Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease
Abstract
:1. Introduction
2. Effects of Diet on GM Composition
2.1. Carbohydrates
2.2. Proteins
2.3. Fat
2.4. Vitamins
3. GM-Derived Metabolites
3.1. Trimethylamine-N-oxide
3.2. Short-Chain Fatty Acids
3.3. Bile Acid-Derived Metabolites
3.4. Amino Acids and Their Metabolites
3.5. Conjugated Linoleic Acids
4. GUT Dysbiosis and CVD
4.1. Metabolic Syndrome
4.2. Atherosclerotic Cardiovascular Disease
4.3. Acute Myocardial Infarction
4.4. Heart Failure
5. Diet Intervention to Target the Heart/GM Axis
5.1. Prebiotics, GM Remodeling and CMD
5.2. Personalized Nutrition
6. Conclusions and Future Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- Rimm, E.; Qi, L.; Rexrode, K.; Albert, C.M.; Sun, Q.; Willett, W.C.; Hu, F.B.; Manson, J.E. Diet, Lifestyle, Biomarkers, Genetic Factors, and Risk of Cardiovascular Disease in the Nurses’ Health Studies. Am. J. Public Health 2016, 106, 1616–1623. [Google Scholar] [CrossRef]
- Lichtenstein, A.H.; Appel, L.J.; Vadiveloo, M.; Hu, F.B.; Kris-Etherton, P.M.; Rebholz, C.M.; Sacks, F.M.; Thorndike, A.N.; Van Horn, L.; Wylie-Rosett, J.; et al. Dietary Guidance to Improve Cardiovascular Health: A Scientific Statement From the American Heart Association. Circulation 2021, 144, e476–e487. [Google Scholar] [CrossRef] [PubMed]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
- Brown, J.M.; Hazen, S.L. The gut microbial endocrine organ: Bacterially derived signals driving cardiometabolic diseases. Annu. Rev. Med. 2015, 66, 343–359. [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]
- 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]
- Tang, W.W.; Chen, H.-C.; Chen, C.-Y.; Yen, C.Y.; Lin, C.-J.; Prajnamitra, R.P.; Chen, L.-L.; Ruan, S.-C.; Lin, J.-H.; Lin, P.-J.; et al. Loss of Gut Microbiota Alters Immune System Composition and Cripples Postinfarction Cardiac Repair. Circulation 2019, 139, 647–659. [Google Scholar] [CrossRef] [PubMed]
- Tang, W.H.W.; Li, D.Y.; Hazen, S.L. Dietary metabolism, the gut microbiome, and heart failure. Nat. Rev. Cardiol. 2019, 16, 137–154. [Google Scholar] [CrossRef]
- 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.; Bobryshev, Y.V.; Kozarov, E.; Sobenin, I.A.; Orekhov, A.N. Role of gut microbiota in the modulation of atherosclerosis-associated immune response. Front. Microbiol. 2015, 6, 671. [Google Scholar] [CrossRef] [Green Version]
- Brown, J.M.; Hazen, S.L. Microbial modulation of cardiovascular disease. Nat. Rev. Microbiol. 2018, 16, 171–181. [Google Scholar] [CrossRef] [PubMed]
- Seo, Y.S.; Lee, H.B.; Kim, Y.; Park, H.Y. Dietary Carbohydrate Constituents Related to Gut Dysbiosis and Health. Microorganisms 2020, 8, 427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Collins, K.H.; Paul, H.A.; Hart, D.A.; Reimer, R.A.; Smith, I.C.; Rios, J.L.; Seerattan, R.A.; Herzog, W. A High-Fat High-Sucrose Diet Rapidly Alters Muscle Integrity, Inflammation and Gut Microbiota in Male Rats. Sci. Rep. 2016, 6, 37278. [Google Scholar] [CrossRef] [PubMed]
- Mastrocola, R.; Ferrocino, I.; Liberto, E.; Chiazza, F.; Cento, A.S.; Collotta, D.; Querio, G.; Nigro, D.; Bitonto, V.; Cutrin, J.C.; et al. Fructose liquid and solid formulations differently affect gut integrity, microbiota composition and related liver toxicity: A comparative in vivo study. J. Nutr. Biochem. 2018, 55, 185–199. [Google Scholar] [CrossRef]
- Kawano, Y.; Edwards, M.; Huang, Y.; Bilate, A.M.; Araujo, L.P.; Tanoue, T.; Atarashi, K.; Ladinsky, M.S.; Reiner, S.L.; Wang, H.H.; et al. Microbiota imbalance induced by dietary sugar disrupts immune-mediated protection from metabolic syndrome. Cell 2022, 185, 3501–3519.e20. [Google Scholar] [CrossRef]
- Cheng, W.L.; Li, S.J.; Lee, T.I.; Lee, T.W.; Chung, C.C.; Kao, Y.H.; Chen, Y.J. Sugar Fructose Triggers Gut Dysbiosis and Metabolic Inflammation with Cardiac Arrhythmogenesis. Biomedicines 2021, 9, 728. [Google Scholar] [CrossRef]
- Bhattacharya, T.; Ghosh, T.S.; Mande, S.S. Global Profiling of Carbohydrate Active Enzymes in Human Gut Microbiome. PLoS ONE 2015, 10, e0142038. [Google Scholar] [CrossRef] [Green Version]
- La Rosa, S.L.; Ostrowski, M.P.; de León, A.V.-P.; McKee, L.S.; Larsbrink, J.; Eijsink, V.G.; Lowe, E.C.; Martens, E.C.; Pope, P.B. Glycan processing in gut microbiomes. Curr. Opin. Microbiol. 2022, 67, 102143. [Google Scholar] [CrossRef]
- Pushpass, R.-A.G.; Alzoufairi, S.; Jackson, K.G.; Lovegrove, J.A. Circulating bile acids as a link between the gut microbiota and cardiovascular health: Impact of prebiotics, probiotics and polyphenol-rich foods. Nutr. Res. Rev. 2022, 35, 161–180. [Google Scholar] [CrossRef]
- Neis, E.P.; Dejong, C.H.; Rensen, S.S. The role of microbial amino acid metabolism in host metabolism. Nutrients 2015, 7, 2930–2946. [Google Scholar] [CrossRef] [Green Version]
- Dai, Z.L.; Wu, G.; Zhu, W.Y. Amino acid metabolism in intestinal bacteria: Links between gut ecology and host health. Front. Biosci. 2011, 16, 1768–1786. [Google Scholar] [CrossRef] [Green Version]
- Lin, R.; Liu, W.; Piao, M.; Zhu, H. A review of the relationship between the gut microbiota and amino acid metabolism. Amino Acids 2017, 49, 2083–2090. [Google Scholar] [CrossRef]
- Davila, A.M.; Blachier, F.; Gotteland, M.; Andriamihaja, M.; Benetti, P.H.; Sanz, Y.; Tomé, D. Intestinal luminal nitrogen metabolism: Role of the gut microbiota and consequences for the host. Pharmacol. Res. 2013, 68, 95–107. [Google Scholar] [CrossRef]
- Diether, N.; Willing, B. Microbial fermentation of dietary protein: An important factor in diet–microbe–host interaction. Microorganisms 2019, 7, 19. [Google Scholar] [CrossRef] [Green Version]
- Fan, P.; Li, L.; Rezaei, A.; Eslamfam, S.; Che, D.; Ma, X. Metabolites of dietary protein and peptides by intestinal microbes and their impacts on gut. Curr. Protein Pept. Sci. 2015, 16, 646–654. [Google Scholar] [CrossRef] [PubMed]
- Kim, E.; Kim, D.-B.; Park, J.-Y. Changes of mouse gut microbiota diversity and composition by modulating dietary protein and carbohydrate contents: A pilot study. Prev. Nutr. Food Sci. 2016, 21, 57–61. [Google Scholar] [CrossRef] [Green Version]
- Bekhit, A.E.A.; Giteru, S.G.; Holman, B.W.B.; Hopkins, D.L. Total volatile basic nitrogen and trimethylamine in muscle foods: Potential formation pathways and effects on human health. Compr. Rev. Food Sci. Food Saf. 2021, 20, 3620–3666. [Google Scholar] [CrossRef]
- Zhu, Y.; Lin, X.; Zhao, F.; Shi, X.; Li, H.; Li, Y.; Zhu, W.; Xu, X.; Li, C.; Zhou, G. Meat, dairy and plant proteins alter bacterial composition of rat gut bacteria. Sci. Rep. 2015, 5, 15220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.; Shi, X.; Lin, X.; Ye, K.; Xu, X.; Li, C.; Zhou, G. Beef, chicken, and soy proteins in diets induce different gut microbiota and metabolites in rats. Front. Microbiol. 2017, 8, 1395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Higuchi, Y.; Hosojima, M.; Kabasawa, H.; Kuwahara, S.; Goto, S.; Toba, K.; Kaseda, R.; Tanaka, T.; Kitamura, N.; Takihara, H.; et al. Rice endosperm protein administration to juvenile mice regulates gut microbiota and suppresses the development of high-fat diet-induced obesity and related disorders in adulthood. Nutrients 2019, 11, 2919. [Google Scholar] [CrossRef] [Green Version]
- Huang, H.; Krishnan, H.B.; Pham, Q.; Yu, L.L.; Wang, T.T.Y. Soy and Gut Microbiota: Interaction and Implication for Human Health. J. Agric. Food Chem. 2016, 64, 8695–8709. [Google Scholar] [CrossRef]
- Singh, R.K.; Chang, H.-W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Cotillard, A.; Kennedy, S.P.; Kong, L.C.; Prifti, E.; Pons, N.; Le Chatelier, E.; Almeida, M.; Quinquis, B.; Levenez, F.; Galleron, N.; et al. Dietary intervention impact on gut microbial gene richness. Nature 2013, 500, 585–588. [Google Scholar] [CrossRef]
- Wiątecka, D.; Narbad, A.; Ridgway, P.K.; Kostyra, H. The study on the impact of glycated pea proteins on human intestinal bacteria. Int. J. Food. Microbiol. 2011, 145, 267–272. [Google Scholar]
- Kim, C.H.; Park, J.; Kim, M. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw. 2014, 14, 277. [Google Scholar] [CrossRef] [Green Version]
- De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696. [Google Scholar] [CrossRef] [Green Version]
- Hertzler, S.R.; Lieblein-Boff, J.C.; Weiler, M.; Allgeier, C. Plant Proteins: Assessing Their Nutritional Quality and Effects on Health and Physical Function. Nutrients 2020, 12, 3704. [Google Scholar] [CrossRef] [PubMed]
- Pereira, P.M.; Vicente, A.F. Meat nutritional composition and nutritive role in the human diet. Meat Sci. 2013, 93, 586–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gilbert, J.A.; Bendsen, N.T.; Tremblay, A.; Astrup, A. Effect of proteins from different sources on body composition. Nutr. Metab. Cardiovasc. Dis. 2011, 21, B16–B31. [Google Scholar] [CrossRef] [PubMed]
- Mariotti, F. Plant Protein, Animal Protein, and Protein Quality. In Vegetarian and Plant-Based Diets in Health and Disease Prevention, 1st ed.; Mariotti, F., Ed.; Elsevier Academic Press: Cambridge, MA, USA, 2017; pp. 621–642. [Google Scholar]
- Wu, S.; Bhat, Z.F.; Gounder, R.S.; Ahmed, I.A.M.; Al-Juhaimi, F.Y.; Ding, Y.; Bekhit, A.E.-D.A. Effect of Dietary Protein and Processing on Gut Microbiota—A Systematic Review. Nutrients 2022, 14, 453. [Google Scholar] [CrossRef] [PubMed]
- Cândido, F.G.; Valente, F.X.; Grześkowiak, Ł.M.; Moreira, A.P.B.; Rocha, D.M.U.P.; de Cássia Gonçalves Alfenas, R. Impact of dietary fat on gut microbiota and low-grade systemic inflammation: Mechanisms and clinical implications on Obesity. Int. J. Food. Sci. Nutr. 2018, 69, 125–143. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Amar, J.; Burcelin, R.; Ruidavets, J.B.; Cani, P.D.; Fauvel, J.; Alessi, M.C.; Chamontin, B.; Ferriéres, J. Energy intake is associated with endotoxemia in apparently healthy men. Am. J. Clin. Nutr. 2008, 87, 1219–1223. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
- Gruber, L.; Hemmerling, J.; Schüppel, V.; Müller, M.; Boekschoten, M.V.; Haller, D. Maternal High-fat Diet Accelerates Development of Crohn’s Disease-like Ileitis in TNFΔARE/WT Offspring. Inflamm. Bowel Dis. 2015, 21, 2016–2025. [Google Scholar] [CrossRef]
- Choi, H.; Rao, M.C.; Chang, E.B. Gut microbiota as a transducer of dietary cues to regulate host circadian rhythms and metabolism. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 679–689. [Google Scholar] [CrossRef]
- Yoo, W.; Zieba, J.K.; Foegeding, N.J.; Torres, T.P.; Shelton, C.D.; Shealy, N.G.; Byndloss, A.J.; Cevallos, S.A.; Gertz, E.; Tiffany, C.R.; et al. High-fat diet-induced colonocyte dysfunction escalates microbiota-derived trimethylamine N-oxide. Science 2021, 373, 813–818. [Google Scholar] [CrossRef] [PubMed]
- Martinez-Guryn, K.; Hubert, N.; Frazier, K.; Urlass, S.; Musch, M.W.; Ojeda, P.; Pierre, J.F.; Miyoshi, J.; Sontag, T.J.; Cham, C.M.; et al. Small Intestine Microbiota Regulate Host Digestive and Absorptive Adaptive Responses to Dietary Lipids. Cell Host Microbe 2018, 23, 458–469.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bidu, C.; Escoula, Q.; Bellenger, S.; Spor, A.; Galan, M.; Geissler, A.; Bouchot, A.; Dardevet, D.; Morio, B.; Cani, P.D.; et al. The Transplantation of ω3 PUFA-Altered Gut Microbiota of fat-1 Mice to Wild-Type Littermates Prevents Obesity and Associated Metabolic Disorders. Diabetes 2018, 67, 1512–1523. [Google Scholar] [CrossRef] [Green Version]
- Lam, Y.Y.; Ha, C.W.Y.; Storlien, L.H.; Hoffmann, J.M.A.; Oscarsson, J.; Dinudom, A.; Mather, T.J.; Cook, D.I.; Hunt, N.H.; Caterson, I.D.; et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obesity 2015, 23, 1429–1439. [Google Scholar] [CrossRef]
- Mozaffarian, D.; Ludwig, D.S. The 2015 US Dietary Guidelines: Lifting the Ban on Total Dietary Fat. JAMA 2015, 313, 2421–2422. [Google Scholar] [CrossRef] [PubMed]
- Schoeler, M.; Caesar, R. Dietary lipids, gut microbiota and lipid metabolism. Rev. Endocr. Metab. Disord. 2019, 20, 461–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jackman, J.A.; Yoon, B.K.; Li, D.; Cho, N.J. Nanotechnology Formulations for Antibacterial Free Fatty Acids and Monoglycerides. Molecules 2016, 21, 305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, J.; Prince, A.L.; Bader, D.; Hu, M.; Ganu, R.; Baquero, K.; Blundell, P.; Harris, R.A.; Frias, A.E.; Grove, K.L.; et al. High-fat maternal diet during pregnancy persistently alters the offspring microbiome in a primate model. Nat. Commun. 2014, 5, 3889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Myles, I.A.; Fontecilla, N.M.; Janelsins, B.M.; Vithayathil, P.J.; Segre, J.A.; Datta, S.K. Parental dietary fat intake alters offspring microbiome and immunity. J. Immunol. 2013, 191, 3200–3209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wankhade, U.D.; Zhong, Y.; Kang, P.; Alfaro, M.; Chintapalli, S.V.; Thakali, K.M.; Shankar, K. Enhanced offspring predisposition to steatohepatitis with maternal high-fat diet is associated with epigenetic and microbiome alterations. PLoS ONE 2017, 12, e0175675. [Google Scholar] [CrossRef] [Green Version]
- Magnusdottir, S.; Ravcheev, D.; De Crécy-Lagard, V.; Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests cooperation among gut microbes. Front. Genet. 2015, 6, 148. [Google Scholar] [CrossRef] [Green Version]
- Pharm, V.T.; Dold, S.; Rehman, A.; Bird, J.K.; Steinert, R.E. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr. Res. 2021, 95, 35–53. [Google Scholar] [CrossRef]
- Pham, V.T.; Calatayud, M.; Rotsaert, C.; Seifert, N.; Richard, N.; Abbeele, P.V.D.; Marzorati, M.; Steinert, R.E. Antioxidant Vitamins and Prebiotic FOS and XOS Differentially Shift Microbiota Composition and Function and Improve Intestinal Epithelial Barrier In Vitro. Nutrients 2021, 13, 1125. [Google Scholar] [CrossRef]
- Steinert, R.E.; Sadaghian Sadabad, M.; Harmsen, H.J.; Weber, P. The prebiotic concept and human health: A changing landscape with riboflavin as a novel prebiotic candidate? Eur. J. Clin. Nutr. 2016, 70, 1348–1353. [Google Scholar] [CrossRef] [PubMed]
- Kau, A.L.; Ahern, P.P.; Griffin, N.W.; Goodman, A.L.; Gordon, J.I. Human nutrition, the gut microbiome and the immune system. Nature 2011, 474, 327–336. [Google Scholar] [CrossRef] [Green Version]
- Hibberd, M.C.; Wu, M.; Rodionov, D.A.; Li, X.; Cheng, J.; Griffin, N.W.; Barratt, M.J.; Giannone, R.J.; Hettich, R.L.; Osterman, A.L.; et al. The effects of micronutrient deficiencies on bacterial species from the human gut microbiota. Sci. Transl. Med. 2017, 9, eaal4069. [Google Scholar] [CrossRef] [Green Version]
- De Medeiros, P.H.Q.S.; Pinto, D.V.; De Almeida, J.Z.; Rêgo, J.M.C.; Rodrigues, F.A.P.; Lima, A.M.; Bolick, D.T.; Guerrant, R.L.; Oriá, R.B. Modulation of Intestinal Immune and Barrier Functions by Vitamin A: Implications for Current Understanding of Malnutrition and Enteric Infections in Children. Nutrients 2018, 10, 1128. [Google Scholar] [CrossRef] [Green Version]
- Czarnewski, P.; Das, S.; Parigi, S.M.; Villablanca, E.J. Retinoic Acid and Its Role in Modulating Intestinal Innate Immunity. Nutrients 2017, 9, 68. [Google Scholar] [CrossRef] [Green Version]
- Nakajima, A.; Vogelzang, A.; Maruya, M.; Miyajima, M.; Murata, M.; Son, A.; Kuwahara, T.; Tsuruyama, T.; Yamada, S.; Matsuura, M.; et al. IgA regulates the composi- tion and metabolic function of gut microbiota by promoting symbiosis between bacteria. J. Exp. Med. 2018, 215, 2019–2034. [Google Scholar] [CrossRef] [Green Version]
- Iyer, N.; Vaishnava, S. Vitamin A at the interface of host-commensal-pathogen interactions. PLoS Pathog. 2019, 15, e1007750. [Google Scholar] [CrossRef] [Green Version]
- McDaniel, K.L.; Restori, K.H.; Dodds, J.W.; Kennett, M.J.; Ross, A.C.; Cantorna, M.T. Vitamin A-Deficient Hosts Become Nonsymptomatic Reservoirs of Escherichia coli-Like Enteric Infections. Infect. Immun. 2015, 83, 2984–2991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellerba, F.; Muzio, V.; Gnagnarella, P.; Facciotti, F.; Chiocca, S.; Bossi, P.; Cortinovis, D.; Chiaradonna, F.; Serrano, D.; Raimondi, S.; et al. The Association between Vitamin D and Gut Microbiota: A Systematic Review of Human Studies. Nutrients 2021, 13, 3378. [Google Scholar] [CrossRef] [PubMed]
- Sukik, A.; Alalwani, J.; Ganji, V. Vitamin D, Gut Microbiota, and Cardiometabolic Diseases-A Possible Three-Way Axis. Int. J. Mol. Sci. 2023, 24, 940. [Google Scholar] [CrossRef]
- Al-Khaldy, N.S.; Al Musharaf, S.; Aljazairy, E.A.; Hussain, S.D.; Alnaami, A.M.; Al-Daghri, N.; Aljuraiban, G. Serum Vitamin D Level and Gut Microbiota in Women. Healthcare 2023, 11, 351. [Google Scholar] [CrossRef]
- Lee, W.J.; Hase, K. Gut microbiota-generated metabolites in animal health and disease. Nat. Chem. Biol. 2014, 10, 416–424. [Google Scholar] [CrossRef] [PubMed]
- Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- Talmor-Barkan, Y.; Bar, N.; Shaul, A.A.; Shahaf, N.; Godneva, A.; Bussi, Y.; Lotan-Pompan, M.; Weinberger, A.; Shechter, A.; Chezar-Azerrad, C.; et al. Metabolomic and microbiome profiling reveals personalized risk factors for coronary artery disease. Nat. Med. 2022, 28, 295–302. [Google Scholar] [CrossRef] [PubMed]
- Kalnins, G.; Kuka, J.; Grinberga, S.; Makrecka-Kuka, M.; Liepinsh, E.; Dambrova, M.; Tars, K. Structure and Function of CutC Choline Lyase from Human Microbiota Bacterium Klebsiella pneumoniae. J. Biol. Chem. 2015, 290, 21732–21740. [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]
- 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, 1600324. [Google Scholar] [CrossRef]
- Wu, W.K.; Chen, C.C.; Liu, P.Y.; Panyod, S.; Liao, B.Y.; Chen, P.C.; Kao, H.L.; Kuo, H.C.; Kuo, C.H.; Chiu, T.H.T.; et al. Identification of TMAO-producer phenotype and host-diet-gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut. 2019, 68, 1439–1449. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- 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] [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] [Green Version]
- 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]
- Tang, W.H.; 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, 1575–1578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Tang, W.H.; 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] [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. Gamma-butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab. 2014, 20, 799–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Senthong, V.; Wang, Z.; Li, X.S.; Fan, Y.; Wu, Y.; Tang, W.H.; 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]
- Senthong, V.; Li, X.S.; Hudec, T.; Coughlin, J.; Wu, Y.; Levison, B.; Wang, Z.; Hazen, S.L.; Tang, W.H. Plasma trimethylamine N-oxide, a gut microbe-generated phosphatidylcholine metabolite, is associated with atherosclerotic burden. J. Am. Coll. Cardiol. 2016, 67, 2620–2628. [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] [PubMed] [Green Version]
- Tang, W.H.; Wang, Z.; Fan, Y.; Levison, B.; Hazen, J.E.; Donahue, L.M.; Wu, Y.; Hazen, S.L. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: Refining the gut hypothesis. J. Am. Coll. Cardiol. 2014, 64, 1908–1914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Organ, C.L.; Otsuka, H.; Bhushan, S.; Wang, Z.; Bradley, J.; Trivedi, R.; Polhemus, D.J.; Tang, W.H.; Wu, Y.; Hazen, S.L.; et al. 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] [PubMed] [Green Version]
- 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]
- Tang, W.H.; Wang, Z.; Kennedy, D.J.; Wu, Y.; Buffa, J.A.; Agatisa-Boyle, B.; Li, X.S.; Levison, B.S.; Hazen, S.L. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ. Res. 2015, 116, 448–455. [Google Scholar] [CrossRef] [PubMed] [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]
- Tang, W.H.; 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] [PubMed] [Green Version]
- Miao, J.; Ling, A.V.; Manthena, P.V.; Gearing, M.E.; Graham, M.J.; Crooke, R.M.; Croce, K.J.; Esquejo, R.M.; Clish, C.B.; Torrecilla, E.; et al. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis. Nat. Commun. 2015, 6, 6498. [Google Scholar] [CrossRef] [Green Version]
- Schugar, R.C.; Shih, D.M.; Warrier, M.; Helsley, R.N.; Burrows, A.; Ferguson, D.; Brown, A.L.; Gromovsky, A.D.; Heine, M.; Chatterjee, A.; et al. The TMAO-Producing Enzyme Flavin-Containing Monooxygenase 3 Regulates Obesity and the Beiging of White Adipose Tissue. Cell Rep. 2017, 19, 2451–2461. [Google Scholar] [CrossRef] [Green Version]
- Zhou, X.; Jin, M.; Liu, L.; Yu, Z.; Lu, X.; Zhang, H. Trimethylamine N-oxide and cardiovascular outcomes in patients with chronic heart failure after myocardial infarction. ESC Heart Fail. 2020, 7, 188–193. [Google Scholar] [CrossRef] [Green Version]
- Simó, C.; García-Cañas, V. Dietary bioactive ingredients to modulate the gut microbiota-derived metabolite TMAO. New opportunities for functional food development. Food Funct. 2020, 11, 6745–6776. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Chen, M.L.; Yi, L.; Zhang, Y.; Zhou, X.; Ran, L.; Yang, J.; Zhu, J.D.; Zhang, Q.Y.; Mi, M.T. Resveratrol Attenuates Trimethylamine-N-Oxide (TMAO)-Induced Atherosclerosis by Regulating TMAO Synthesis and Bile Acid Metabolism via Remodeling of the Gut Microbiota. mBio 2016, 7, e02210-15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ke, Y.; Li, D.; Zhao, M.; Liu, C.; Liu, J.; Zeng, A.; Shi, X.; Cheng, S.; Pan, B.; Zheng, L.; et al. Gut flora-dependent metabolite Trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress. Free Radic. Biol. Med. 2018, 116, 88–100. [Google Scholar] [CrossRef]
- Ma, G.; Pan, B.; Chen, Y.; Guo, C.; Zhao, M.; Zheng, L.; Chen, B. Trimethylamine N-oxide in atherogenesis: Impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci. Rep. 2017, 37, BSR20160244. [Google Scholar] [CrossRef] [Green Version]
- Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.; 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] [PubMed] [Green Version]
- Zhang, X.; Li, Y.; Yang, P.; Liu, X.; Lu, L.; Chen, Y.; Zhong, X.; Li, Z.; Liu, H.; Ou, C.; et al. Trimethylamine-N-Oxide Promotes Vascular Calcification through Activation of NLRP3 (Nucleotide-Binding Domain, Leucine-Rich-Containing Family, Pyrin Domain-Containing-3) Inflammasome and NF-κB (Nuclear Factor κB) Signals. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 751–765. [Google Scholar] [CrossRef]
- Cheng, X.; Qiu, X.; Liu, Y.; Yuan, C.; Yang, X. Trimethylamine N-oxide promotes tissue factor expression and activity in vascular endothelial cells: A new link between trimethylamine N-oxide and atherosclerotic thrombosis. Thromb. Res. 2019, 177, 110–116. [Google Scholar] [CrossRef]
- 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]
- Costabile, G.; Vetrani, C.; Bozzetto, L.; Giacco, R.; Bresciani, L.; Del Rio, D.; Vitale, M.; Della Pepa, G.; Brighenti, F.; Riccardi, G.; et al. Plasma TMAO increase after healthy diets: Results from 2 randomized controlled trials with dietary fish, polyphenols, and whole-grain cereals. Am. J. Clin. Nutr. 2021, 114, 1342–1350. [Google Scholar] [CrossRef]
- Bergeron, N.; Williams, P.T.; Lamendella, R.; Faghihnia, N.; Grube, A.; Li, X.; Wang, Z.; Knight, R.; Jansson, J.K.; Hazen, S.L.; et al. Diets high in resistant starch increase plasma levels of trimethylamine-N-oxide, a gut microbiome metabolite associated with CVD risk. Br. J. Nutr. 2016, 116, 2020–2029. [Google Scholar] [CrossRef] [Green Version]
- Koay, Y.C.; Chen, Y.-C.; Wali, J.A.; Luk, A.W.S.; Li, M.; Doma, H.; Reimark, R.; Zaldivia, M.T.K.; Habtom, H.T.; Franks, A.; et al. Plasma levels of trimethylamine-N-oxide can be increased with ‘healthy’ and ‘unhealthy’ diets and do not correlate with the extent of atherosclerosis but with plaque instability. Cardiovasc. Res. 2021, 117, 435–449. [Google Scholar] [CrossRef]
- Chen, S.; Henderson, A.; Petriello, M.C.; Romano, K.A.; Gearing, M.; Miao, J.; Schell, M.; Sandoval-Espinola, W.J.; Tao, J.; Sha, B.; et al. Trimethylamine N-Oxide Binds and Activates PERK to Promote Metabolic Dysfunction. Cell Metab. 2019, 30, 1141–1151.e5. [Google Scholar] [CrossRef]
- Jaworska, K.; Bielinska, K.; Gawrys-Kopczynska, M.; Ufnal, M. TMA (trimethylamine), but not its oxide TMAO (trimethylamine-oxide), exerts haemodynamic effects: Implications for interpretation of cardiovascular actions of gut microbiome. Cardiovasc. Res. 2019, 115, 1948–1949. [Google Scholar] [CrossRef]
- Restini, C.B.A.; Fink, G.D.; Watts, S.W. Vascular reactivity stimulated by TMA and TMAO: Are perivascular adipose tissue and endothelium involved? Pharmacol. Res. 2021, 163, 105273. [Google Scholar] [CrossRef]
- Wang, J.; Jia, H. Metagenome-wide association studies: Fine-mining the microbiome. Nat. Rev. Microbiol. 2016, 14, 508–522. [Google Scholar] [CrossRef] [PubMed]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deleu, S.; Machiels, K.; Raes, J.; Verbeke, K.; Vermeire, S. Short chain fatty acids and its producing organisms: An overlooked therapy for IBD? EBioMedicine. 2021, 66, 103293. [Google Scholar] [CrossRef] [PubMed]
- Brubaker, P.L.; Anini, Y. Direct and indirect mechanisms regulating secretion of glucagon-like peptide-1 and glucagon-like peptide-2. Can. J. Physiol. Pharmacol. 2003, 81, 1005–1012. [Google Scholar] [CrossRef]
- Sun, J.; Furio, L.; Mecheri, R.; van der Does, A.M.; Lundeberg, E.; Saveanu, L.; Chen, Y.; van Endert, P.; Agerberth, B.; Diana, J. Pancreatic β-Cells Limit Autoimmune Diabetes via an Immunoregulatory Antimicrobial Peptide Expressed under the Influence of the Gut Microbiota. Immunity 2015, 43, 304–317. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Li, Y.; Yang, S.; Lu, J.; Jin, X.; Wu, M. Diet-gut microbiota-epigenetics in metabolic diseases: From mechanisms to therapeutics. Biomed. Pharmacother. 2022, 153, 113290. [Google Scholar] [CrossRef]
- De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.; Crowley, J.; Yanagisawa, M.; et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA 2008, 105, 16767–16772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Delaere, F.; Duchampt, A.; Mounien, L.; Seyer, P.; Duraffourd, C.; Zitoun, C.; Thorens, B.; Mithieux, G. The role of sodium-coupled glucose co-transporter 3 in the satiety effect of portal glucose sensing. Mol. Metab. 2012, 2, 47–53. [Google Scholar] [CrossRef] [PubMed]
- Gautier-Stein, A.; Mithieux, G. Intestinal gluconeogenesis: Metabolic benefits make sense in the light of evolution. Nat. Rev. Gastroenterol. Hepatol. 2022, 20, 183–194. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; van Esch, B.C.A.M.; Henricks, P.A.J.; Folkerts, G.; Garssen, J. The Anti-inflammatory Effects of Short Chain Fatty Acids on Lipopolysaccharide- or Tumor Necrosis Factor α-Stimulated Endothelial Cells via Activation of GPR41/43 and Inhibition of HDACs. Front. Pharmacol. 2018, 9, 533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Dawson, P.A.; Lan, T.; Rao, A. Bile acid transporters. J. Lipid Res. 2009, 50, 2340–2357. [Google Scholar] [CrossRef] [Green Version]
- Joyce, S.A.; Gahan, C.G. Bile Acid Modifications at the Microbe-Host Interface: Potential for Nutraceutical and Pharmaceutical Interventions in Host Health. Annu. Rev. Food Sci. Technol. 2016, 7, 313–333. [Google Scholar] [CrossRef]
- Devlin, A.S.; Fischbach, M.A. A biosynthetic pathway for a prominent class of microbiota-derived bile acids. Nat. Chem. Biol. 2015, 11, 685–690. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Chen, J.; Hollister, K.; Sowers, L.C.; Forman, B.M. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol. Cell 1999, 3, 543–553. [Google Scholar] [CrossRef]
- Kawamata, Y.; Fujii, R.; Hosoya, M.; Harada, M.; Yoshida, H.; Miwa, M.; Fukusumi, S.; Habata, Y.; Itoh, T.; Shintani, Y.; et al. A G protein-coupled receptor responsive to bile acids. J. Biol. Chem. 2003, 278, 9435–9440. [Google Scholar] [CrossRef] [Green Version]
- Rosen, H.; Gonzalez-Cabrera, P.J.; Sanna, M.G.; Brown, S. Sphingosine 1-phosphate receptor signaling. Annu. Rev. Biochem. 2009, 78, 743–768. [Google Scholar] [CrossRef]
- Raufman, J.P.; Chen, Y.; Zimniak, P.; Cheng, K. Deoxycholic acid conjugates are muscarinic cholinergic receptor antagonists. Pharmacology 2002, 65, 215–221. [Google Scholar] [CrossRef] [PubMed]
- Massafra, V.; Pellicciari, R.; Gioiello, A.; van Mil, S.W.C. Progress and challenges of selective Farnesoid X Receptor modulation. Pharmacol. Ther. 2018, 191, 162–177. [Google Scholar] [CrossRef] [PubMed]
- Calkin, A.C.; Tontonoz, P. Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR. Nat. Rev. Mol. Cell Biol. 2012, 13, 213–224. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.T.; Swales, K.E.; Thomas, G.J.; Warner, T.D.; Bishop-Bailey, D. Farnesoid x receptor ligands inhibit vascular smooth muscle cell inflammation and migration. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2606–2611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Li, J.; Weng, X.; Lan, X.; Chi, X. Farnesoid X receptor agonist CDCA reduces blood pressure and regulates vascular tone in spontaneously hypertensive rats. J. Am. Soc. Hypertens. 2015, 9, 507–516.e7. [Google Scholar] [CrossRef]
- Watanabe, M.; Houten, S.M.; Mataki, C.; Christoffolete, M.A.; Kim, B.W.; Sato, H.; Messaddeq, N.; Harney, J.W.; Ezaki, O.; Kodama, T.; et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 2006, 439, 484–489. [Google Scholar] [CrossRef]
- Thomas, C.; Gioiello, A.; Noriega, L.; Strehle, A.; Oury, J.; Rizzo, G.; Macchiarulo, A.; Yamamoto, H.; Mataki, C.; Pruzanski, M.; et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009, 10, 167–177. [Google Scholar] [CrossRef] [Green Version]
- Gill, S.R.; Pop, M.; Deboy, R.T.; Eckburg, P.B.; Turnbaugh, P.J.; Samuel, B.S.; Gordon, J.I.; Relman, D.A.; Fraser-Liggett, C.M.; Nelson, K.E. Metagenomic analysis of the human distal gut microbiome. Science 2006, 312, 1355–1359. [Google Scholar] [CrossRef] [Green Version]
- Ciarlo, E.; Heinonen, T.; Herderschee, J.; Fenwick, C.; Mombelli, M.; Le Roy, D.; Roger, T. Impact of the microbial derived short chain fatty acid propionate on host susceptibility to bacterial and fungal infections in vivo. Sci. Rep. 2016, 6, 37944. [Google Scholar] [CrossRef]
- Dodd, D.; Spitzer, M.H.; Van Treuren, W.; Merrill, B.D.; Hryckowian, A.J.; Higginbottom, S.K.; Le, A.; Cowan, T.M.; Nolan, G.P.; Fischbach, M.A.; et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017, 551, 648–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Venkatesh, M.; Mukherjee, S.; Wang, H.; Li, H.; Sun, K.; Benechet, A.P.; Qiu, Z.; Maher, L.; Redinbo, M.R.; Phillips, R.S.; et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity 2014, 41, 296–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013, 39, 372–385. [Google Scholar] [CrossRef] [Green Version]
- Xue, H.; Chen, X.; Yu, C.; Deng, Y.; Zhang, Y.; Chen, S.; Chen, X.; Chen, K.; Yang, Y.; Ling, W. Gut Microbially Produced Indole-3-Propionic Acid Inhibits Atherosclerosis by Promoting Reverse Cholesterol Transport and Its Deficiency Is Causally Related to Atherosclerotic Cardiovascular Disease. Circ. Res. 2022, 131, 404–420. [Google Scholar] [CrossRef]
- YLi, Q.; You, Y.; Zeng, Y.; Wang, X.; Pan, Z.; Pang, J.; Chen, Q.; Zhou, Y.; Jin, Y.; Yang, Y.; et al. Associations between plasma tryptophan and indole-3-propionic acid levels and mortality in patients with coronary artery disease. Am. J. Clin. Nutr. 2022, 116, 1070–1077. [Google Scholar] [CrossRef]
- Zhao, M.; Zhao, L.; Xiong, X.; He, Y.; Huang, W.; Liu, Z.; Ji, L.; Pan, B.; Guo, X.; Wang, L.; et al. TMAVA, a Metabolite of Intestinal Microbes, Is Increased in Plasma From Patients With Liver Steatosis, Inhibits γ-Butyrobetaine Hydroxylase, and Exacerbates Fatty Liver in Mice. Gastroenterology 2020, 158, 2266–2281.e27. [Google Scholar] [CrossRef]
- Zhao, M.; Wei, H.; Li, C.; Zhan, R.; Liu, C.; Gao, J.; Yi, Y.; Cui, X.; Shan, W.; Ji, L.; et al. Gut microbiota production of trimethyl-5-aminovaleric acid reduces fatty acid oxidation and accelerates cardiac hypertrophy. Nat. Commun. 2022, 13, 1757. [Google Scholar] [CrossRef] [PubMed]
- Nemet, I.; Saha, P.P.; Gupta, N.; Zhu, W.; Romano, K.A.; Skye, S.M.; Cajka, T.; Mohan, M.L.; Li, L.; Wu, Y.; et al. A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors. Cell 2020, 180, 862–877.e22. [Google Scholar] [CrossRef]
- Romano, K.A.; Nemet, I.; Saha, P.P.; Haghikia, A.; Li, X.S.; Mohan, M.L.; Lovano, B.; Castel, L.; Witkowski, M.; Buffa, J.A.; et al. Gut Microbiota-Generated Phenylacetylglutamine and Heart Failure. Circ. Heart Fail. 2022, 16, e009972. [Google Scholar] [CrossRef]
- Zhu, Y.; Dwidar, M.; Nemet, I.; Buffa, J.A.; Sangwan, N.; Li, X.S.; Anderson, J.T.; Romano, K.A.; Fu, X.; Funabashi, M.; et al. Two distinct gut microbial pathways contribute to meta-organismal production of phenylacetylglutamine with links to cardiovascular disease. Cell Host Microbe 2023, 31, 18–32.e9. [Google Scholar] [CrossRef]
- Kishino, S.; Takeuchi, M.; Park, S.-B.; Hirata, A.; Kitamura, N.; Kunisawa, J.; Kiyono, H.; Iwamoto, R.; Isobe, Y.; Arita, M.; et al. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc. Natl. Acad. Sci. USA 2013, 110, 17808–17813. [Google Scholar] [CrossRef] [Green Version]
- Ecker, J.; Liebisch, G.; Patsch, W.; Schmitz, G. The conjugated linoleic acid isomer trans-9,trans-11 is a dietary occurring agonist of liver X receptor alpha. Biochem. Biophys. Res. Commun. 2009, 388, 660–666. [Google Scholar] [CrossRef] [PubMed]
- Coakley, M.; Ross, R.P.; Nordgren, M.; Fitzgerald, G.; Devery, R.; Stanton, C. Conjugated linoleic acid biosynthesis by human-derived Bifidobacterium species. J. Appl. Microbiol. 2003, 94, 138–145. [Google Scholar] [CrossRef]
- Lee, H.Y.; Park, J.H.; Seok, S.H.; Baek, M.W.; Kim, D.J.; Lee, K.E.; Paek, K.S.; Lee, Y.; Park, J.H. Human originated bacteria, Lactobacillus rhamnosus PL60, produce conjugated linoleic acid and show anti-obesity effects in diet-induced obese mice. Biochim. Biophys. Acta 2006, 1761, 736–744. [Google Scholar] [CrossRef]
- Goto, T.; Kim, Y.I.; Furuzono, T.; Takahashi, N.; Yamakuni, K.; Yang, H.E.; Li, Y.; Ohue, R.; Nomura, W.; Sugawara, T.; et al. 10-oxo-12(Z)-octadecenoic acid, a linoleic acid metabolite produced by gut lactic acid bacteria, potently activates PPARγ and stimulates adipogenesis. Biochem. Biophys. Res. Commun. 2015, 459, 597–603. [Google Scholar] [CrossRef] [PubMed]
- Miyamoto, J.; Mizukure, T.; Park, S.B.; Kishino, S.; Kimura, I.; Hirano, K.; Bergamo, P.; Rossi, M.; Suzuki, T.; Arita, M.; et al. A gut microbial metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, ameliorates intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. J. Biol. Chem. 2015, 290, 2902–2918. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Violi, F.; Cammisotto, V.; Bartimoccia, S.; Pignatelli, P.; Carnevale, R.; Nocella, C. Gut-derived low-grade endotoxaemia, atherothrombosis and cardiovascular disease. Nat. Rev. Cardiol. 2023, 20, 24–37. [Google Scholar] [CrossRef]
- Caruso, R.; Warner, N.; Inohara, N.; Núñez, G. NOD1 and NOD2: Signaling, host defense, and inflammatory disease. Immunity 2014, 41, 898–908. [Google Scholar] [CrossRef] [Green Version]
- González-Ramos, S.; Fernández-García, V.; Recalde, M.; Rodríguez, C.; Martínez-González, J.; Andrés, V.; Martín-Sanz, P.; Boscá, L. Deletion or Inhibition of NOD1 Favors Plaque Stability and Attenuates Atherothrombosis in Advanced Atherogenesis. Cells 2020, 9, 2067. [Google Scholar] [CrossRef]
- Nishio, H.; Kanno, S.; Onoyama, S.; Ikeda, K.; Tanaka, T.; Kusuhara, K.; Fujimoto, Y.; Fukase, K.; Sueishi, K.; Hara, T. Nod1 ligands induce site-specific vascular inflammation. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 1093–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- El Mokhtari, N.E.; Ott, S.J.; Nebel, A.; Simon, R.; Schreiber, S. A functional variant in the CARD4 gene and risk of premature coronary heart disease. Int. J. Immunogenet. 2006, 33, 307–311. [Google Scholar] [CrossRef]
- Justina, V.D.; Giachini, F.R.; Priviero, F.; Webb, R.C. Double-stranded RNA and Toll-like receptor activation: A novel mechanism for blood pressure regulation. Clin. Sci. 2020, 134, 303–313. [Google Scholar] [CrossRef]
- Lundberg, A.M.; Ketelhuth, D.F.; Johansson, M.E.; Gerdes, N.; Liu, S.; Yamamoto, M.; Akira, S.; Hansson, G.K. Toll-like receptor 3 and 4 signalling through the TRIF and TRAM adaptors in haematopoietic cells promotes atherosclerosis. Cardiovasc. Res. 2013, 99, 364–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kang, J.Y.; Nan, X.; Jin, M.S.; Youn, S.J.; Ryu, Y.H.; Mah, S.; Han, S.H.; Lee, H.; Paik, S.G.; Lee, J.O. Recognition of lipopeptide patterns by Toll-like receptor 2-Toll-like receptor 6 heterodimer. Immunity 2009, 31, 873–884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Little, P.J.; Downey, L.; Afroz, R.; Wu, Y.; Ta, H.T.; Xu, S.; Kamato, D. The Role of Toll-like Receptors in Atherothrombotic Cardiovascular Disease. ACS Pharmacol. Transl. Sci. 2020, 3, 457–471. [Google Scholar] [CrossRef]
- Koulis, C.; Chen, Y.C.; Hausding, C.; Ahrens, I.; Kyaw, T.S.; Tay, C.; Allen, T.; Jandeleit-Dahm, K.; Sweet, M.J.; Akira, S.; et al. Protective role for Toll-like receptor-9 in the development of atherosclerosis in apolipoprotein E-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 516–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vijay-Kumar, M.; Aitken, J.D.; Carvalho, F.A.; Cullender, T.C.; Mwangi, S.; Srinivasan, S.; Sitaraman, S.V.; Knight, R.; Ley, R.E.; Gewirtz, A.T. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 2010, 328, 228–231. [Google Scholar] [CrossRef] [Green Version]
- Yu, L.; Feng, Z. The Role of Toll-Like Receptor Signaling in the Progression of Heart Failure. Mediat. Inflamm. 2018, 2018, 9874109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498, 99–103. [Google Scholar] [CrossRef] [PubMed]
- Forslund, K.; Hildebrand, F.; Nielsen, T.; Falony, G.; Le Chatelier, E.; Sunagawa, S.; Prifti, E.; Vieira-Silva, S.; Gudmundsdottir, V.; Krogh Pedersen, H.; et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015, 528, 262–266. [Google Scholar] [CrossRef] [Green Version]
- Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Belda, E.; Voland, L.; Tremaroli, V.; Falony, G.; Adriouch, S.; Assmann, K.E.; Prifti, E.; Aron-Wisnewsky, J.; Debédat, J.; Le Roy, T.; et al. Impairment of gut microbial biotin metabolism and host biotin status in severe obesity: Effect of biotin and prebiotic supplementation on improved metabolism. Gut 2022, 71, 2463–2480. [Google Scholar] [CrossRef]
- Amedei, A.; Morbidelli, L. Circulating Metabolites Originating from Gut Microbiota Control Endothelial Cell Function. Molecules 2019, 24, 3992. [Google Scholar] [CrossRef] [Green Version]
- Shi, G.; Lin, Y.; Wu, Y.; Zhou, J.; Cao, L.; Chen, J.; Li, Y.; Tan, N.; Zhong, S. Bacteroides fragilis Supplementation Deteriorated Metabolic Dysfunction, Inflammation, and Aorta Atherosclerosis by Inducing Gut Microbiota Dysbiosis in Animal Model. Nutrients 2022, 14, 2199. [Google Scholar] [CrossRef]
- Filardo, S.; Di Pietro, M.; Farcomeni, A.; Schiavoni, G.; Sessa, R. Chlamydia pneumoniae-Mediated Inflammation in Atherosclerosis: A Meta-Analysis. Mediat. Inflamm. 2015, 2015, 378658. [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.; et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4592–4598. [Google Scholar] [CrossRef] [Green Version]
- Mitra, S.; Drautz-Moses, D.I.; Alhede, M.; Maw, M.T.; Liu, Y.; Purbojati, R.W.; Yap, Z.H.; Kushwaha, K.K.; Gheorghe, A.G.; Bjarnsholt, T.; et al. In silico analyses of metagenomes from human atherosclerotic plaque samples. Microbiome 2015, 3, 38. [Google Scholar] [CrossRef] [PubMed] [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]
- Song, T.; Guan, X.; Wang, X.; Qu, S.; Zhang, S.; Hui, W.; Men, L.; Chen, X. Dynamic modulation of gut microbiota improves post-myocardial infarct tissue repair in rats via butyric acid-mediated histone deacetylase inhibition. FASEB J. 2021, 35, e21385. [Google Scholar] [CrossRef]
- Liu, C.; Sun, Z.; Shali, S.; Mei, Z.; Chang, S.; Mo, H.; Xu, L.; Pu, Y.; Guan, H.; Chen, G.C.; et al. The gut microbiome and microbial metabolites in acute myocardial infarction. J. Genet. Genom. 2022, 49, 569–578. [Google Scholar] [CrossRef]
- Van Parys, A.; Lysne, V.; Svingen, G.F.T.; Ueland, P.M.; Dhar, I.; Øyen, J.; Dierkes, J.; Nygård, O.K. Dietary choline is related to increased risk of acute myocardial infarction in patients with stable angina pectoris. Biochimie 2020, 173, 68–75. [Google Scholar] [CrossRef] [Green Version]
- Gagné, M.A.; Barbeau, C.; Frégeau, G.; Gilbert, K.; Mathieu, O.; Auger, J.; Tompkins, T.A.; Charbonney, E.; Godbout, R.; Rousseau, G. Dysbiotic microbiota contributes to the extent of acute myocardial infarction in rats. Sci. Rep. 2022, 12, 16517. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Gong, Z.; Sun, G.; Xu, J.; Qi, C.; Sun, W.; Jiang, H.; Cao, P.; Ju, H. Dysbiosis of Gut Microbiota in Patients with Acute Myocardial Infarction. Front. Microbiol. 2021, 12, 680101. [Google Scholar] [CrossRef] [PubMed]
- Luedde, M.; Winkler, T.; Heinsen, F.A.; Rühlemann, M.C.; Spehlmann, M.E.; Bajrovic, A.; Lieb, W.; Franke, A.; Ott, S.J.; Frey, N. Heart failure is associated with depletion of core intestinal microbiota. ESC Heart Fail. 2017, 4, 282–290. [Google Scholar] [CrossRef]
- Kamo, T.; Akazawa, H.; Suda, W.; Saga-Kamo, A.; Shimizu, Y.; Yagi, H.; Liu, Q.; Nomura, S.; Naito, A.T.; Takeda, N.; et al. Dysbiosis and compositional alterations with aging in the gut microbiota of patients with heart failure. PLoS ONE 2017, 12, e0174099. [Google Scholar] [CrossRef] [Green Version]
- Cui, X.; Ye, L.; Li, J.; Jin, L.; Wang, W.; Li, S.; Bao, M.; Wu, S.; Li, L.; Geng, B.; et al. Metagenomic and metabolomic analyses unveil dysbiosis of gut microbiota in chronic heart failure patients. Sci. Rep. 2018, 8, 635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kummen, M.; Mayerhofer, C.C.K.; Vestad, B.; Broch, K.; Awoyemi, A.; Storm-Larsen, C.; Ueland, T.; Yndestad, A.; Hov, J.R.; Trøseid, M. Gut Microbiota Signature in Heart Failure Defined From Profiling of 2 Independent Cohorts. J. Am. Coll. Cardiol. 2018, 71, 1184–1186. [Google Scholar] [CrossRef] [PubMed]
- Pasini, E.; Aquilani, R.; Testa, C.; Baiardi, P.; Angioletti, S.; Boschi, F.; Verri, M.; Dioguardi, F. Pathogenic Gut Flora in Patients With Chronic Heart Failure. JACC Heart Fail. 2016, 4, 220–227. [Google Scholar] [CrossRef] [PubMed]
- Branchereau, M.; Burcelin, R.; Heymes, C. The gut microbiome and heart failure: A better gut for a better heart. Rev. Endocr. Metab. Disord. 2019, 20, 407–414. [Google Scholar] [CrossRef]
- McMillan, A.; Hazen, S.L. Gut Microbiota Involvement in Ventricular Remodeling Post-Myocardial Infarction. Circulation 2019, 139, 660–662. [Google Scholar] [CrossRef]
- Mayerhofer, C.C.K.; Ueland, T.; Broch, K.; Vincent, R.P.; Cross, G.F.; Dahl, C.P.; Aukrust, P.; Gullestad, L.; Hov, J.R.; Trøseid, M. Increased Secondary/Primary Bile Acid Ratio in Chronic Heart Failure. J. Card. Fail. 2017, 23, 666–671. [Google Scholar] [CrossRef] [Green Version]
- von Haehling, S.; Schefold, J.C.; Jankowska, E.A.; Springer, J.; Vazir, A.; Kalra, P.R.; Sandek, A.; Fauler, G.; Stojakovic, T.; Trauner, M.; et al. Ursodeoxycholic acid in patients with chronic heart failure: A double-blind, randomized, placebo-controlled, crossover trial. J. Am. Coll. Cardiol. 2012, 59, 585–592. [Google Scholar] [CrossRef]
- Tuerhongjiang, G.; Guo, M.; Qiao, X.; Lou, B.; Wang, C.; Wu, H.; Wu, Y.; Yuan, Z.; She, J. Interplay Between Gut Microbiota and Amino Acid Metabolism in Heart Failure. Front. Cardiovasc. Med. 2021, 8, 752241. [Google Scholar] [CrossRef]
- Hayashi, T.; Yamashita, T.; Takahashi, T.; Tabata, T.; Watanabe, H.; Gotoh, Y.; Shinohara, M.; Kami, K.; Tanaka, H.; Matsumoto, K.; et al. Role of Gut Microbiota in Amino Acid Metabolic Disturbances in Heart Failure Through Metagenomic Analysis. Front. Cardiovasc. Med. 2021, 8, 789325. [Google Scholar] [CrossRef]
- Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swanson, K.S.; de Vos, W.M.; Martens, E.C.; Gilbert, J.A.; Menon, R.S.; Soto-Vaca, A.; Hautvast, J.; Meyer, P.D.; Borewicz, K.; Vaughan, E.E.; et al. Effect of fructans, prebiotics and fibres on the human gut microbiome assessed by 16S rRNA-based approaches: A review. Benef. Microbes 2020, 11, 101–129. [Google Scholar] [CrossRef] [PubMed]
- Farhangi, M.A.; Dehghan, P.; Namazi, N. Prebiotic supplementation modulates advanced glycation end-products (AGEs), soluble receptor for AGEs (sRAGE), and cardiometabolic risk factors through improving metabolic endotoxemia: A randomized-controlled clinical trial. Eur. J. Nutr. 2020, 59, 3009–3021. [Google Scholar] [CrossRef]
- Xu, D.; Feng, M.; Chu, Y.; Wang, S.; Shete, V.; Tuohy, K.M.; Liu, F.; Zhou, X.; Kamil, A.; Pan, D.; et al. The Prebiotic Effects of Oats (avena) on Blood Lipids, Gut Microbiota, and Short-Chain Fatty Acids in Mildly Hypercholesterolemic Subjects Compared With Rice: A Randomized, Controlled Trial. Front. Immunol. 2021, 12, 787797. [Google Scholar] [CrossRef] [PubMed]
- Xu, D.; Wang, S.; Feng, M.; Shete, V.; Chu, Y.; Kamil, A.; Yang, C.; Liu, H.; Xia, H.; Wang, X.; et al. Serum Metabolomics Reveals Underlying Mechanisms of Cholesterol-Lowering Effects of Oat Consumption: A Randomized Controlled Trial in a Mildly Hypercholesterolemic Population. Mol. Nutr. Food Res. 2021, 65, e2001059. [Google Scholar] [CrossRef] [PubMed]
- Deehan, E.C.; Zhang, Z.; Riva, A.; Armet, A.M.; Perez-Muñoz, M.E.; Nguyen, N.K.; Krysa, J.A.; Seethaler, B.; Zhao, Y.Y.; Cole, J.; et al. Elucidating the role of the gut microbiota in the physiological effects of dietary fiber. Microbiome 2022, 10, 77. [Google Scholar] [CrossRef]
- Wang, Y.; Harding, S.V.; Thandapilly, S.J.; Tosh, S.M.; Jones, P.J.H.; Ames, N.P. Barley β-glucan reduces blood cholesterol levels via interrupting bile acid metabolism. Br. J. Nutr. 2017, 118, 822–829. [Google Scholar] [CrossRef] [Green Version]
- Naumann, S.; Haller, D.; Eisner, P.; Schweiggert-Weisz, U. Mechanisms of interactions between bile acids and plant compounds—A review. Int. J. Mol. Sci. 2020, 21, 6495. [Google Scholar] [CrossRef]
- 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]
- Haas, E.A.; Saad, M.J.A.; Santos, A.; Vitulo, N.; Lemos, W.J.F.; Martins, A.M.A.; Picossi, C.R.C.; Favarato, D.; Gaspar, R.S.; Magro, D.O.; et al. A red wine intervention does not modify plasma trimethylamine N-oxide but is associated with broad shifts in the plasma metabolome and gut microbiota composition. Am. J. Clin. Nutr. 2022, 116, 1515–1529. [Google Scholar] [CrossRef]
- Petersen, C.; Bharat, D.; Wankhade, U.D.; Kim, J.S.; Cutler, B.R.; Denetso, C.; Gholami, S.; Nelson, S.; Bigley, J.; Johnson, A.; et al. Dietary Blueberry Ameliorates Vascular Complicationsin Diabetic Mice Possibly through NOX4 and Modulates Composition and Functional Diversity of Gut Microbes. Mol. Nutr. Food Res. 2022, 66, e2100784. [Google Scholar] [CrossRef]
- Miller, J.C.; Babu, A.K.S.; Petersen, C.; Wankhade, U.D.; Robeson, M.S.; Putich, M.N.; Mueller, J.E.; O’Farrell, A.S.; Cho, J.M.; Chintapalli, S.V.; et al. Gut Microbes Are Associated with the Vascular Beneficial Effects of Dietary Strawberry on Metabolic Syndrome-Induced Vascular Inflammation. Mol. Nutr. Food Res. 2022, 66, e2200112. [Google Scholar] [CrossRef] [PubMed]
- Medina-Larqué, A.S.; Rodríguez-Daza, M.C.; Roquim, M.; Dudonné, S.; Pilon, G.; Levy, É.; Marette, A.; Roy, D.; Jacques, H.; Desjardins, Y. Cranberry polyphenols and agave agavins impact gut immune response and microbiota composition while improving gut barrier function, inflammation, and glucose metabolism in mice fed an obesogenic diet. Front. Immunol. 2022, 13, 871080. [Google Scholar] [CrossRef]
- Vezza, T.; Rodríguez-Nogales, A.; Algieri, F.; Garrido-Mesa, J.; Romero, M.; Sánchez, M.; Toral, M.; Martín-García, B.; Gómez-Caravaca, A.M.; Arráez-Román, D.; et al. The metabolic and vascular protective effects of olive (Olea europaea L.) leaf extract in diet-induced obesity in mice are related to the amelioration of gut microbiota dysbiosis and to its immunomodulatory properties. Pharmacol. Res. 2019, 150, 104487. [Google Scholar] [CrossRef] [PubMed]
- 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, X.; Zhao, A.; Sandhu, A.K.; Edirisinghe, I.; Burton-Freeman, B.M. Red Raspberry and Fructo-Oligosaccharide Supplementation, Metabolic Biomarkers, and the Gut Microbiota in Adults with Prediabetes: A Randomized Crossover Clinical Trial. J. Nutr. 2022, 152, 1438–1449. [Google Scholar] [CrossRef]
- Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [Green Version]
- Berry, S.; Valdes, A.; Davies, R.; Khatib, H.A.; Delahanty, L.; Drew, D.; Chan, A.T.; Segata, N.; Franks, P.; Spector, T. Large Inter-individual Variation in Postprandial Lipemia Following a Mixed Meal in over 1000 Twins and Singletons from the UK and US: The PREDICT I Study (OR19-06-19). Curr. Dev. Nutr 2019, 3 (Suppl. S1), nzz046.OR19-06-19. [Google Scholar] [CrossRef]
- 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]
- Jie, Z.; Yu, X.; Liu, Y.; Sun, L.; Chen, P.; Ding, Q.; Gao, Y.; Zhang, X.; Yu, M.; Liu, Y.; et al. The Baseline Gut Microbiota Directs Dieting-Induced Weight Loss Trajectories. Gastroenterology 2021, 160, 2029–2042.e16. [Google Scholar] [CrossRef] [PubMed]
- Mendes-Soares, H.; Raveh-Sadka, T.; Azulay, S.; Edens, K.; Ben-Shlomo, Y.; Cohen, Y.; Ofek, T.; Bachrach, D.; Stevens, J.; Colibaseanu, D.; et al. Assessment of a Personalized Approach to Predicting Postprandial Glycemic Responses to Food Among Individuals Without Diabetes. JAMA Netw. Open 2019, 2, e188102. [Google Scholar] [CrossRef] [Green Version]
- Palmnäs, M.; Brunius, C.; Shi, L.; Rostgaard-Hansen, A.; Torres, N.E.; González-Domínguez, R.; Zamora-Ros, R.; Ye, Y.L.; Halkjær, J.; Tjønneland, A.; et al. Perspective: Metabotyping-A Potential Personalized Nutrition Strategy for Precision Prevention of Cardiometabolic Disease. Adv. Nutr. 2020, 11, 524–532. [Google Scholar] [CrossRef] [Green Version]
- Leeming, E.R.; Johnson, A.J.; Spector, T.D.; Le Roy, C.I. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862. [Google Scholar] [CrossRef] [Green Version]
- Ratiner, K.; Shapiro, H.; Goldenberg, K.; Elinav, E. Time-limited diets and the gut microbiota in cardiometabolic disease. J. Diabetes 2022, 14, 377–393. [Google Scholar] [CrossRef] [PubMed]
- Maffei, S.; Forini, F.; Canale, P.; Nicolini, G.; Guiducci, L. Gut Microbiota and Sex Hormones: Crosstalking Players in Cardiometabolic and Cardiovascular Disease. Int. J. Mol. Sci. 2022, 23, 7154. [Google Scholar] [CrossRef] [PubMed]
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
Guiducci, L.; Nicolini, G.; Forini, F. Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease. Metabolites 2023, 13, 760. https://doi.org/10.3390/metabo13060760
Guiducci L, Nicolini G, Forini F. Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease. Metabolites. 2023; 13(6):760. https://doi.org/10.3390/metabo13060760
Chicago/Turabian StyleGuiducci, Letizia, Giuseppina Nicolini, and Francesca Forini. 2023. "Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease" Metabolites 13, no. 6: 760. https://doi.org/10.3390/metabo13060760
APA StyleGuiducci, L., Nicolini, G., & Forini, F. (2023). Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease. Metabolites, 13(6), 760. https://doi.org/10.3390/metabo13060760