: A key orchestrator of cancer therapy. Nat. Rev. Cancer 2017, 17, 271–285. [Google Scholar] [CrossRef]
- Micheel, C.M.; Nass, S.J.; Omenn, G.S. Omics-based clinical discovery: Science, technology, and applications. In Evolution of Translational Omics: Lessons Learned and the Path Forward; National Academies Press: Washington, DC, USA, 2012. [Google Scholar]
- Maruvada, P.; Leone, V.; Kaplan, L.M.; Chang, E.B. The human microbiome and obesity: Moving beyond associations. Cell Host Microbe 2017, 22, 589–599. [Google Scholar] [CrossRef] [PubMed]
- Miyamoto, J.; Igarashi, M.; Watanabe, K.; Karaki, S.I.; Mukouyama, H.; Kishino, S.; Li, X.; Ichimura, A.; Irie, J.; Sugimoto, Y.; et al. Gut microbiota confers host resistance to obesity by metabolizing dietary polyunsaturated fatty acids. Nat. Commun. 2019, 10, 4007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rajagopala, S.V.; Vashee, S.; Oldfield, L.M.; Suzuki, Y.; Venter, J.C.; Telenti, A.; Nelson, K.E. The human microbiome and cancer. Cancer Prev. Res. 2017, 10, 226–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, W.; Yang, Y.; Wang, H.; Wang, H.; Yu, X.; Lu, Y.; Shen, S.; Teng, L. Gut microbiota shifts in patients with gastric cancer in perioperative period. Medicine (Baltimore) 2019, 98, e16626. [Google Scholar] [CrossRef]
- Kostic, A.D.; Gevers, D.; Pedamallu, C.S.; Michaud, M.; Duke, F.; Earl, A.M.; Ojesina, A.I.; Jung, J.; Bass, A.J.; Tabernero, J.; et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012, 22, 292–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McCoy, A.N.; Araujo-Perez, F.; Azcarate-Peril, A.; Yeh, J.J.; Sandler, R.S.; Keku, T.O. Fusobacterium is associated with colorectal adenomas. PLoS ONE 2013, 8, e53653. [Google Scholar] [CrossRef] [PubMed]
- McIlroy, J.; Ianiro, G.; Mukhopadhya, I.; Hansen, R.; Hold, G.L. Review article: The gut microbiome in inflammatory bowel disease-avenues for microbial management. Aliment. Pharmacol. Ther. 2018, 47, 26–42. [Google Scholar] [CrossRef]
- Sitkin, S.; Demyanova, E.; Vakhitov, T.; Pokrotnieks, J. Altered sphingolipid metabolism and its interaction with the intestinal microbiome is another key to the pathogenesis of inflammatory bowel disease. Inflamm. Bowel Dis. 2019, 25, e157–e158. [Google Scholar] [CrossRef]
- Yeoh, N.; Burton, J.P.; Suppiah, P.; Reid, G.; Stebbings, S. The role of the microbiome in rheumatic diseases. Curr. Rheumatol. Rep. 2013, 15, 314. [Google Scholar] [CrossRef]
- van Dijkhuizen, E.H.P.; Del Chierico, F.; Malattia, C.; Russo, A.; Pires Marafon, D.; Ter Haar, N.M.; Magni-Manzoni, S.; Vastert, S.J.; Dallapiccola, B.; Prakken, B.; et al. Microbiome analytics of the gut microbiota in patients with juvenile idiopathic arthritis: A longitudinal observational cohort study. Arthritis Rheumatol. 2019, 71, 1000–1010. [Google Scholar] [CrossRef]
- Ascher, S.; Reinhardt, C. The gut microbiota: An emerging risk factor for cardiovascular and cerebrovascular disease. Eur. J. Immunol. 2018, 48, 564–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miele, L.; Giorgio, V.; Alberelli, M.A.; De Candia, E.; Gasbarrini, A.; Grieco, A. Impact of gut microbiota on obesity, diabetes, and cardiovascular disease risk. Curr. Cardiol. Rep. 2015, 17, 120. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Zhu, W.; Gregory, J.C.; Org, E.; Buffa, J.A.; Gupta, N.; Wang, Z.; Li, L.; Fu, X.; Wu, Y.; Mehrabian, M.; et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016, 165, 111–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benakis, C.; Brea, D.; Caballero, S.; Faraco, G.; Moore, J.; Murphy, M.; Sita, G.; Racchumi, G.; Ling, L.; Pamer, E.G.; et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal gammadelta T cells. Nat. Med. 2016, 22, 516–523. [Google Scholar] [CrossRef]
- Lin, L.; Zhang, J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol. 2017, 18, 2. [Google Scholar] [CrossRef] [Green Version]
- Kohling, H.L.; Plummer, S.F.; Marchesi, J.R.; Davidge, K.S.; Ludgate, M. The microbiota and autoimmunity: Their role in thyroid autoimmune diseases. Clin. Immunol. 2017, 183, 63–74. [Google Scholar] [CrossRef]
- Chambers, E.S.; Preston, T.; Frost, G.; Morrison, D.J. Role of gut microbiota-generated short-chain fatty acids in metabolic and cardiovascular health. Curr. Nutr. Rep. 2018, 7, 198–206. [Google Scholar] [CrossRef] [Green Version]
- Pluznick, J.L.; Protzko, R.J.; Gevorgyan, H.; Peterlin, Z.; Sipos, A.; Han, J.; Brunet, I.; Wan, L.X.; Rey, F.; Wang, T.; et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc. Natl. Acad. Sci. USA 2013, 110, 4410–4415. [Google Scholar] [CrossRef] [Green Version]
- Natarajan, N.; Hori, D.; Flavahan, S.; Steppan, J.; Flavahan, N.A.; Berkowitz, D.E.; Pluznick, J.L. Microbial short chain fatty acid metabolites lower blood pressure via endothelial G protein-coupled receptor 41. Physiol. Genom. 2016, 48, 826–834. [Google Scholar] [CrossRef]
- Wang, B.; Jiang, X.; Cao, M.; Ge, J.; Bao, Q.; Tang, L.; Chen, Y.; Li, L. Altered fecal microbiota correlates with liver biochemistry in nonobese patients with non-alcoholic fatty liver disease. Sci. Rep. 2016, 6, 32002. [Google Scholar] [CrossRef]
- Perry, R.J.; Peng, L.; Barry, N.A.; Cline, G.W.; Zhang, D.; Cardone, R.L.; Petersen, K.F.; Kibbey, R.G.; Goodman, A.L.; Shulman, G.I. Acetate mediates a microbiome-brain-beta-cell axis to promote metabolic syndrome. Nature 2016, 534, 213–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Backhed, F. From dietary fiber to host physiology: Short-Chain fatty acids as key bacterial metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rios-Covian, D.; Gueimonde, M.; Duncan, S.H.; Flint, H.J.; de los Reyes-Gavilan, C.G. Enhanced butyrate formation by cross-feeding between Faecalibacterium prausnitzii and Bifidobacterium adolescentis. FEMS Microbiol. Lett. 2015, 362. [Google Scholar] [CrossRef] [Green Version]
- Hoek, M.; Merks, R.M.H. Emergence of microbial diversity due to cross-feeding interactions in a spatial model of gut microbial metabolism. BMC Syst. Biol. 2017, 11, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [Green Version]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [Green Version]
- Granado-Serrano, A.B.; Martin-Gari, M.; Sanchez, V.; Riart Solans, M.; Berdun, R.; Ludwig, I.A.; Rubio, L.; Vilaprinyo, E.; Portero-Otin, M.; Serrano, J.C.E. Faecal bacterial and short-chain fatty acids signature in hypercholesterolemia. Sci. Rep. 2019, 9, 1772. [Google Scholar] [CrossRef]
- Yamashita, M.; Okubo, H.; Kobuke, K.; Ohno, H.; Oki, K.; Yoneda, M.; Tanaka, J.; Hattori, N. Alteration of gut microbiota by a Westernized lifestyle and its correlation with insulin resistance in non-diabetic Japanese men. J. Diabetes Investig. 2019, 10, 1463–1470. [Google Scholar] [CrossRef] [Green Version]
- Zhong, H.; Penders, J.; Shi, Z.; Ren, H.; Cai, K.; Fang, C.; Ding, Q.; Thijs, C.; Blaak, E.E.; Stehouwer, C.D.A.; et al. Impact of early events and lifestyle on the gut microbiota and metabolic phenotypes in young school-age children. Microbiome 2019, 7, 2. [Google Scholar] [CrossRef] [Green Version]
- Ruggles, K.V.; Wang, J.; Volkova, A.; Contreras, M.; Noya-Alarcon, O.; Lander, O.; Caballero, H.; Dominguez-Bello, M.G. Changes in the gut microbiota of urban subjects during an immersion in the traditional diet and lifestyle of a rainforest village. mSphere 2018, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dubois, G.; Girard, C.; Lapointe, F.J.; Shapiro, B.J. The Inuit gut microbiome is dynamic over time and shaped by traditional foods. Microbiome 2017, 5, 151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palmer, C.; Bik, E.M.; DiGiulio, D.B.; Relman, D.A.; Brown, P.O. Development of the human infant intestinal microbiota. PLoS Biol. 2007, 5, e177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Human Microbiome Project, C. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Faith, J.J.; Guruge, J.L.; Charbonneau, M.; Subramanian, S.; Seedorf, H.; Goodman, A.L.; Clemente, J.C.; Knight, R.; Heath, A.C.; Leibel, R.L.; et al. The long-term stability of the human gut microbiota. Science 2013, 341, 1237439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, Y.; Wan, J.; Choe, U.; Pham, Q.; Schoene, N.W.; He, Q.; Li, B.; Yu, L.; Wang, T.T.Y. Interactions between food and gut microbiota: Impact on human health. Annu. Rev. Food Sci. Technol. 2019, 10, 389–408. [Google Scholar] [CrossRef] [PubMed]
- Proctor, C.; Thiennimitr, P.; Chattipakorn, N.; Chattipakorn, S.C. Diet, gut microbiota and cognition. Metab. Brain Dis. 2017, 32, 1–17. [Google Scholar] [CrossRef]
- Mailing, L.J.; Allen, J.M.; Buford, T.W.; Fields, C.J.; Woods, J.A. Exercise and the gut microbiome: A review of the evidence, potential mechanisms, and implications for human health. Exerc. Sport Sci. Rev. 2019, 47, 75–85. [Google Scholar] [CrossRef]
- Neufeld, K.A.; Kang, N.; Bienenstock, J.; Foster, J.A. Effects of intestinal microbiota on anxiety-like behavior. Commun. Integr. Biol. 2011, 4, 492–494. [Google Scholar] [CrossRef]
- Lach, G.; Schellekens, H.; Dinan, T.G.; Cryan, J.F. Anxiety, depression, and the microbiome: A role for gut peptides. Neurother. J. Am. Soc. Exp. Neurother. 2018, 15, 36–59. [Google Scholar] [CrossRef] [Green Version]
- Petra, A.I.; Panagiotidou, S.; Hatziagelaki, E.; Stewart, J.M.; Conti, P.; Theoharides, T.C. Gut-microbiota-brain axis and its effect on neuropsychiatric disorders with suspected immune dysregulation. Clin. Ther. 2015, 37, 984–995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, N.; Yun, M.; Oh, Y.J.; Choi, H.J. Mind-altering with the gut: Modulation of the gut-brain axis with probiotics. J. Microbiol. 2018, 56, 172–182. [Google Scholar] [CrossRef] [PubMed]
- Dumas, M.E.; Kinross, J.; Nicholson, J.K. Metabolic phenotyping and systems biology approaches to understanding metabolic syndrome and fatty liver disease. Gastroenterology 2014, 146, 46–62. [Google Scholar] [CrossRef] [PubMed]
- Dumas, M.-E.; Barton, R.H.; Toye, A.; Cloarec, O.; Blancher, C.; Rothwell, A.; Fearnside, J.; Tatoud, R.; Blanc, V.; Lindon, J.C. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc. Natl. Acad. Sci. USA 2006, 103, 12511–12516. [Google Scholar] [CrossRef] [Green Version]
- Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013, 341, 1241214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turnbaugh, P.J.; Gordon, J.I. The core gut microbiome, energy balance and obesity. J. Physiol. 2009, 587, 4153–4158. [Google Scholar] [CrossRef]
- Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480. [Google Scholar] [CrossRef] [Green Version]
- Barrett, E.; Kwan, H. Bacterial reduction of trimethylamine oxide. Annu. Rev. Microbiol. 1985, 39, 131–149. [Google Scholar] [CrossRef]
- Smith, J.L.; Wishnok, J.S.; Deen, W.M. Metabolism and excretion of methylamines in rats. Toxicol. Appl. Pharmacol. 1994, 125, 296–308. [Google Scholar] [CrossRef]
- Zhang, A.; Mitchell, S.; Smith, R. Dietary precursors of trimethylamine in man: A pilot study. Food Chem. Toxicol. 1999, 37, 515–520. [Google Scholar] [CrossRef]
- Bain, M.A.; Fornasini, G.; Evans, A.M. Trimethylamine: Metabolic, pharmacokinetic and safety aspects. Curr. Drug Metab. 2005, 6, 227–240. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Vance, D.E. Thematic review series: Glycerolipids. Phosphatidylcholine and choline homeostasis. J. Lipid Res. 2008, 49, 1187–1194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Geiger, O.; López-Lara, I.M.; Sohlenkamp, C. Phosphatidylcholine biosynthesis and function in bacteria. Biochim. Et Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2013, 1831, 503–513. [Google Scholar] [CrossRef] [PubMed]
- Tang, W.H.; Hazen, S.L. The contributory role of gut microbiota in cardiovascular disease. J. Clin. Investig. 2014, 124, 4204–4211. [Google Scholar] [CrossRef] [PubMed]
- 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. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. Invest. 2019, 129. [Google Scholar] [CrossRef]
- Tang, W.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]
- Lever, M.; George, P.M.; Slow, S.; Bellamy, D.; Young, J.M.; Ho, M.; McEntyre, C.J.; Elmslie, J.L.; Atkinson, W.; Molyneux, S.L. Betaine and trimethylamine-N-oxide as predictors of cardiovascular outcomes show different patterns in diabetes mellitus: An observational study. PLoS ONE 2014, 9, e114969. [Google Scholar] [CrossRef] [Green Version]
- Mente, A.; Chalcraft, K.; Ak, H.; Davis, A.D.; Lonn, E.; Miller, R.; Potter, M.A.; Yusuf, S.; Anand, S.S.; McQueen, M.J. The relationship between trimethylamine-N-oxide and prevalent cardiovascular disease in a multiethnic population living in Canada. Can. J. Cardiol. 2015, 31, 1189–1194. [Google Scholar] [CrossRef]
- Trøseid, M.; Ueland, T.; Hov, J.R.; Svardal, A.; Gregersen, I.; Dahl, C.P.; Aakhus, S.; Gude, E.; Bjørndal, B.; Halvorsen, B.; et al. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J. Intern. Med. 2015, 277, 717–726. [Google Scholar] [CrossRef]
- Kaluza, J.; Åkesson, A.; Wolk, A. Processed and unprocessed red meat consumption and risk of heart failure: Prospective study of men. Circ. Heart Fail. 2014, 7, 552–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pan, A.; Sun, Q.; Bernstein, A.M.; Schulze, M.B.; Manson, J.E.; Stampfer, M.J.; Willett, W.C.; Hu, F.B. Red meat consumption and mortality: Results from 2 prospective cohort studies. Arch. Intern. Med. 2012, 172, 555–563. [Google Scholar] [PubMed] [Green Version]
- Spence, J.D.; Jenkins, D.J.; Davignon, J. Egg yolk consumption and carotid plaque. Atherosclerosis 2012, 224, 469–473. [Google Scholar] [CrossRef] [PubMed]
- Kaluza, J.; Wolk, A.; Larsson, S.C. Red meat consumption and risk of stroke: A meta-analysis of prospective studies. Stroke 2012, 43, 2556–2560. [Google Scholar] [CrossRef] [Green Version]
- Larsson, S.C.; Orsini, N. Red meat and processed meat consumption and all-cause mortality: A meta-analysis. Am. J. Epidemiol. 2013, 179, 282–289. [Google Scholar] [CrossRef] [Green Version]
- Djoussé, L.; Gaziano, J.M. Egg consumption in relation to cardiovascular disease and mortality: The Physicians’ Health Study. Am. J. Clin. Nutr. 2008, 87, 964–969. [Google Scholar] [CrossRef]
- Djoussé, L.; Gaziano, J.M. Egg consumption and risk of heart failure in the Physicians’ Health Study. Circulation 2008, 117, 512. [Google Scholar] [CrossRef] [Green Version]
- Qureshi, A.I.; Suri, M.F.K.; Ahmed, S.; Nasar, A.; Divani, A.A.; Kirmani, J.F. Regular egg consumption does not increase the risk of stroke and cardiovascular diseases. Med. Sci. Monit. 2007, 13, 8. [Google Scholar]
- Scrafford, C.G.; Tran, N.L.; Barraj, L.M.; Mink, P.J. Egg consumption and CHD and stroke mortality: A prospective study of US adults. Public Health Nutr. 2011, 14, 261–270. [Google Scholar] [CrossRef] [Green Version]
- Tran, N.L.; Barraj, L.M.; Heilman, J.M.; Scrafford, C.G. Egg consumption and cardiovascular disease among diabetic individuals: A systematic review of the literature. Diabetes Metab. Syndr. Obes. Targ. Ther. 2014, 7, 121. [Google Scholar] [CrossRef] [Green Version]
- Miller, C.A.; Corbin, K.D.; da Costa, K.-A.; Zhang, S.; Zhao, X.; Galanko, J.A.; Blevins, T.; Bennett, B.J.; O’Connor, A.; Zeisel, S.H. Effect of egg ingestion on trimethylamine-N-oxide production in humans: A randomized, controlled, dose-response study. Am. J. Clin. Nutr. 2014, 100, 778–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Tang, W.W.; Buffa, J.A.; Fu, X.; Britt, E.B.; Koeth, R.A.; Levison, B.S.; Fan, Y.; Wu, Y.; Hazen, S.L. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur. Heart J. 2014, 35, 904–910. [Google Scholar] [CrossRef] [PubMed]
- Klinder, A.; Shen, Q.; Heppel, S.; Lovegrove, J.A.; Rowland, I.; Tuohy, K.M. Impact of increasing fruit and vegetables and flavonoid intake on the human gut microbiota. Food Funct. 2016, 7, 1788–1796. [Google Scholar] [CrossRef] [PubMed]
- Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Dekker Nitert, M. Increased systolic and diastolic blood pressure is associated with altered gut microbiota composition and butyrate production in early pregnancy. Hypertension 2016, 68, 974–981. [Google Scholar] [CrossRef] [PubMed]
- Heianza, Y.; Ma, W.; Manson, J.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. [Google Scholar] [CrossRef] [PubMed]
- Vázquez-Fresno, R.; Llorach, R.; Perera, A.; Mandal, R.; Feliz, M.; Tinahones, F.J.; Wishart, D.S.; Andres-Lacueva, C. Clinical phenotype clustering in cardiovascular risk patients for the identification of responsive metabotypes after red wine polyphenol intake. J. Nutr. Biochem. 2016, 28, 114–120. [Google Scholar] [CrossRef] [Green Version]
- Guasch-Ferre, M.; Hu, F.B.; Ruiz-Canela, M.; Bullo, M.; Yu, E.; Zheng, Y.; Toledo, E.; Wang, D.D.; Hruby, A.; Corella, D.; et al. Gut microbiota related plasma metabolites and risk of cardiovascular disease in the PREDIMED study. Circulation 2017, 135, A23. [Google Scholar]
- Kerley, C.P. Dietary patterns and components to prevent and treat heart failure: A comprehensive review of human studies. Nutr. Res. Rev. 2018. [Google Scholar] [CrossRef]
- Qi, J.; You, T.; Li, J.; Pan, T.; Xiang, L.; Han, Y.; Zhu, L. Circulating trimethylamine N-oxide and the risk of cardiovascular diseases: A systematic review and meta-analysis of 11 prospective cohort studies. J. Cell. Mol. Med. 2018, 22, 185–194. [Google Scholar] [CrossRef] [PubMed]
- Yao, M.E.; Liao, P.D.; Zhao, X.J.; Wang, L. Trimethylamine-N-oxide has prognostic value in coronary heart disease: A meta-analysis and dose-response analysis. BMC Cardiovasc. Disord. 2020, 20, 7. [Google Scholar] [CrossRef]
- Abbasi, J. TMAO and heart disease: The new red meat risk? JAMA 2019, 321, 2149–2151. [Google Scholar] [CrossRef] [PubMed]
- McCarty, M.F. L-carnitine consumption, its metabolism by intestinal microbiota, and cardiovascular health. Mayo. Clin. Proc. 2013, 88, 786–789. [Google Scholar] [CrossRef] [PubMed] [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] [PubMed] [Green Version]
- Ussher, J.R.; Lopaschuk, G.D.; Arduini, A. Gut microbiota metabolism of L-carnitine and cardiovascular risk. Atherosclerosis 2013, 231, 456–461. [Google Scholar] [CrossRef]
- Meyer, K.A.; Benton, T.Z.; Bennett, B.J.; Jacobs, D.R., Jr.; Lloyd-Jones, D.M.; Gross, M.D.; Carr, J.J.; Gordon-Larsen, P.; Zeisel, S.H. Microbiota-dependent metabolite trimethylamine N-oxide and coronary artery calcium in the Coronary Artery Risk Development in Young Adults Study (CARDIA). J. Am. Heart Assoc. 2016, 5. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
- Taki, K.; Tsuruta, Y.; Niwa, T. Indoxyl sulfate and atherosclerotic risk factors in hemodialysis patients. Am. J. Nephrol. 2007, 27, 30–35. [Google Scholar] [CrossRef]
- Meijers, B.K.; Verbeke, K.; Dehaen, W.; Vanrenterghem, Y.; Hoylaerts, M.F.; Evenepoel, P. The uremic retention solute p-cresyl sulfate and markers of endothelial damage. Am. J. Kidney Dis. 2009, 54, 891–901. [Google Scholar] [CrossRef]
- Buffie, C.G.; Pamer, E.G. Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol. 2013, 13, 790–801. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- Collins, S.M. A role for the gut microbiota in IBS. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 497. [Google Scholar] [CrossRef] [PubMed]
- Tsai, S.; Winer, S.; Winer, D.A. Gut T cells feast on GLP-1 to modulate cardiometabolic disease. Cell Metab. 2019, 29, 787–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alphonse, P.A.S.; Jones, P.J.H. Revisiting human cholesterol synthesis and absorption: The reciprocity paradigm and its key regulators. Lipids 2016, 51, 519–536. [Google Scholar] [CrossRef] [PubMed]
- Lyte, M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: Microbial endocrinology in the design and use of probiotics. Bioessays 2011, 33, 574–581. [Google Scholar] [CrossRef]
- Lan, T.H.; Huang, X.Q.; Tan, H.M. Vascular fibrosis in atherosclerosis. Cardiovasc. Pathol. Off. J. Soc. Cardiovasc. Pathol. 2013, 22, 401–407. [Google Scholar] [CrossRef]
- Karbach, S.H.; Schönfelder, T.; Brandão, I.; Wilms, E.; Hörmann, N.; Jäckel, S.; Schüler, R.; Finger, S.; Knorr, M.; Lagrange, J. Gut microbiota promote angiotensin II–induced arterial hypertension and vascular dysfunction. J. Am. Heart Assoc. 2016, 5, e003698. [Google Scholar] [CrossRef] [Green Version]
- Geurts, L.; Lazarevic, V.; Derrien, M.; Everard, A.; Van Roye, M.; Knauf, C.; Valet, P.; Girard, M.; Muccioli, G.G.; François, P. Altered gut microbiota and endocannabinoid system tone in obese and diabetic leptin-resistant mice: Impact on apelin regulation in adipose tissue. Front. Microbiol. 2011, 2, 149. [Google Scholar] [CrossRef] [Green Version]
- Huart, J.; Leenders, J.; Taminiau, B.; Descy, J.; Saint-Remy, A.; Daube, G.; Krzesinski, J.M.; Melin, P.; de Tullio, P.; Jouret, F. Gut microbiota and fecal levels of short-chain fatty acids differ upon 24-hour blood pressure levels in men. Hypertension 2019, 74, 1005–1013. [Google Scholar] [CrossRef]
- Loo, R.L.; Zou, X.; Appel, L.J.; Nicholson, J.K.; Holmes, E. Characterization of metabolic responses to healthy diets and association with blood pressure: Application to the Optimal Macronutrient Intake Trial for Heart Health (OmniHeart), a randomized controlled study. Am. J. Clin. Nutr. 2018, 107, 323–334. [Google Scholar] [CrossRef]
- van den Munckhof, I.C.L.; Kurilshikov, A.; ter Horst, R.; Riksen, N.P.; Joosten, L.A.B.; Zhernakova, A.; Fu, J.; Keating, S.T.; Netea, M.G.; de Graaf, J.; et al. Role of gut microbiota in chronic low-grade inflammation as potential driver for atherosclerotic cardiovascular disease: A systematic review of human studies. Obes. Rev. 2018, 19, 1719–1734. [Google Scholar] [CrossRef] [Green Version]
- Lam, O.L.T.; Zhang, W.; Samaranayake, L.P.; Li, L.S.W.; McGrath, C. A systematic review of the effectiveness of oral health promotion activities among patients with cardiovascular disease. Int. J. Cardiol. 2011, 151, 261–267. [Google Scholar] [CrossRef] [PubMed]
- Renvert, S.; Pettersson, T.; Ohlsson, O.; Persson, G.R. Bacterial profile and burden of periodontal infection in subjects with a diagnosis of acute coronary syndrome. J. Periodontol. 2006, 77, 1110–1119. [Google Scholar] [CrossRef] [PubMed]
- Pussinen, P.J.; Alfthan, G.; Rissanen, H.; Reunanen, A.; Asikainen, S.; Knekt, P. Antibodies to periodontal pathogens and stroke risk. Stroke 2004, 35, 2020–2023. [Google Scholar] [CrossRef] [Green Version]
- Beck, J.D.; Eke, P.; Heiss, G.; Madianos, P.; Couper, D.; Lin, D.; Moss, K.; Elter, J.; Offenbacher, S. Periodontal disease and coronary heart disease: A reappraisal of the exposure. Circulation 2005, 112, 19–24. [Google Scholar] [CrossRef] [Green Version]
- Yang, S.; Li, X.; Yang, F.; Zhao, R.; Pan, X.; Liang, J.; Tian, L.; Li, X.; Liu, L.; Xing, Y.; et al. Gut Microbiota-Dependent Marker TMAO in Promoting Cardiovascular Disease: Inflammation Mechanism, Clinical Prognostic, and Potential as a Therapeutic Target. Front Pharmacol. 2019, 10, 1360. [Google Scholar] [CrossRef] [PubMed]
- Mell, B.; Jala, V.R.; Mathew, A.V.; Byun, J.; Waghulde, H.; Zhang, Y.; Haribabu, B.; Vijay-Kumar, M.; Pennathur, S.; Joe, B. Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol. Genom. 2015, 47, 187–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karlsson, C.; Ahrné, S.; Molin, G.; Berggren, A.; Palmquist, I.; Fredrikson, G.N.; Jeppsson, B. Probiotic therapy to men with incipient arteriosclerosis initiates increased bacterial diversity in colon: A randomized controlled trial. Atherosclerosis 2010, 208, 228–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernández-Ruiz, I. Acute coronary syndromes: Microbial-dependent TMAO as a prognostic marker in ACS. Nat. Rev. Cardiol. 2017, 14, 128. [Google Scholar] [CrossRef]
- Organ, C.L.; Otsuka, H.; Bhushan, S.; Wang, Z.; Bradley, J.; Trivedi, R.; Polhemus, D.J.; Tang, W.W.; Wu, Y.; Hazen, S.L. Choline diet and its gut microbe–derived metabolite, trimethylamine N-oxide, exacerbate pressure overload–induced heart failure. Circ. Heart Fail. 2016, 9, e002314. [Google Scholar] [CrossRef] [Green Version]
- Koren, O.; Spor, A.; Felin, J.; Fåk, F.; Stombaugh, J.; Tremaroli, V.; Behre, C.J.; Knight, R.; Fagerberg, B.; Ley, R.E. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc. Natl. Acad. Sci. USA 2011, 108, 4592–4598. [Google Scholar] [CrossRef] [Green Version]
- Ott, S.J.; El Mokhtari, N.E.; Musfeldt, M.; Hellmig, S.; Freitag, S.; Rehman, A.; Kuhbacher, T.; Nikolaus, S.; Namsolleck, P.; Blaut, M. Detection of diverse bacterial signatures in atherosclerotic lesions of patients with coronary heart disease. Circulation 2006, 113, 929–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mattila, K.J.; Nieminen, M.S.; Valtonen, V.V.; Rasi, V.P.; Kesäniemi, Y.A.; Syrjälä, S.L.; Jungell, P.S.; Isoluoma, M.; Hietaniemi, K.; Jokinen, M.J. Association between dental health and acute myocardial infarction. BMJ 1989, 298, 779–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hyvärinen, K.; Mäntylä, P.; Buhlin, K.; Paju, S.; Nieminen, M.S.; Sinisalo, J.; Pussinen, P.J. A common periodontal pathogen has an adverse association with both acute and stable coronary artery disease. Atherosclerosis 2012, 223, 478–484. [Google Scholar] [CrossRef] [PubMed]
- Fåk, F.; Tremaroli, V.; Bergström, G.; Bäckhed, F. Oral microbiota in patients with atherosclerosis. Atherosclerosis 2015, 243, 573–578. [Google Scholar] [CrossRef]
- e Silva Filho, W.S.; Casarin, R.C.; Junior, E.L.N.; Passos, H.M.; Sallum, A.W.; Gonçalves, R.B. Microbial diversity similarities in periodontal pockets and atheromatous plaques of cardiovascular disease patients. PLoS ONE 2014, 9, e109761. [Google Scholar] [CrossRef]
- Jonsson, A.L.; Backhed, F. Role of gut microbiota in atherosclerosis. Nat. Rev. Cardiol. 2017, 14, 79–87. [Google Scholar] [CrossRef]
- Dziubak, A.; Wojcicka, G.; Wojtak, A.; Beltowski, J. Metabolic effects of metformin in the failing heart. Int. J. Mol. Sci. 2018, 19. [Google Scholar] [CrossRef] [Green Version]
- McCreight, L.J.; Bailey, C.J.; Pearson, E.R. Metformin and the gastrointestinal tract. Diabetologia 2016, 59, 426–435. [Google Scholar] [CrossRef] [Green Version]
- Munch Roager, H.; Vogt, J.K.; Kristensen, M.; Hansen, L.B.S.; Ibrügger, S.; Maerkedahl, R.B.; Bahl, M.I.; Lind, M.V.; Nielsen, R.L.; Frøkiaer, H.; et al. Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: A randomised cross-over trial. Gut 2019, 68, 83–93. [Google Scholar] [CrossRef] [Green Version]
- Plaza-Diaz, J.; Gomez-Llorente, C.; Abadia-Molina, F.; Saez-Lara, M.J.; Campana-Martin, L.; Munoz-Quezada, S.; Romero, F.; Gil, A.; Fontana, L. Effects of Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035 and Lactobacillus rhamnosus CNCM I-4036 on hepatic steatosis in Zucker rats. PLoS ONE 2014, 9, e98401. [Google Scholar] [CrossRef]
- Battson, M.L.; Lee, D.M.; Weir, T.L.; Gentile, C.L. The gut microbiota as a novel regulator of cardiovascular function and disease. J. Nutr. Biochem. 2018, 56, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Kiechl, S.; Egger, G.; Mayr, M.; Wiedermann, C.J.; Bonora, E.; Oberhollenzer, F.; Muggeo, M.; Xu, Q.; Wick, G.; Poewe, W.; et al. Chronic infections and the risk of carotid atherosclerosis: Prospective results from a large population study. Circulation 2001, 103, 1064–1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Niebauer, J.; Volk, H.D.; Kemp, M.; Dominguez, M.; Schumann, R.R.; Rauchhaus, M.; Poole-Wilson, P.A.; Coats, A.J.; Anker, S.D. Endotoxin and immune activation in chronic heart failure: A prospective cohort study. Lancet 1999, 353, 1838–1842. [Google Scholar] [CrossRef]
- Miller, M.A.; McTernan, P.G.; Harte, A.L.; Silva, N.F.; Strazzullo, P.; Alberti, K.G.; Kumar, S.; Cappuccio, F.P. Ethnic and sex differences in circulating endotoxin levels: A novel marker of atherosclerotic and cardiovascular risk in a British multi-ethnic population. Atherosclerosis 2009, 203, 494–502. [Google Scholar] [CrossRef]
- Li, J.; Lin, S.; Vanhoutte, P.M.; Woo, C.W.; Xu, A. Akkermansia muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in apoe-/- mice. Circulation 2016, 133, 2434–2446. [Google Scholar] [CrossRef] [Green Version]
- Lehr, H.A.; Sagban, T.A.; Ihling, C.; Zahringer, U.; Hungerer, K.D.; Blumrich, M.; Reifenberg, K.; Bhakdi, S. Immunopathogenesis of atherosclerosis: Endotoxin accelerates atherosclerosis in rabbits on hypercholesterolemic diet. Circulation 2001, 104, 914–920. [Google Scholar] [CrossRef] [Green Version]
- Denou, E.; Lolmede, K.; Garidou, L.; Pomie, C.; Chabo, C.; Lau, T.C.; Fullerton, M.D.; Nigro, G.; Zakaroff-Girard, A.; Luche, E.; et al. Defective NOD2 peptidoglycan sensing promotes diet-induced inflammation, dysbiosis, and insulin resistance. EMBO Mol. Med. 2015, 7, 259–274. [Google Scholar] [CrossRef]
- Galluzzo, S.; Patti, G.; Dicuonzo, G.; Di Sciascio, G.; Tonini, G.; Ferraro, E.; Spoto, C.; Campanale, R.; Zoccoli, A.; Angeletti, S. Association between NOD2/CARD15 polymorphisms and coronary artery disease: A case-control study. Hum. Immunol. 2011, 72, 636–640. [Google Scholar] [CrossRef]
- Yuan, H.; Zelkha, S.; Burkatovskaya, M.; Gupte, R.; Leeman, S.E.; Amar, S. Pivotal role of NOD2 in inflammatory processes affecting atherosclerosis and periodontal bone loss. Proc. Natl. Acad. Sci. USA 2013, 110, E5059–E5068. [Google Scholar] [CrossRef] [Green Version]
- Patel, P.; Mendall, M.A.; Carrington, D.; Strachan, D.P.; Leatham, E.; Molineaux, N.; Levy, J.; Blakeston, C.; Seymour, C.A.; Camm, A.J.; et al. Association of Helicobacter pylori and Chlamydia pneumoniae infections with coronary heart disease and cardiovascular risk factors. BMJ 1995, 311, 711–714. [Google Scholar] [CrossRef] [Green Version]
- Wright, A.T. Gut commensals make choline too. Nat. Microbiol. 2019, 4, 4–5. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, C.; Yoon, W.; Goldschmidt-Clermont, P.J. DNA methylation and atherosclerosis. J. Nutr. 2002, 132, 2406S–2409S. [Google Scholar] [CrossRef] [PubMed] [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] [PubMed]
- Salmon, W.D.; Newberne, P.M. Cardiovascular disease in choline-deficient rats. Effects of choline deficiency, nature and level of dietary lipids and proteins, and duration of feeding on plasma and liver lipid values and cardiovascular lesions. Arch. Pathol. 1962, 73, 190–209. [Google Scholar] [PubMed]
- Mudd, S.H.; Finkelstein, J.D.; Refsum, H.; Ueland, P.M.; Malinow, M.R.; Lentz, S.R.; Jacobsen, D.W.; Brattstrom, L.; Wilcken, B.; Wilcken, D.E.; et al. Homocysteine and its disulfide derivatives: A suggested consensus terminology. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1704–1706. [Google Scholar] [CrossRef] [Green Version]
- McCully, K.S.; Ragsdale, B.D. Production of arteriosclerosis by homocysteinemia. Am. J. Pathol. 1970, 61, 1–11. [Google Scholar]
- Marti-Carvajal, A.J.; Sola, I.; Lathyris, D.; Dayer, M. Homocysteine-lowering interventions for preventing cardiovascular events. Cochrane Database Syst. Rev. 2017, 8, CD006612. [Google Scholar] [CrossRef]
- Millard, H.R.; Musani, S.K.; Dibaba, D.T.; Talegawkar, S.A.; Taylor, H.A.; Tucker, K.L.; Bidulescu, A. Dietary choline and betaine; associations with subclinical markers of cardiovascular disease risk and incidence of CVD, coronary heart disease and stroke: The Jackson Heart Study. Eur. J. Nutr. 2018, 57, 51–60. [Google Scholar] [CrossRef] [Green Version]
- Magnúsdóttir, S.; Ravcheev, D.; de Crécy-Lagard, V.; Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Front. Genet. 2015, 6, 148. [Google Scholar] [CrossRef] [Green Version]
- Said, H.M. Intestinal absorption of water-soluble vitamins in health and disease. Biochem. J. 2011, 437, 357–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeon, J.; Park, K. Dietary vitamin B6 intake associated with a decreased risk of cardiovascular disease: A prospective cohort study. Nutrients 2019, 11, 1484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Group, H.T.C.; Landray, M.J.; Haynes, R.; Hopewell, J.C.; Parish, S.; Aung, T.; Tomson, J.; Wallendszus, K.; Craig, M.; Jiang, L.; et al. Effects of extended-release niacin with laropiprant in high-risk patients. N. Engl. J. Med. 2014, 371, 203–212. [Google Scholar] [CrossRef] [Green Version]
- Jenkins, D.J.A.; Spence, J.D.; Giovannucci, E.L.; Kim, Y.I.; Josse, R.; Vieth, R.; Blanco Mejia, S.; Viguiliouk, E.; Nishi, S.; Sahye-Pudaruth, S.; et al. Supplemental vitamins and minerals for CVD prevention and treatment. J. Am. Coll. Cardiol. 2018, 71, 2570–2584. [Google Scholar] [CrossRef] [PubMed]
- Aung, K.; Htay, T. Review: Folic acid may reduce risk for CVD and stroke, and B-vitamin complex may reduce risk for stroke. Ann. Intern. Med. 2018, 169, JC44. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Jin, Y.; Wang, Y.; Li, L.; Liao, Y.; Zhang, Y.; Yu, D. The effect of folic acid in patients with cardiovascular disease: A systematic review and meta-analysis. Medicine (Baltimore) 2019, 98, e17095. [Google Scholar] [CrossRef] [PubMed]
- Valentini, L.; Pinto, A.; Bourdel-Marchasson, I.; Ostan, R.; Brigidi, P.; Turroni, S.; Hrelia, S.; Hrelia, P.; Bereswill, S.; Fischer, A.; et al. Impact of personalized diet and probiotic supplementation on inflammation, nutritional parameters and intestinal microbiota—The “RISTOMED project”: Randomized controlled trial in healthy older people. Clin. Nutr. 2015, 34, 593–602. [Google Scholar] [CrossRef] [Green Version]
- Larsen, J.M. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 2017, 151, 363–374. [Google Scholar] [CrossRef] [Green Version]
- Pearson, T.A.; Mensah, G.A.; Alexander, R.W.; Anderson, J.L.; Cannon III, R.O.; Criqui, M.; Fadl, Y.Y.; Fortmann, S.P.; Hong, Y.; Myers, G.L. Markers of inflammation and cardiovascular disease: Application to clinical and public health practice: A statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation 2003, 107, 499–511. [Google Scholar] [CrossRef]
- Ostan, R.; Bene, M.C.; Spazzafumo, L.; Pinto, A.; Donini, L.M.; Pryen, F.; Charrouf, Z.; Valentini, L.; Lochs, H.; Bourdel-Marchasson, I.; et al. Impact of diet and nutraceutical supplementation on inflammation in elderly people. Results from the RISTOMED study, an open-label randomized control trial. Clin. Nutr. 2016, 35, 812–818. [Google Scholar] [CrossRef] [Green Version]
- Simone, C.D.; Ciardi, A.; Grassi, A.; Gardini, S.L.; Tzantzoglou, S.; Trinchieri, V.; Moretti, S.; Jirillo, E. Effect of Bifidobacterium bifidum and Lactobacillus acidophilus on gut mucosa and peripheral blood B lymphocytes. Immunopharmacol. Immunotoxicol. 1992, 14, 331–340. [Google Scholar] [CrossRef] [PubMed]
- Guillemard, E.; Tondu, F.; Lacoin, F.; Schrezenmeir, J. Consumption of a fermented dairy product containing the probiotic Lactobacillus casei DN-114001 reduces the duration of respiratory infections in the elderly in a randomised controlled trial. Br. J. Nutr. 2010, 103, 58–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mane, J.; Pedrosa, E.; Loren, V.; Gassull, M.A.; Espadaler, J.; Cune, J.; Audivert, S.; Bonachera, M.A.; Cabre, E. A mixture of Lactobacillus plantarum CECT 7315 and CECT 7316 enhances systemic immunity in elderly subjects. A dose-response, double-blind, placebo-controlled, randomized pilot trial. Nutr. Hosp. 2011, 26, 228–235. [Google Scholar] [PubMed]
- Dong, H.; Rowland, I.; Thomas, L.V.; Yaqoob, P. Immunomodulatory effects of a probiotic drink containing Lactobacillus casei Shirota in healthy older volunteers. Eur. J. Nutr. 2013, 52, 1853–1863. [Google Scholar] [CrossRef]
- Moro-Garcia, M.A.; Alonso-Arias, R.; Baltadjieva, M.; Fernandez Benitez, C.; Fernandez Barrial, M.A.; Diaz Ruisanchez, E.; Alonso Santos, R.; Alvarez Sanchez, M.; Saavedra Mijan, J.; Lopez-Larrea, C. Oral supplementation with Lactobacillus delbrueckii subsp. bulgaricus 8481 enhances systemic immunity in elderly subjects. Age 2013, 35, 1311–1326. [Google Scholar] [CrossRef]
- Ouwehand, A.C.; Tiihonen, K.; Saarinen, M.; Putaala, H.; Rautonen, N. Influence of a combination of Lactobacillus acidophilus NCFM and lactitol on healthy elderly: Intestinal and immune parameters. Br. J. Nutr. 2009, 101, 367–375. [Google Scholar] [CrossRef] [Green Version]
- Schiffrin, E.J.; Parlesak, A.; Bode, C.; Bode, J.C.; van’t Hof, M.A.; Grathwohl, D.; Guigoz, Y. Probiotic yogurt in the elderly with intestinal bacterial overgrowth: Endotoxaemia and innate immune functions. Br. J. Nutr. 2008, 101, 961–966. [Google Scholar] [CrossRef] [Green Version]
- Gil-Cruz, C.; Perez-Shibayama, C.; De Martin, A.; Ronchi, F.; van der Borght, K.; Niederer, R.; Onder, L.; Lutge, M.; Novkovic, M.; Nindl, V.; et al. Microbiota-derived peptide mimics drive lethal inflammatory cardiomyopathy. Science 2019, 366, 881–886. [Google Scholar] [CrossRef]
- Adnan, S.; Nelson, J.W.; Ajami, N.J.; Venna, V.R.; Petrosino, J.F.; Bryan Jr, R.M.; Durgan, D.J. Alterations in the gut microbiota can elicit hypertension in rats. Physiol. Genom. 2016, 49, 96–104. [Google Scholar] [CrossRef] [Green Version]
- Yang, T.; Santisteban, M.M.; Rodriguez, V.; Li, E.; Ahmari, N.; Carvajal, J.M.; Zadeh, M.; Gong, M.; Qi, Y.; Zubcevic, J. Gut dysbiosis is linked to hypertension. Hypertension 2015, 65, 1331–1340. [Google Scholar] [CrossRef] [Green Version]
- Guzmán-Castañeda, S.J.; Ortega-Vega, E.L.; de la Cuesta-Zuluaga, J.; Velásquez-Mejía, E.P.; Rojas, W.; Bedoya, G.; Escobar, J.S. Gut microbiota composition explains more variance in the host cardiometabolic risk than genetic ancestry. Gut Microbes 2019, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Kelly, T.N.; Bazzano, L.A.; Ajami, N.J.; He, H.; Zhao, J.; Petrosino, J.F.; Correa, A.; He, J. Gut microbiome associates with lifetime cardiovascular disease risk profile among bogalusa heart study participants. Circ. Res. 2016, 119, 956–964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Guo, Y.; Gui, Y.; Xu, D. Physical exercise, gut, gut microbiota, and atherosclerotic cardiovascular diseases. Lipids Health Dis. 2018, 17, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Denou, E.; Marcinko, K.; Surette, M.G.; Steinberg, G.R.; Schertzer, J.D. High-intensity exercise training increases the diversity and metabolic capacity of the mouse distal gut microbiota during diet-induced obesity. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E982–E993. [Google Scholar] [CrossRef]
- Lambert, J.E.; Myslicki, J.P.; Bomhof, M.R.; Belke, D.D.; Shearer, J.; Reimer, R.A. Exercise training modifies gut microbiota in normal and diabetic mice. Appl. Physiol. Nutr. Metab. 2015, 40, 749–752. [Google Scholar] [CrossRef]
- Matsumoto, M.; Inoue, R.; Tsukahara, T.; Ushida, K.; Chiji, H.; Matsubara, N.; Hara, H. Voluntary running exercise alters microbiota composition and increases n-butyrate concentration in the rat cecum. Biosci. Biotechnol. Biochem. 2008, 72, 572–576. [Google Scholar] [CrossRef]
- Queipo-Ortuno, M.I.; Seoane, L.M.; Murri, M.; Pardo, M.; Gomez-Zumaquero, J.M.; Cardona, F.; Casanueva, F.; Tinahones, F.J. Gut microbiota composition in male rat models under different nutritional status and physical activity and its association with serum leptin and ghrelin levels. PLoS ONE 2013, 8, e65465. [Google Scholar] [CrossRef]
- Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Liu, H.-Y.; Zhou, H.; Zhan, Q.; Lai, W.; Zeng, Q.; Ren, H.; Xu, D. Moderate-intensity exercise affects gut microbiome composition and influences cardiac function in myocardial infarction mice. Front. Microbiol. 2017, 8, 1687. [Google Scholar] [CrossRef]
- Bermudez-Brito, M.; Plaza-Díaz, J.; Munoz-Quezada, S.; Gomez-Llorente, C.; Gil, A. Probiotic mechanisms of action. Ann. Nutr. Metab. 2012, 61, 160–174. [Google Scholar] [CrossRef]
- Plaza-Díaz, J.; Fernández-Caballero, J.Á.; Chueca, N.; García, F.; Gómez-Llorente, C.; Sáez-Lara, M.J.; Fontana, L.; Gil, Á. Pyrosequencing analysis reveals changes in intestinal microbiota of healthy adults who received a daily dose of immunomodulatory probiotic strains. Nutrients 2015, 7, 3999–4015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, D.; Jin, D.; Huang, S.; Wu, J.; Xu, M.; Liu, T.; Dong, W.; Liu, X.; Wang, S.; Zhong, W. Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett. 2020, 469, 456–467. [Google Scholar] [CrossRef] [PubMed]
- Plaza-Díaz, J.; Ruiz-Ojeda, F.J.; Vilchez-Padial, L.M.; Gil, A. Evidence of the anti-inflammatory effects of probiotics and synbiotics in intestinal chronic diseases. Nutrients 2017, 9, 555. [Google Scholar] [CrossRef] [Green Version]
- Plaza-Díaz, J.; Robles-Sánchez, C.; Abadía-Molina, F.; Morón-Calvente, V.; Sáez-Lara, M.J.; Ruiz-Bravo, A.; Jiménez-Valera, M.; Gil, Á.; Gómez-Llorente, C.; Fontana, L. Adamdec1, Ednrb and Ptgs1/Cox1, inflammation genes upregulated in the intestinal mucosa of obese rats, are downregulated by three probiotic strains. Sci. Rep. 2017, 7, 1939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munoz-Quezada, S.; Bermudez-Brito, M.; Chenoll, E.; Genovés, S.; Gomez-Llorente, C.; Plaza-Diaz, J.; Matencio, E.; Bernal, M.J.; Romero, F.; Ramón, D. Competitive inhibition of three novel bacteria isolated from faeces of breast milk-fed infants against selected enteropathogens. Br. J. Nutr. 2013, 109, S63–S69. [Google Scholar] [CrossRef] [Green Version]
- Plaza-Diaz, J.; Gomez-Llorente, C.; Campaña-Martin, L.; Matencio, E.; Ortuño, I.; Martínez-Silla, R.; Gomez-Gallego, C.; Periago, M.J.; Ros, G.; Chenoll, E. Safety and immunomodulatory effects of three probiotic strains isolated from the feces of breast-fed infants in healthy adults: SETOPROB study. PLoS ONE 2013, 8, e78111. [Google Scholar] [CrossRef]
- Ebel, B.; Lemetais, G.; Beney, L.; Cachon, R.; Sokol, H.; Langella, P.; Gervais, P. Impact of probiotics on risk factors for cardiovascular diseases. A review. Crit. Rev. Food Sci. Nutr. 2014, 54, 175–189. [Google Scholar] [CrossRef]
- Thushara, R.M.; Gangadaran, S.; Solati, Z.; Moghadasian, M.H. Cardiovascular benefits of probiotics: A review of experimental and clinical studies. Food Funct. 2016, 7, 632–642. [Google Scholar] [CrossRef]
- Hassan, A.; Din, A.U.; Zhu, Y.; Zhang, K.; Li, T.; Wang, Y.; Luo, Y.; Wang, G. Updates in understanding the hypocholesterolemia effect of probiotics on atherosclerosis. Appl. Microbiol. Biotechnol. 2019, 103, 5993–6006. [Google Scholar] [CrossRef]
- Ejtahed, H.-S.; Angoorani, P.; Soroush, A.-R.; Atlasi, R.; Hasani-Ranjbar, S.; Mortazavian, A.M.; Larijani, B. Probiotics supplementation for the obesity management; A systematic review of animal studies and clinical trials. J. Funct. Foods 2019, 52, 228–242. [Google Scholar] [CrossRef]
- Salgaço, M.K.; Oliveira, L.G.S.; Costa, G.N.; Bianchi, F.; Sivieri, K. Relationship between gut microbiota, probiotics, and type 2 diabetes mellitus. Appl. Microbiol. Biotechnol. 2019, 103, 9229–9238. [Google Scholar] [CrossRef] [PubMed]
- DiRienzo, D.B. Effect of probiotics on biomarkers of cardiovascular disease: Implications for heart-healthy diets. Nutr. Rev. 2014, 72, 18–29. [Google Scholar] [CrossRef] [PubMed]
- Sáez-Lara, M.J.; Robles-Sanchez, C.; Ruiz-Ojeda, F.J.; Plaza-Diaz, J.; Gil, A. Effects of probiotics and synbiotics on obesity, insulin resistance syndrome, type 2 diabetes and non-alcoholic fatty liver disease: A review of human clinical trials. Int. J. Mol. Sci. 2016, 17, 928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reis, S.; Conceição, L.; Rosa, D.; Siqueira, N.; Peluzio, M. Mechanisms responsible for the hypocholesterolaemic effect of regular consumption of probiotics. Nutr. Res. Rev. 2017, 30, 36–49. [Google Scholar] [CrossRef]
- Arora, T.; Sharma, R.; Frost, G. Propionate. Anti-obesity and satiety enhancing factor? Appetite 2011, 56, 511–515. [Google Scholar] [CrossRef]
- Korhonen, H. Milk-derived bioactive peptides: From science to applications. J. Funct. Foods 2009, 1, 177–187. [Google Scholar] [CrossRef]
- Ahtesh, F.B.; Stojanovska, L.; Apostolopoulos, V. Anti-hypertensive peptides released from milk proteins by probiotics. Maturitas 2018, 115, 103–109. [Google Scholar] [CrossRef] [Green Version]
- Ardeshirlarijani, E.; Tabatabaei-Malazy, O.; Mohseni, S.; Qorbani, M.; Larijani, B.; Jalili, R.B. Effect of probiotics supplementation on glucose and oxidative stress in type 2 diabetes mellitus: A meta-analysis of randomized trials. Daru J. Pharm. Sci. 2019, 27, 827–837. [Google Scholar] [CrossRef]
- Balakumar, M.; Prabhu, D.; Sathishkumar, C.; Prabu, P.; Rokana, N.; Kumar, R.; Raghavan, S.; Soundarajan, A.; Grover, S.; Batish, V.K. Improvement in glucose tolerance and insulin sensitivity by probiotic strains of Indian gut origin in high-fat diet-fed C57BL/6J mice. Eur. J. Nutr. 2018, 57, 279–295. [Google Scholar] [CrossRef]
- Amar, J.; Chabo, C.; Waget, A.; Klopp, P.; Vachoux, C.; Bermúdez-Humarán, L.G.; Smirnova, N.; Bergé, M.; Sulpice, T.; Lahtinen, S. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: Molecular mechanisms and probiotic treatment. EMBO Mol. Med. 2011, 3, 559–572. [Google Scholar] [CrossRef]
- Olek, A.; Woynarowski, M.; Ahren, I.L.; Kierkus, J.; Socha, P.; Larsson, N.; Onning, G. Efficacy and safety of lactobacillus plantarum DSM 9843 (LP299V) in the prevention of antibiotic-associated gastrointestinal symptoms in children-randomized, double-blind, placebo-controlled study. J. Pediatr. 2017, 186, 82–86. [Google Scholar] [CrossRef] [PubMed]
- Pereg, D.; Kimhi, O.; Tirosh, A.; Orr, N.; Kayouf, R.; Lishner, M. The effect of fermented yogurt on the prevention of diarrhea in a healthy adult population. Am. J. Infect. Control 2005, 33, 122–125. [Google Scholar] [CrossRef] [PubMed]
- Jafarnejad, S.; Shab-Bidar, S.; Speakman, J.R.; Parastui, K.; Daneshi-Maskooni, M.; Djafarian, K. Probiotics Reduce the Risk of Antibiotic-Associated Diarrhea in Adults (18–64 Years) but Not the Elderly (>65 Years): A Meta-Analysis. Nutr. Clin. Pract. Off. Publ. Am. Soc. Parenter. Enter. Nutr. 2016, 31, 502–513. [Google Scholar] [CrossRef]
- Allen, S.J.; Wareham, K.; Wang, D.; Bradley, C.; Hutchings, H.; Harris, W.; Dhar, A.; Brown, H.; Foden, A.; Gravenor, M.B.; et al. Lactobacilli and bifidobacteria in the prevention of antibiotic-associated diarrhoea and Clostridium difficile diarrhoea in older inpatients (PLACIDE): A randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2013, 382, 1249–1257. [Google Scholar] [CrossRef] [Green Version]
- Zeng, W.; Shen, J.; Bo, T.; Peng, L.; Xu, H.; Nasser, M.I.; Zhuang, Q.; Zhao, M. Cutting edge: Probiotics and fecal microbiota transplantation in immunomodulation. J. Immunol. Res. 2019, 2019, 1603758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leshem, A.; Horesh, N.; Elinav, E. Fecal microbial transplantation and its potential application in cardiometabolic syndrome. Front. Immunol. 2019, 10, 1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brandt, L.J.; Aroniadis, O.C.; Mellow, M.; Kanatzar, A.; Kelly, C.; Park, T.; Stollman, N.; Rohlke, F.; Surawicz, C. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am. J. Gastroenterol. 2012, 107, 1079–1087. [Google Scholar] [CrossRef]
- van Nood, E.; Vrieze, A.; Nieuwdorp, M.; Fuentes, S.; Zoetendal, E.G.; de Vos, W.M.; Visser, C.E.; Kuijper, E.J.; Bartelsman, J.F.; Tijssen, J.G.; et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 2013, 368, 407–415. [Google Scholar] [CrossRef] [Green Version]
- Schwan, A.; Sjolin, S.; Trottestam, U.; Aronsson, B. Relapsing Clostridium difficile enterocolitis cured by rectal infusion of homologous faeces. Lancet 1983, 2, 845. [Google Scholar] [CrossRef]
- Cohen, S.H.; Gerding, D.N.; Johnson, S.M.; Kelly, C.P.; Loo, V.G.; McDonald, L.C. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect. Control Hosp. Epidemiol. 2010, 31, 431–455. [Google Scholar] [CrossRef]
- Gregory, J.C.; Buffa, J.A.; Org, E.; Wang, Z.; Levison, B.S.; Zhu, W.; Wagner, M.A.; Bennett, B.J.; Li, L.; DiDonato, J.A.; et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J. Biol. Chem. 2015, 290, 5647–5660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vrieze, A.; Van Nood, E.; Holleman, F.; Salojarvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012, 143, 913–916.e917. [Google Scholar] [CrossRef] [PubMed]
- Choi, H.H.; Cho, Y.S. Fecal microbiota transplantation: Current applications, effectiveness, and future perspectives. Clin. Endosc. 2016, 49, 257–265. [Google Scholar] [CrossRef] [PubMed]
- De Leon, L.M.; Watson, J.B.; Kelly, C.R. Transient flare of ulcerative colitis after fecal microbiota transplantation for recurrent Clostridium difficile infection. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2013, 11, 1036–1038. [Google Scholar] [CrossRef] [PubMed]
- Brandt, L.J. FMT: First step in a long journey. Am. J. Gastroenterol. 2013, 108, 1367–1368. [Google Scholar] [CrossRef]
- Zeevi, D.; Korem, T.; Zmora, N.; Israeli, D.; Rothschild, D.; Weinberger, A.; Ben-Yacov, O.; Lador, D.; Avnit-Sagi, T.; Lotan-Pompan, M.; et al. Personalized nutrition by prediction of glycemic responses. Cell 2015, 163, 1079–1094. [Google Scholar] [CrossRef] [Green Version]
- Kolodziejczyk, A.A.; Zheng, D.; Elinav, E. Diet-microbiota interactions and personalized nutrition. Nat. Rev. Microbiol. 2019, 17, 742–753. [Google Scholar] [CrossRef]
- Mendes-Soares, H.; Raveh-Sadka, T.; Azulay, S.; Ben-Shlomo, Y.; Cohen, Y.; Ofek, T.; Stevens, J.; Bachrach, D.; Kashyap, P.; Segal, L.; et al. Model of personalized postprandial glycemic response to food developed for an Israeli cohort predicts responses in Midwestern American individuals. Am. J. Clin. Nutr. 2019, 110, 63–75. [Google Scholar] [CrossRef] [Green Version]
- Kovatcheva-Datchary, P.; Nilsson, A.; Akrami, R.; Lee, Y.S.; De Vadder, F.; Arora, T.; Hallen, A.; Martens, E.; Bjorck, I.; Backhed, F. Dietary fiber-induced improvement in glucose metabolism is associated with increased abundance of prevotella. Cell Metab. 2015, 22, 971–982. [Google Scholar] [CrossRef] [Green Version]
- Martinez, I.; Lattimer, J.M.; Hubach, K.L.; Case, J.A.; Yang, J.; Weber, C.G.; Louk, J.A.; Rose, D.J.; Kyureghian, G.; Peterson, D.A.; et al. Gut microbiome composition is linked to whole grain-induced immunological improvements. ISME J. 2013, 7, 269–280. [Google Scholar] [CrossRef] [Green Version]
- Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hughes, R.L.; Marco, M.L.; Hughes, J.P.; Keim, N.L.; Kable, M.E. The role of the gut microbiome in predicting response to diet and the development of precision nutrition models-part I: Overview of current methods. Adv. Nutr. 2019, 10, 953–978. [Google Scholar] [CrossRef]
- Lacatusu, C.M.; Grigorescu, E.D.; Floria, M.; Onofriescu, A.; Mihai, B.M. The mediterranean diet: From an environment-driven food culture to an emerging medical prescription. Int. J. Environ. Res. Public Health 2019, 16, 942. [Google Scholar] [CrossRef] [Green Version]
- Ciecierska, A.; Drywien, M.E.; Hamulka, J.; Sadkowski, T. Nutraceutical functions of beta-glucans in human nutrition. Rocz. Panstw. Zakl. Hig. 2019, 70, 315–324. [Google Scholar] [CrossRef]
- He, Z.; Hao, W.; Kwek, E.; Lei, L.; Liu, J.; Zhu, H.; Ma, K.Y.; Zhao, Y.; Ho, H.M.; He, W.S.; et al. Fish oil is more potent than flaxseed oil in modulating gut microbiota and reducing trimethylamine-N-oxide-exacerbated atherogenesis. J. Agric. Food Chem. 2019, 67, 13635–13647. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; He, Z.; Ma, N.; Chen, Z.Y. Beneficial effects of dietary polyphenols on high-fat diet-induced obesity linking with modulation of gut microbiota. J. Agric. Food Chem. 2020, 68, 33–47. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Han, Y.; Song, M.; Gu, M.; Ren, D.; Zhu, X.; Cao, X.; Li, F.; Wang, W.; Cai, X.; Yuan, B.; et al. Dietary intake of whole strawberry inhibited colonic inflammation in dextran-sulfate-sodium-treated mice via restoring immune homeostasis and alleviating gut microbiota dysbiosis. J. Agric. Food Chem. 2019, 67, 9168–9177. [Google Scholar] [CrossRef] [PubMed]
- Larrosa, M.; Gonzalez-Sarrias, A.; Yanez-Gascon, M.J.; Selma, M.V.; Azorin-Ortuno, M.; Toti, S.; Tomas-Barberan, F.; Dolara, P.; Espin, J.C. Anti-inflammatory properties of a pomegranate extract and its metabolite urolithin-A in a colitis rat model and the effect of colon inflammation on phenolic metabolism. J. Nutr. Biochem. 2010, 21, 717–725. [Google Scholar] [CrossRef] [PubMed]
- Sousa, T.; Paterson, R.; Moore, V.; Carlsson, A.; Abrahamsson, B.; Basit, A.W. The gastrointestinal microbiota as a site for the biotransformation of drugs. Int. J. Pharm. 2008, 363, 1–25. [Google Scholar] [CrossRef]
- Tuteja, S.; Ferguson, J.F. Gut microbiome and response to cardiovascular drugs. Circ. Genom. Precis. Med. 2019, 12, 421–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spanogiannopoulos, P.; Bess, E.N.; Carmody, R.N.; Turnbaugh, P.J. The microbial pharmacists within us: A metagenomic view of xenobiotic metabolism. Nat. Rev. Microbiol. 2016, 14, 273–287. [Google Scholar] [CrossRef] [PubMed]
- Haiser, H.J.; Turnbaugh, P.J. Developing a metagenomic view of xenobiotic metabolism. Pharmacol. Res. 2013, 69, 21–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sanchez-Rodriguez, E.; Egea-Zorrilla, A.; Plaza-Díaz, J.; Aragón-Vela, J.; Muñoz-Quezada, S.; Tercedor-Sánchez, L.; Abadia-Molina, F. The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases. Nutrients 2020, 12, 605. https://doi.org/10.3390/nu12030605
Sanchez-Rodriguez E, Egea-Zorrilla A, Plaza-Díaz J, Aragón-Vela J, Muñoz-Quezada S, Tercedor-Sánchez L, Abadia-Molina F. The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases. Nutrients. 2020; 12(3):605. https://doi.org/10.3390/nu12030605
Chicago/Turabian StyleSanchez-Rodriguez, Estefania, Alejandro Egea-Zorrilla, Julio Plaza-Díaz, Jerónimo Aragón-Vela, Sergio Muñoz-Quezada, Luis Tercedor-Sánchez, and Francisco Abadia-Molina. 2020. "The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases" Nutrients 12, no. 3: 605. https://doi.org/10.3390/nu12030605
APA StyleSanchez-Rodriguez, E., Egea-Zorrilla, A., Plaza-Díaz, J., Aragón-Vela, J., Muñoz-Quezada, S., Tercedor-Sánchez, L., & Abadia-Molina, F. (2020). The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases. Nutrients, 12(3), 605. https://doi.org/10.3390/nu12030605