Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation
Abstract
:1. Introduction
2. The Role of Epithelial Cellular Adhesion Molecule (Ep-CAM) and Trefoil Factor 3 (TFF3) in Inflammation and Cardiovascular Disease (CVD)
3. Obesity, Proteins Contactin 1 (CNTN1), Notch 3, and Elastin
4. Lipoproteins and Paraoxonases (PONS)
5. Gut Microbiota and Inflammation
6. Short-Chain Fatty Acids and Bile Acids
7. Trimethylamine N-oxide
8. Conclusions
Funding
Conflicts of Interest
References
- 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] [PubMed]
- Knights, D.; Silverberg, M.S.; Weersma, R.K.; Gevers, D.; Dijkstra, G.; Huang, H.; Tyler, A.D.; van Sommeren, S.; Imhann, F.; Stempak, J.M.; et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014, 6, 107. [Google Scholar] [CrossRef] [PubMed]
- Imhann, F.; Vila, A.V.; Bonder, M.J.; Fu, J.; Gevers, D.; Visschedijk, M.C.; Spekhorst, L.M.; Alberts, R.; Franke, L.; van Dullemen, H.M.; et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018, 67, 108–119. [Google Scholar] [CrossRef]
- Nakatsu, G.; Li, X.; Zhou, H.; Sheng, J.; Wong, S.H.; Wu, W.K.K.; Ng, S.C.; Tsoi, H.; Dong, Y.; Zhang, N.; et al. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat. Commun. 2015, 6, 8727. [Google Scholar] [CrossRef]
- Sender, R.; Fuchs, S.; Milo, R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef]
- Sender, R.; Fuchs, S.; Milo, R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef] [PubMed]
- Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T. Human microbiome project consortium: Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar]
- Nelson, W.C.; Weinstock, G.M.; Highlander, S.K.; Worley, K.C.; Creasy, H.H.; Wortman, J.R.; Rusch, D.B.; Mitreva, M.; Sodergren, E.; Chinwalla, A.T.; et al. The human microbiome jumpstart reference strains consortium. A catalog of reference genomes from the human microbiome. Science 2010, 328, 994–999. [Google Scholar]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
- Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; Nielsen, T.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar] [CrossRef]
- Hugon, P.; Dufour, J.-C.; Colson, P.; Fournier, P.-E.; Sallah, K.; Raoult, D. A comprehensive repertoire of prokaryotic species; identified in human beings. Lancet Infect. Dis. 2015, 15, 1211–1219. [Google Scholar] [CrossRef] [PubMed]
- Bilen, M.; Dufour, J.-C.; Lagier, J.-C.; Cadoret, F.; Daoud, Z.; Dubourg, G.; Raoult, D. The contribution of culturomics to the repertoire of isolated human bacterial and archaeal species. Microbiome 2018, 6, 94. [Google Scholar] [CrossRef] [PubMed]
- Giovannini, M.G.; Lana, D.; Traini, C.; Vannucchi, M.G. The microbiota–gut–brain axis and Alzheimer disease. From dysbiosis to neurodegeneration: Focus on the central nervous system glial cells. J. Clin. Med. 2021, 10, 2358. [Google Scholar] [CrossRef]
- Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef]
- Bauer, T.M.; Schwacha, H.; Steinbrückner, B.; E Brinkmann, F.; Ditzen, A.K.; Kist, M.; E Blum, F. Diagnosis of small intestinal bacterial overgrowth in patients with cirrhosis of the liver: Poor performance of the glucose breath hydrogen test. J. Hepatol. 2000, 33, 382–386. [Google Scholar] [CrossRef]
- Simren, M.; Stotzer, P.O. Use and abuse of hydrogen breath tests. Gut 2006, 55, 297–303. [Google Scholar] [CrossRef]
- Van De Wouw, M.; Boehme, M.; Lyte, J.M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain-gut axis alterations. J. Physiol. 2018, 596, 4923–4944. [Google Scholar] [CrossRef] [PubMed]
- Agus, A.; Planchais, J.; Sokol, H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
- MahmoudianDehkordi, S.; Arnold, M.; Nho, K.; Ahmad, S.; Jia, W.; Xie, G.; Louie, G.; Kueider-Paisley, A.; Moseley, M.A.; Thompson, J.W.; et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease—An emerging role for gut microbiome. Alzheimer’s Dement 2019, 15, 76–92. [Google Scholar] [CrossRef]
- Baj, A.; Moro, E.; Bistoletti, M.; Orlandi, V.; Crema, F.; Giaroni, C. Glutamatergic signaling along the microbiota-gut-brain axis. Int. J. Mol. Sci. 2019, 20, 1482. [Google Scholar] [CrossRef] [PubMed]
- Tracey, K.J.; Chavan, S.S. Nerve Stimulation for Treatment of Diseases and Disorders. International Patent PCT/US2016/018573, 19 February 2016. [Google Scholar]
- Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
- Zhernakova, D.V.; Le, T.H.; Kurilshikov, A.; Atanasovska, B.; Bonder, M.J.; Sanna, S.; Claringbould, A.; Võsa, U.; Deelen, P.; Franke, L.; et al. Individual variations in cardiovascular disease-related protein levels are driven by genetics and gut microbiome. Nat. Genet. 2018, 50, 1524–1532. [Google Scholar] [CrossRef]
- Bjorkqvist, J.; Jamsa, A.; Renne, T. Plasma kallikrein: The bradykinin-producing enzyme. Thromb. Haemost. 2013, 110, 399–407. [Google Scholar] [CrossRef] [PubMed]
- Stavrou, E.; Schmaier, A.H. Factor XII: What does it contribute to our understanding of the physiology and pathophysiology of hemostasis & thrombosis. Thromb. Res. 2010, 125, 210–215. [Google Scholar]
- Hofman, Z.; de Maat, S.; Hack, C.E.; Maas, C. Product of the coagulation system. Clin. Rev. Allergy Immunol. 2016, 51, 152–161. [Google Scholar] [CrossRef] [PubMed]
- Kozan, P.A.; McGeough, M.D.; Peña, C.A.; Mueller, J.L.; Barrett, K.E.; Marchelletta, R.R.; Sivagnanam, M. Mutation of EpCAM leads to intestinal barrier and ion transport dysfunction. J. Mol. Med. 2015, 93, 535–545. [Google Scholar] [CrossRef]
- Liu, X.; Mao, B.; Gu, J.; Wu, J.; Cui, S.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Blautia—A new functional genus with potential probiotic properties? Gut Microbes 2021, 13, 1875796. [Google Scholar] [CrossRef]
- Yang, Y.; Lin, Z.; Lin, Q.; Bei, W.; Guo, J. Pathological and therapeutic roles of bioactive peptide trefoil factor 3 in diverse diseases: Recent progress and perspective. Cell Death Dis. 2022, 13, 62. [Google Scholar] [CrossRef]
- Trim, W.; Turner, J.E.; Thompson, D. Parallels in Immunometabolic adipose tissue dysfunction with ageing and obesity. Front. Immunol. 2018, 9, 169. [Google Scholar] [CrossRef]
- Lau, E.S.; Paniagua, S.M.; Zarbafian, S.; Hoffman, U.; Long, M.T.; Hwang, S.; Courchesne, P.; Yao, C.; Ma, J.; Larson, M.G.; et al. Cardiovascular biomarkers of obesity and overlap with cardiometabolic dysfunction. J. Am. Heart. Assoc. 2021, 10, e020215. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Lv, T.; Wang, X.; Wu, M.; Zhang, R.; Yang, X.; Fu, Y.; Liu, Z. Role of the microbiota–gut–heart axis between bile acids and cardiovascular disease. Biomed. Pharmacother. 2024, 174, 116567. [Google Scholar] [CrossRef] [PubMed]
- Dicks, L.M.T. Gut bacteria and neurotransmitters. Microorganisms 2022, 10, 1838. [Google Scholar] [CrossRef]
- Dicks, L.M.T. How does quorum sensing of intestinal bacteria affect our health and mental status? Microorganisms 2022, 10, 1969. [Google Scholar] [CrossRef]
- Yin, X.; Subramanian, S.; Hwang, S.-J.; O’Donnell, C.J.; Fox, C.S.; Courchesne, P.; Muntendam, P.; Gordon, N.; Adourian, A.; Juhasz, P.; et al. Protein biomarkers of new-onset cardiovascular disease: Prospective study from the systems approach to biomarker research in cardiovascular disease initiative. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 939–945. [Google Scholar] [CrossRef]
- Malka, K.; Liaw, L. NOTCH3 as a modulator of vascular disease: A target in elastin deficiency and arterial pathologies. J. Clin. Investig. 2022, 132, e157007. [Google Scholar] [CrossRef]
- Bray, S.J. Notch Signalling: A simple pathway becomes complex. Nat. Rev. Mol. Cell Biol 2006, 7, 678–689. [Google Scholar] [CrossRef]
- Rascher-Eggstein, G.; Liebner, S.; Wolburg, H. The Blood–Brain Barrier in the Human Glioma. In Blood-Spinal Cord and Brain Barriers in Health and Disease; Sharma, H.S., Westman, J., Eds.; Academic Press: Academic Press, San Diego, 2004; pp. 561–576. ISBN 9780126390117. [Google Scholar]
- Merla, G.; Brunetti-Pierri, N.; Piccolo, P.; Micale, L.; Loviglio, M.N. Supravalvular aortic stenosis: Elastin arteriopathy. Circ. Cardiovasc. Genet. 2012, 5, 692–696. [Google Scholar] [CrossRef] [PubMed]
- Cocciolone, A.J.; Hawes, J.Z.; Staiculescu, M.C.; Johnson, E.O.; Murshed, M.; Wagenseil, J.E. Elastin, arterial mechanics, and cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H189–H205. [Google Scholar] [CrossRef]
- Durrington, P.N.; Mackness, B.; Mackness, M.I. Paraoxonase and atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 473–480. [Google Scholar] [CrossRef]
- Khattib, A.; Musa, S.; Halabi, M.; Hayek, T.; Khatib, S. Lyso-DGTS. Lipid derivatives enhance PON1 activities and prevent oxidation of LDL: A structure-activity relationship study. Antioxidants 2022, 11, 2058. [Google Scholar] [CrossRef] [PubMed]
- Ng, C.J.; Shih, D.M.; Hama, S.Y.; Villa, N.; Navab, M.; Reddy, S.T. The paraoxonase gene family and atherosclerosis. Free. Radic. Biol. Med. 2005, 38, 153–163. [Google Scholar] [CrossRef] [PubMed]
- Billecke, S.; Draganov, D.; Counsell, R.; Stetson, P.; Watson, C.; Hsu, C.; La Du, B.N. Human serum paraoxonase (PON1) isozymes Q and R hydrolyze lactones and cyclic carbonate esters. Drug Metab. Dispos. 2000, 28, 1335–1342. [Google Scholar] [PubMed]
- Draganov, D.I.; Teiber, J.F.; Speelman, A.; Osawa, Y.; Sunahara, R.; La Du, B.N. Human paraoxonases (PON1, PON2, and PON3) are lactonases with overlapping and distinct substrate specificities. J. Lip. Res. 2005, 46, 1239–1247. [Google Scholar] [CrossRef]
- Devarajan, A.; Bourquard, N.; Hama, S.; Navab, M.; Grijalva, V.R.; Morvardi, S.; Clarke, C.F.; Vergnes, L.; Reue, K.; Teiber, J.F.; et al. Paraoxonase 2 deficiency alters mitochondrial function and exacerbates the development of atherosclerosis. Antioxid. Redox. Signal. 2011, 14, 341–351. [Google Scholar] [CrossRef]
- Priyanka, K.; Singh, S.; Gill, K. Paraoxonase 3: Structure and its role in pathophysiology of coronary artery disease. Biomolecules 2019, 9, 817. [Google Scholar] [CrossRef]
- Wang, L.; Wang, S.; Zhang, Q.; He, C.; Fu, C.; Wei, Q. The role of the gut microbiota in health and cardiovascular diseases. Mol. Biomed. 2022, 3, 30. [Google Scholar] [CrossRef]
- Trøseid, M.; Nestvold, T.K.; Rudi, K.; Thoresen, H.; Nielsen, E.W.; Lappegård, K.T. Plasma lipopolysaccharide is closely associated with glycemic control and abdominal obesity. Evidence from bariatric surgery. Diabetes Care 2013, 36, 3627–3632. [Google Scholar] [CrossRef]
- Brandsma, E.; Kloosterhuis, N.J.; Koster, M.; Dekker, D.C.; Gijbels, M.J.; van der Velden, S.; Ríos-Morales, M.; van Faassen, M.J.; Loreti, M.G.; de Bruin, A.; et al. A proinflammatory gut microbiota increases systemic inflammation and accelerates atherosclerosis. Circ. Res. 2019, 124, 94–100. [Google Scholar] [CrossRef]
- An, H.; Qian, C.; Cao, X. Regulation of Toll-like receptor signaling in the innate immunity. Sci. China Life Sci. 2010, 53, 34–43. [Google Scholar] [CrossRef]
- Kienes, I.; Weidl, T.; Mirza, N.; Chamaillard, M.; Kufer, T.A. Role of NLRs in the regulation of type I interferon signaling, host defense and tolerance to inflammation. Int. J. Mol. Sci. 2021, 22, 1301. [Google Scholar] [CrossRef] [PubMed]
- Jaén, R.I.; Val-Blasco, A.; Prieto, P.; Gil-Fernández, M.; Smani, T.; López-Sendón, J.L.; Delgado, C.; Boscá, L.; Fernández-Velasco, M. Innate immune receptors, key actors in cardiovascular diseases. JACC Basic Transl. Sci. 2020, 5, 735–749. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.-Q.; Zhang, X.-Y.; Edfeldt, K.; Nijhuis, M.O.; Idborg, H.; Bäck, M.; Roy, J.; Hedin, U.; Jakobsson, P.-J.; Laman, J.D.; et al. NOD2-mediated innate immune signaling regulates the eicosanoids in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2193–2201. [Google Scholar] [CrossRef] [PubMed]
- Moreno, L.; Gatheral, T. Therapeutic targeting of NOD1 receptors. Br. J. Pharmacol. 2013, 170, 475–485. [Google Scholar] [CrossRef] [PubMed]
- Grylls, A.; Seidler, K.; Neil, J. Link between microbiota and hypertension: Focus on LPS/TLR4 pathway in endothelial dysfunction and vascular inflammation, and therapeutic implication of probiotics. Biomed. Pharmacother. 2021, 137, 111334. [Google Scholar] [CrossRef]
- Zhi, C.; Huang, J.; Wang, J.; Cao, H.; Bai, Y.; Guo, J.; Su, Z. Connection between gut microbiome and the development of obesity. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 1987–1998. [Google Scholar] [CrossRef]
- Subah, P.C. Estrogen protection, oxidized LDL, endothelial dysfunction and vasorelaxation in cardiovascular disease: New insights into a complex issue. Cardiovasc. Res. 2007, 73, 6–7. [Google Scholar]
- Rath, E.; Moschetta, A.; Haller, D. Mitochondrial function—Gatekeeper of intestinal epithelial cell homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2018, 158, 497–516. [Google Scholar] [CrossRef]
- Tang, W.W.; Wang, Z.; Shrestha, K.; Borowski, A.G.; Wu, Y.; Troughton, R.W.; Klein, A.L.; Hazen, S.L. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J. Card. Fail. 2015, 21, 91–96. [Google Scholar] [CrossRef]
- Campbell, E.L.; Colgan, S.P. Control and dysregulation of redox signalling in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 2018, 16, 106–120. [Google Scholar] [CrossRef]
- Kossmann, S.; Schwenk, M.; Hausding, M.; Karbach, S.H.; Schmidgen, M.I.; Brandt, M.; Knorr, M.; Hu, H.; Kröller-Schön, S.; Schönfelder, T.; et al. Angiotensin Ii-induced vascular dysfunction depends on interferon-g- driven immune cell recruitment and mutual activation of monocytes and Nk-cells. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 1313–1319. [Google Scholar] [CrossRef] [PubMed]
- Karbach, S.H.; Schönfelder, T.; Brandão, I.; Wilms, E.; Hörmann, N.; Jäckel, S.; Schüler, R.; Finger, S.; Knorr, M.; Lagrange, J.; et al. Gut Microbiota Promote Angiotensin II-Induced Arterial Hypertension and Vascular Dysfunction. J. Am. Heart. Assoc. 2016, 5, e003698. [Google Scholar] [CrossRef] [PubMed]
- Parada Venegas, D.; De La Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front. Immunol. 2019, 10, 277. [Google Scholar]
- Cushing, K.; Alvarado, D.M.; Ciorba, M.A. Butyrate and mucosal inflammation: New scientific evidence supports clinical observation. Clin. Transl. Gastroenterol. 2015, 6, e108. [Google Scholar] [CrossRef]
- 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]
- De Vadder, F.; Grasset, E.; Holm, L.M.; Karsenty, G.; MacPherson, A.J.; Olofsson, L.E.; Bäckhed, F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc. Natl. Acad. Sci. USA 2018, 115, 6458–6463. [Google Scholar] [CrossRef]
- Egerod, K.L.; Petersen, N.; Timshel, P.N.; Rekling, J.C.; Wang, Y.; Liu, Q.; Schwartz, T.W.; Gautron, L. Profiling of G protein-coupled receptors in vagal afferents reveals novel gut-to-brain sensing mechanisms. Mol. Metab. 2018, 12, 62–75. [Google Scholar] [CrossRef]
- 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]
- Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015, 39, 424–429. [Google Scholar] [CrossRef]
- Zhou, J.; Martin, R.J.; Tulley, R.T.; Raggio, A.M.; McCutcheon, K.L.; Shen, L.; Danna, S.C.; Tripathy, S.; Hegsted, M.; Keenan, M.J. Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E1160–E1166. [Google Scholar] [CrossRef]
- Liu, T.; Li, J.; Liu, Y.; Xiao, N.; Suo, H.; Xie, K.; Yang, C.; Wu, C. Short-chain fatty acids suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory cytokines through inhibition of NF-κB pathway in RAW264.7 cells. Inflammation 2012, 35, 1676–1684. [Google Scholar] [CrossRef] [PubMed]
- Takeuchi, O.; Akira, S. Pattern Recognition Receptors and Inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [PubMed]
- Cheng, D.; Xu, J.-H.; Li, J.-Y.; Wang, S.-Y.; Wu, T.-F.; Chen, Q.-K.; Yu, T. Butyrate Ameliorated-NLRC3 Protects the Intestinal Barrier in a GPR43-Dependent Manner. Exp. Cell Res. 2018, 368, 101–110. [Google Scholar] [CrossRef]
- Tong, L.-C.; Wang, Y.; Wang, Z.-B.; Liu, W.-Y.; Sun, S.; Li, L.; Su, D.-F.; Zhang, L.-C. Propionate Ameliorates Dextran Sodium Sulfate-Induced Colitis by Improving Intestinal Barrier Function and Reducing Inflammation and Oxidative Stress. Front. Pharmacol. 2016, 7, 253. [Google Scholar] [CrossRef]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; Van Der Veeken, J.; DeRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
- Campos-Perez, W.; Martinez-Lopez, E. Effects of short chain fatty acids on metabolic and inflammatory processes in human health. Biochim Biophys Acta Mol Cell Biol Lipids 2021, 1866, 158900. [Google Scholar] [CrossRef] [PubMed]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly-Y, M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, shortchain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef]
- Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
- Tan, J.; McKenzie, C.; Vuillermin, P.J.; Goverse, G.; Vinuesa, C.G.; Mebius, R.E.; Macia, L.; Mackay, C.R. Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep. 2016, 15, 2809–2824. [Google Scholar] [CrossRef]
- Wu, W.; Sun, M.; Chen, F.; Cao, A.; Liu, H.; Zhao, Y.; Huang, X.; Xiao, Y.; Yao, S.; Zhao, Q.; et al. Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal. Immunol. 2017, 10, 946–956. [Google Scholar] [CrossRef]
- Sun, M.; Wu, W.; Chen, L.; Yang, W.; Huang, X.; Ma, C.; Chen, F.; Xiao, Y.; Zhao, Y.; Ma, C.; et al. Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat. Commun. 2018, 9, 3555. [Google Scholar] [CrossRef] [PubMed]
- Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef]
- Armet, A.M.; Deehan, E.C.; Thöne, J.V.; Hewko, S.J.; Walter, J. The effect of isolated and synthetic dietary fibers on markers of metabolic diseases in human intervention studies: A systematic review. Adv. Nutr. 2020, 11, 420–438. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Zhang, F.; Ding, X.; Wu, G.; Lam, Y.Y.; Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.; et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018, 359, 1151–1156. [Google Scholar] [CrossRef] [PubMed]
- Desai, M.S.; Seekatz, A.M.; Koropatkin, N.M.; Kamada, N.; Hickey, C.A.; Wolter, M.; Pudlo, N.A.; Kitamoto, S.; Terrapon, N.; Muller, A.; et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 2016, 167, 1339–1353.e21. [Google Scholar] [CrossRef]
- Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.-I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. New Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef]
- Vangay, P.; Johnson, A.J.; Ward, T.L.; Al-Ghalith, G.A.; Shields-Cutler, R.R.; Hillmann, B.M.; Lucas, S.K.; Beura, L.K.; Thompson, E.A.; Till, L.M.; et al. US immigration westernizes the human gut microbiome. Cell 2018, 175, 962–972. [Google Scholar] [CrossRef]
- Sonnenburg, E.D.; Smits, S.A.; Tikhonov, M.; Higginbottom, S.K.; Wingreen, N.S.; Sonnenburg, J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016, 529, 212–215. [Google Scholar] [CrossRef] [PubMed]
- Wastyk, H.C.; Fragiadakis, G.K.; Perelman, D.; Dahan, D.; Merrill, B.D.; Yu, F.B.; Topf, M.; Gonzalez, C.G.; Van Treuren, W.; Han, S.; et al. Gut-microbiota-targeted diets modulate human immune status. Cell 2021, 184, 4137–4153.e14. [Google Scholar] [CrossRef]
- Johnson, A.J.; Vangay, P.; Al-Ghalith, G.A.; Hillmann, B.M.; Ward, T.L.; Shields-Cutler, R.R.; Kim, A.D.; Shmagel, A.K.; Syed, A.N.; Walter, J.; et al. Daily sampling reveals personalized diet-microbiome associations in humans. Cell Host Microbe 2019, 25, 789–802. [Google Scholar] [CrossRef]
- Roshanravan, N.; Mahdavi, R.; Alizadeh, E.; Jafarabadi, M.A.; Hedayati, M.; Ghavami, A.; Alipour, S.; Alamdari, N.M.; Barati, M.; Ostadrahimi, A. Effect of butyrate and inulin supplementation on glycemic status, lipid profile and glucagon-like peptide 1 level in patients with type 2 diabetes: A randomized double-blind, placebo-controlled trial. Horm. Metab. Res. 2017, 49, 886–891. [Google Scholar] [CrossRef] [PubMed]
- Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Nitert, M.D. 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]
- Isibor, P.; Akinduti, P.; Aworunse, O.; Oyewale, J.; Oshamika, O.; Ugboko, H.; Taiwo, O.; Ahuekwe, E.; Obafemi, Y.; Onibokun, E.; et al. Significance of African diets in biotherapeutic modulation of the gut microbiome. Bioinform. Biol. Insights 2021, 15, 11779322211012697. [Google Scholar] [CrossRef]
- Statovci, D.; Aguilera, M.; MacSharry, J.; Melgar, S. The impact of Western diet and nutrients on the microbiota and immune response at mucosal interfaces. Front. Immunol. 2017, 8, 838. [Google Scholar] [CrossRef]
- Wrzosek, L.; Miquel, S.; Noordine, M.-L.; Bouet, S.; Chevalier-Curt, M.J.; Robert, V.; Philippe, C.; Bridonneau, C.; Cherbuy, C.; Robbe-Masselot, C.; et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. 2013, 11, 61. [Google Scholar] [CrossRef]
- So, D.; Whelan, K.; Rossi, M.; Morrison, M.; Holtmann, G.; Kelly, J.T.; Shanahan, E.R.; Staudacher, H.M.; Campbell, K.L. Dietary fiber intervention on gut microbiota composition in healthy adults: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2018, 107, 965–983. [Google Scholar] [CrossRef]
- Dicks, L.M.T.; Grobbelaar, M.J. Double-barrel shotgun: Probiotic lactic acid bacteria with antiviral properties modified to serve as vaccines. Microorganisms 2021, 9, 1565. [Google Scholar] [CrossRef]
- Dicks, L.M.T.; Geldenhuys, J.; Mikkelsen, L.S.; Brandsborg, E.; Marcotte, H. Our gut microbiota: A long walk to homeostasis. Benef. Microbes 2018, 9, 3–19. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Long, W.; Zhang, C.; Liu, S.; Zhao, L.; Hamaker, B.R. Fiber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota. Sci. Rep. 2017, 7, 2594. [Google Scholar] [CrossRef]
- McDonald, D.; Hyde, E.; Debelius, J.W.; Morton, J.T.; Gonzalez, A.; Ackermann, G.; Aksenov, A.A.; Behsaz, B.; Brennan, C.; Chen, Y.; et al. American gut: An open platform for citizen science microbiome research. mSystems 2018, 3, e00031-18. [Google Scholar] [CrossRef]
- Blaak, E.E.; Canfora, E.E.; Theis, S.; Frost, G.; Groen, A.K.; Mithieux, G.; Nauta, A.; Scott, K.; Stahl, B.; Van Harsselaar, J.; et al. Short chain fatty acids in human gut and metabolic health. Benef. Microbes 2020, 11, 411–455. [Google Scholar] [CrossRef] [PubMed]
- Diaz, M.; Kellingray, L.; Akinyemi, N.; Adefiranye, O.O.; Olaonipekun, A.B.; Bayili, G.R.; Ibezim, J.; du Plessis, A.S.; Houngbédji, M.; Kamya, D.; et al. Comparison of the microbial composition of African fermented foods using amplicon sequencing. Sci. Rep. 2019, 9, 13863. [Google Scholar] [CrossRef] [PubMed]
- Heiman, M.L.; Greenway, F.L. A healthy gastrointestinal microbiome is dependent on dietary diversity. Mol. Metab. 2016, 5, 317–320. [Google Scholar] [CrossRef] [PubMed]
- Guizoni, D.M.; Vettorazzi, J.F.; Carneiro, E.M.; Davel, A.P. Modulation of endothelium-derived nitric oxide production and activity by taurine and taurine-conjugated bile acids. Nitric Oxide 2020, 94, 48–53. [Google Scholar] [CrossRef]
- Callender, C.; Attaye, I.; Nieuwdorp, M. The interaction between the gut microbiome and bile acids in cardiometabolic diseases. Metabolites 2022, 12, 65. [Google Scholar] [CrossRef]
- Pols, T.W.; Nomura, M.; Harach, T.; Sasso, G.L.; Oosterveer, M.H.; Thomas, C.; Rizzo, G.; Gioiello, A.; Adorini, L.; Pellicciari, R.; et al. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab. 2011, 14, 747–757. [Google Scholar] [CrossRef]
- Pols, T.W. TGR5 in inflammation and cardiovascular disease. Biochem. Soc. Trans. 2014, 42, 244–249. [Google Scholar] [CrossRef]
- Baghdasaryan, A.; Claudel, T.; Gumhold, J.; Silbert, D.; Adorini, L.; Roda, A.; Vecchiotti, S.; Gonzalez, F.J.; Schoonjans, K.; Strazzabosco, M.; et al. Dual farnesoid X receptor/TGR5 agonist INT-767 reduces liver injury in the Mdr2-/- (Abcb4-/-) mouse cholangiopathy model by promoting biliary HCO⁻₃ output. Hepatology 2011, 54, 1303–1312. [Google Scholar] [CrossRef]
- Kaplan, J.; Kanwal, A.; Ahmed, I.; Lala, V. Reentrant Arrhythmias. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK557775/ (accessed on 2 August 2024).
- 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]
- 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]
- Katsimichas, T.; Antonopoulos, A.S.; Katsimichas, A.; Ohtani, T.; Sakata, Y.; Tousoulis, D. The intestinal microbiota and cardiovascular disease. Cardiovasc. Res. 2019, 115, 1471–1486. [Google Scholar] [CrossRef] [PubMed]
- Xie, G.; Yan, A.; Lin, P.; Wang, Y.; Guo, L. Trimethylamine N-oxide-a marker for atherosclerotic vascular disease. Rev. Cardiovasc. Med. 2021, 22, 787–797. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.L.; Zhu, X.H.; Ran, L.; Lang, H.D.; Yi, L.; Mi, M.T. Trimethylamine-N-oxide induces vascular inflammation by activating the NLRP3 inflammasome through the sirt3-sod2-mtros signaling pathway. J. Am. Heart Assoc. 2017, 6, e003698. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Ding, L.; Chang, M.; Guo, Y.; Zhang, L.; Xue, C.; Yanagita, T.; Zhang, T.; Wang, Y. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids Health Dis. 2018, 17, 286. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, L.; Ren, P.; Yang, Y.; Li, S.; Qin, X.; Zhang, M.; Zhou, M.; Liu, W. Qing-xue-xiao-zhi formula attenuates atherosclerosis by inhibiting macrophage lipid accumulation and inflammatory response via TLR4/myd88/NF-κB pathway regulation. Phytomedicine 2021, 93, 153812. [Google Scholar] [CrossRef]
- Liu, X.; Shao, Y.; Tu, J.; Sun, J.; Li, L.; Tao, J.; Chen, J. Trimethylamine-N-oxide stimulated hepatocyte-derived exosomes promote inflammation and endothelial dysfunction through nuclear factor-kappa b signaling. Ann. Transl. Med. 2021, 9, 1670. [Google Scholar] [CrossRef]
- Schuett, K.; Kleber, M.E.; Scharnagl, H.; Lorkowski, S.; März, W.; Niessner, A.; Marx, N.; Meinitzer, A. Trimethylamine-N-oxide and heart failure with reduced versus preserved ejection fraction. J. Am. Coll. Cardiol. 2017, 70, 3202–3204. [Google Scholar] [CrossRef]
- Akerman, B.; Lemass, H.; Chow, L.; Lambert, D.; Greenberg, C.; Bibeau, C.; Mamer, O.; Treacy, E. Trimethylaminuria is caused by mutations of the FMO3 gene in a North American cohort. Mol. Genet. Metab. 1999, 68, 24–31. [Google Scholar] [CrossRef]
- Cashman, J.R.; Bi, Y.-A.; Lin, J.; Youil, R.; Knight, M.; Forrest, S.; Treacy, E. Human flavin-containing monooxygenase form 3: cDNA expression of the enzymes containing amino acid substitutions observed in individuals with trimethylaminuria. Chem. Res. Toxicol. 1997, 10, 837–841. [Google Scholar] [CrossRef]
- Treacy, E.P.; Akerman, B.R.; Chow, L.M.L.; Youil, R.; Lin, C.B.; Bruce, A.G.; Knight, M.; Danks, D.M.; Cashman, J.R.; Forrest, S.M. Mutations of the flavin-containing monooxygenase gene (FMO3) cause trimethylaminuria, a defect in detoxication. Hum. Mol. Genet. 1998, 7, 839–845. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Zuo, K.; Liu, X.; Wang, P.; Jiao, J.; Han, C.; Liu, Z.; Yin, X.; Li, J.; Yang, X. Metagenomic data mining reveals enrichment of trimethylamine-N-oxide synthesis in gut microbiome in atrial fibrillation patients. BMC Genom. 2020, 21, 526. [Google Scholar] [CrossRef] [PubMed]
- Yu, L.; Meng, G.; Huang, B.; Zhou, X.; Stavrakis, S.; Wang, M.; Li, X.; Zhou, L.; Wang, Y.; Wang, M.; et al. A potential relationship between gut microbes and atrial fibrillation: Trimethylamine N-oxide, a gut microbe-derived metabolite, facilitates the progression of atrial fibrillation. Int. J. Cardiol. 2018, 255, 92–98. [Google Scholar] [CrossRef] [PubMed]
- Hou, Y.; Scherlag, B.J.; Lin, J.; Zhang, Y.; Lu, Z.; Truong, K.; Patterson, E.; Lazzara, R.; Jackman, W.M.; Po, S.S. Ganglionated plexi modulate extrinsic cardiac autonomic nerve input: Effects on sinus rate, atrioventricular conduction, refractoriness, and inducibility of atrial fibrillation. J. Am. Coll. Cardiol. 2007, 50, 61–68. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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. [Google Scholar] [CrossRef]
- Chen, Y.-Y.; Sun, Z.-W.; Jiang, J.-P.; Kang, X.-D.; Wang, L.-L.; Shen, Y.-L.; Xie, X.-D.; Zheng, L.-R. A-adrenoceptor-mediated enhanced inducibility of atrial fibrillation in a canine system inflammation model. Mol. Med. Rep. 2017, 15, 3767–3774. [Google Scholar] [CrossRef]
- Zuo, K.; Li, J.; Li, K.; Hu, C.; Gao, Y.; Chen, M.; Hu, R.; Liu, Y.; Chi, H.; Wang, H.; et al. Disordered gut microbiota and alterations in metabolic patterns are associated with atrial fibrillation. Gigascience 2019, 8, giz058. [Google Scholar] [CrossRef]
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. |
© 2024 by the author. 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
Dicks, L.M.T. Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation. Int. J. Mol. Sci. 2024, 25, 10634. https://doi.org/10.3390/ijms251910634
Dicks LMT. Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation. International Journal of Molecular Sciences. 2024; 25(19):10634. https://doi.org/10.3390/ijms251910634
Chicago/Turabian StyleDicks, Leon M. T. 2024. "Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation" International Journal of Molecular Sciences 25, no. 19: 10634. https://doi.org/10.3390/ijms251910634