236
Views
3
CrossRef citations to date
0
Altmetric
REVIEW

Intestinal Flora Derived Metabolites Affect the Occurrence and Development of Cardiovascular Disease

, , , , , , & show all
Pages 2591-2603 | Received 23 Mar 2022, Accepted 10 Aug 2022, Published online: 09 Nov 2022

References

  • Nitsa A, Toutouza M, Machairas N, Mariolis A, Philippou A, Koutsilieris M. Vitamin D in cardiovascular disease. In Vivo (Brooklyn). 2018;32(5):977–981. doi:10.21873/invivo.11338
  • Roth GA, Johnson C, Abajobir A, et al. Global, regional, and National Burden of Cardiovascular Diseases for 10 causes, 1990 to 2015. J Am Coll Cardiol. 2017;70(1):1–25. doi:10.1016/j.jacc.2017.04.052
  • Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar Reddy D. Role of the normal gut microbiota. World J Gastroenterol. 2015;21(29):8787–8803. doi:10.3748/wjg.v21.i29.8787
  • Heintz-Buschart A, Wilmes P. Human gut microbiome: function matters. Trends Microbiol. 2018;26(7):563–574. doi:10.1016/j.tim.2017.11.002
  • Wampach L, Heintz-Buschart A, Hogan A, et al. Colonization and succession within the human gut microbiome by archaea, bacteria, and microeukaryotes during the first year of life. Front Microbiol. 2017;8:738. doi:10.3389/fmicb.2017.00738
  • Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005;308(5728):1635–1638. doi:10.1126/science.1110591
  • Demarquoy J, Georges B, Rigault C, et al. Radioisotopic determination of l-carnitine content in foods commonly eaten in Western countries. Food Chem. 2004;86(1):137–142. doi:10.1016/j.foodchem.2003.09.023
  • Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164(3):337–340. doi:10.1016/j.cell.2016.01.013
  • Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. doi:10.1038/nature08821
  • Lim ES, Zhou Y, Zhao G, et al. Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med. 2015;21(10):1228–1234. doi:10.1038/nm.3950
  • Martinez-Guryn K, Leone V, Chang EB. Regional diversity of the gastrointestinal microbiome. Cell Host Microbe. 2019;26(3):314–324. doi:10.1016/j.chom.2019.08.011
  • Robles-Vera I, Toral M, de la Visitación N, et al. Changes to the gut microbiota induced by losartan contributes to its antihypertensive effects. Br J Pharmacol. 2020;177(9):2006–2023. doi:10.1111/bph.14965
  • Robles-Vera I, Toral M, Romero M, et al. Antihypertensive effects of probiotics. Curr Hypertens Rep. 2017;19(4):26. doi:10.1007/s11906-017-0723-4
  • Yang T, Santisteban MM, Rodriguez V, et al. Gut dysbiosis is linked to hypertension. Hypertension. 2015;65(6):1331–1340. doi:10.1161/HYPERTENSIONAHA.115.05315
  • Hoyles L, Jiménez-Pranteda ML, Chilloux J, et al. Metabolic retroconversion of trimethylamine N-oxide and the gut microbiota. Microbiome. 2018;6(1):73. doi:10.1186/s40168-018-0461-0
  • Ufnal M, Jazwiec R, Dadlez M, Drapala A, Sikora M, Skrzypecki J. Trimethylamine-N-oxide: a carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin II in rats. Can J Cardiol. 2014;30(12):1700–1705. doi:10.1016/j.cjca.2014.09.010
  • D’Odorico I, Di Bella S, Monticelli J, Giacobbe DR, Boldock E, Luzzati R. Role of fecal microbiota transplantation in inflammatory bowel disease. J Dig Dis. 2018;19(6):322–334. doi:10.1111/1751-2980.12603
  • Gallo A, Passaro G, Gasbarrini A, Landolfi R, Montalto M. Modulation of microbiota as treatment for intestinal inflammatory disorders: an uptodate. World J Gastroenterol. 2016;22(32):7186–7202. doi:10.3748/wjg.v22.i32.7186
  • Brown JM, Hazen SL. The gut microbial endocrine organ: bacterially derived signals driving cardiometabolic diseases. Annu Rev Med. 2015;66:343–359. doi:10.1146/annurev-med-060513-093205
  • Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63. doi:10.1038/nature09922
  • Yan G, Wang J, Yi T, et al. Baicalin prevents pulmonary arterial remodeling in vivo via the AKT/ERK/NF-κB signaling pathways. Pulm Circ. 2019;9(4):2045894019878599. doi:10.1177/2045894019878599
  • Seldin MM, Meng Y, Qi H, et al. Trimethylamine N-Oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear Factor-κB. J Am Heart Assoc. 2016;5(2). doi:10.1161/JAHA.115.002767
  • Yang G, Lin CC, Yang Y, et al. Nobiletin prevents trimethylamine oxide-induced vascular inflammation via inhibition of the NF-κB/MAPK pathways. J Agric Food Chem. 2019;67(22):6169–6176. doi:10.1021/acs.jafc.9b01270
  • Wang X, Wang Y, Antony V, Sun H, Liang G. Metabolism-Associated Molecular Patterns (MAMPs). Trends Endocrinol Metab. 2020;31(10):712–724. doi:10.1016/j.tem.2020.07.001
  • Wu Q, Zhang X, Zhao Y, Yang X. High l-Carnitine ingestion impairs liver function by disordering gut bacteria composition in mice. J Agric Food Chem. 2020;68(20):5707–5714. doi:10.1021/acs.jafc.9b08313
  • Guignabert C, Humbert M. Targeting transforming growth factor-β receptors in pulmonary hypertension. Eur Respir J. 2021;57(2):2002341. doi:10.1183/13993003.02341-2020
  • Tang WH, Wang Z, Kennedy DJ, et al. 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(3):448–455. doi:10.1161/CIRCRESAHA.116.305360
  • El-Deeb OS, Atef MM, Hafez YM. The interplay between microbiota-dependent metabolite trimethylamine N-oxide, Transforming growth factor β/SMAD signaling and inflammasome activation in chronic kidney disease patients: a new mechanistic perspective. J Cell Biochem. 2019;120(9):14476–14485. doi:10.1002/jcb.28707
  • Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med. 2020;217(3):e20190103. doi:10.1084/jem.20190103
  • Kojonazarov B, Novoyatleva T, Boehm M, et al. p38 MAPK inhibition improves heart function in pressure-loaded right ventricular hypertrophy. Am J Respir Cell Mol Biol. 2017;57(5):603–614. doi:10.1165/rcmb.2016-0374OC
  • Chowdhury A, Sarkar J, Pramanik PK, Chakraborti T, Chakraborti S. Cross talk between MMP2-Spm-Cer-S1P and ERK1/2 in proliferation of pulmonary artery smooth muscle cells under angiotensin II stimulation. Arch Biochem Biophys. 2016;603:91–101. doi:10.1016/j.abb.2016.05.013
  • Sarkar J, Chakraborti T, Chowdhury A, Bhuyan R, Chakraborti S. Protective role of epigallocatechin-3-gallate in NADPH oxidase-MMP2-Spm-Cer-S1P signalling axis mediated ET-1 induced pulmonary artery smooth muscle cell proliferation. J Cell Commun Signal. 2019;13(4):473–489. doi:10.1007/s12079-018-00501-7
  • Wang Y, Zhang X, Gao L, et al. Cortistatin exerts antiproliferation and antimigration effects in vascular smooth muscle cells stimulated by Ang II through suppressing ERK1/2, p38 MAPK, JNK and ERK5 signaling pathways. Ann Transl Med. 2019;7(20):561. doi:10.21037/atm.2019.09.45
  • Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165(1):111–124.
  • Duttaroy AK. Role of gut microbiota and their metabolites on atherosclerosis, hypertension and human blood platelet function: a review. Nutrients. 2021;13(1):144. doi:10.3390/nu13010144
  • Bennett BJ, De Aguiar Vallim TQ, Wang Z, et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013;17(1):49–60. doi:10.1016/j.cmet.2012.12.011
  • Ding L, Chang M, Guo Y, et al. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids Health Dis. 2018;17(1):286. doi:10.1186/s12944-018-0939-6
  • Fang Y, Yan C, Zhao Q, et al. The roles of microbial products in the development of colorectal cancer: a review. Bioengineered. 2021;12(1):720–735. doi:10.1080/21655979.2021.1889109
  • Dumas ME, Rothwell AR, Hoyles L, et al. Microbial-host co-metabolites are prodromal markers predicting phenotypic heterogeneity in behavior, obesity, and impaired glucose tolerance. Cell Rep. 2017;20(1):136–148. doi:10.1016/j.celrep.2017.06.039
  • Guo S, Luo Y. Brain Foxp3(+) regulatory T cells can be expanded by Interleukin-33 in mouse ischemic stroke. Int Immunopharmacol. 2020;81:106027. doi:10.1016/j.intimp.2019.106027
  • Zhao ZH, Xin FZ, Zhou D, et al. Trimethylamine N-oxide attenuates high-fat high-cholesterol diet-induced steatohepatitis by reducing hepatic cholesterol overload in rats. World J Gastroenterol. 2019;25(20):2450–2462. doi:10.3748/wjg.v25.i20.2450
  • Govindarajulu M, Pinky PD, Steinke I, et al. Gut metabolite TMAO induces synaptic plasticity deficits by promoting endoplasmic reticulum stress. Front Mol Neurosci. 2020;13:138. doi:10.3389/fnmol.2020.00138
  • Wang Z, Roberts AB, Buffa JA, et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163(7):1585–1595. doi:10.1016/j.cell.2015.11.055
  • Huang Y, Lin F, Tang R, et al. Gut microbial metabolite Trimethylamine N-Oxide aggravates pulmonary hypertension. Am J Respir Cell Mol Biol. 2022;66(4):452–460. doi:10.1165/rcmb.2021-0414OC
  • Shen X, Li L, Sun Z, et al. Gut microbiota and atherosclerosis-focusing on the plaque stability. Front Cardiovasc Med. 2021;8:668532. doi:10.3389/fcvm.2021.668532
  • Pluznick JL, Protzko RJ, Gevorgyan H, et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci U S A. 2013;110(11):4410–4415. doi:10.1073/pnas.1215927110
  • Ufnal M, Zadlo A, Ostaszewski R. TMAO: a small molecule of great expectations. Nutrition. 2015;31(11–12):1317–1323. doi:10.1016/j.nut.2015.05.006
  • Nøhr MK, Egerod KL, Christiansen SH, et al. Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience. 2015;290:126–137. doi:10.1016/j.neuroscience.2015.01.040
  • Brown AJ, Goldsworthy SM, Barnes AA, et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem. 2003;278(13):11312–11319. doi:10.1074/jbc.M211609200
  • Tang C, Offermanns S. FFA2 and FFA3 in metabolic regulation. Handb Exp Pharmacol. 2017;236:205–220.
  • Sun M, Wu W, Liu Z, Cong Y. Microbiota metabolite short chain fatty acids, GPCR, and inflammatory bowel diseases. J Gastroenterol. 2017;52(1):1–8. doi:10.1007/s00535-016-1242-9
  • Hsu CN, Yu HR, Lin IC, et al. Sodium butyrate modulates blood pressure and gut microbiota in maternal tryptophan-free diet-induced hypertension rat offspring. J Nutr Biochem. 2022;108:109090. doi:10.1016/j.jnutbio.2022.109090
  • Gurav A, Sivaprakasam S, Bhutia YD, Boettger T, Singh N, Ganapathy V. Slc5a8, a Na+-coupled high-affinity transporter for short-chain fatty acids, is a conditional tumour suppressor in colon that protects against colitis and colon cancer under low-fibre dietary conditions. Biochem J. 2015;469(2):267–278. doi:10.1042/BJ20150242
  • Park J, Kim M, Kang SG, et al. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol. 2015;8(1):80–93. doi:10.1038/mi.2014.44
  • Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–573. doi:10.1126/science.1241165
  • Lin MY, de Zoete MR, van Putten JP, Strijbis K. Redirection of epithelial immune responses by short-chain fatty acids through inhibition of histone deacetylases. Front Immunol. 2015;6:554. doi:10.3389/fimmu.2015.00554
  • Asarat M, Apostolopoulos V, Vasiljevic T, Donkor O. Short-chain fatty acids produced by synbiotic mixtures in skim milk differentially regulate proliferation and cytokine production in peripheral blood mononuclear cells. Int J Food Sci Nutr. 2015;66(7):755–765. doi:10.3109/09637486.2015.1088935
  • Yeo Y, Yi ES, Kim JM, et al. FGF12 (Fibroblast Growth Factor 12) Inhibits vascular smooth muscle cell remodeling in pulmonary arterial hypertension. Hypertension. 2020;76(6):1778–1786. doi:10.1161/HYPERTENSIONAHA.120.15068
  • Wu PH, Chiu YW, Zou HB, et al. Exploring the Benefit of 2-Methylbutyric acid in patients undergoing hemodialysis using a cardiovascular proteomics approach. Nutrients. 2019;11(12):3033. doi:10.3390/nu11123033
  • Bartolomaeus H, Balogh A, Yakoub M, et al. Short-chain fatty acid propionate protects from hypertensive cardiovascular damage. Circulation. 2019;139(11):1407–1421. doi:10.1161/CIRCULATIONAHA.118.036652
  • Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem. 2003;72:137–174. doi:10.1146/annurev.biochem.72.121801.161712
  • Chambers KF, Day PE, Aboufarrag HT, Kroon PA. Polyphenol effects on cholesterol metabolism via bile acid biosynthesis, CYP7A1: a review. Nutrients. 2019;11(11):2588. doi:10.3390/nu11112588
  • Dawson PA, Karpen SJ. Intestinal transport and metabolism of bile acids. J Lipid Res. 2015;56(6):1085–1099. doi:10.1194/jlr.R054114
  • Xu G, Pan LX, Li H, et al. Dietary cholesterol stimulates CYP7A1 in rats because farnesoid X receptor is not activated. Am J Physiol Gastrointest Liver Physiol. 2004;286(5):G730–735. doi:10.1152/ajpgi.00397.2003
  • Chen P, Zhang R, Mou L, Li X, Qin Y, Li X. An impaired hepatic clock system effects lipid metabolism in rats with nephropathy. Int J Mol Med. 2018;42(5):2720–2736. doi:10.3892/ijmm.2018.3833
  • Taniguchi T, Chen J, Cooper AD. Regulation of cholesterol 7 alpha-hydroxylase gene expression in Hep-G2 cells. Effect of serum, bile salts, and coordinate and noncoordinate regulation with other sterol-responsive genes. J Biol Chem. 1994;269(13):10071–10078. doi:10.1016/S0021-9258(17)36991-0
  • Li T, Francl JM, Boehme S, Chiang JY. Regulation of cholesterol and bile acid homeostasis by the cholesterol 7α-hydroxylase/steroid response element-binding protein 2/microRNA-33a axis in mice. Hepatology. 2013;58(3):1111–1121. doi:10.1002/hep.26427
  • Song KH, Li T, Owsley E, Chiang JY. A putative role of micro RNA in regulation of cholesterol 7alpha-hydroxylase expression in human hepatocytes. J Lipid Res. 2010;51(8):2223–2233. doi:10.1194/jlr.M004531
  • Yokota A, Fukiya S, Islam KB, et al. Is bile acid a determinant of the gut microbiota on a high-fat diet? Gut Microbes. 2012;3(5):455–459. doi:10.4161/gmic.21216
  • Desai MS, Mathur B, Eblimit Z, et al. Bile acid excess induces cardiomyopathy and metabolic dysfunctions in the heart. Hepatology. 2017;65(1):189–201. doi:10.1002/hep.28890
  • Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–585. doi:10.1038/nm.3145
  • Kazemian N, Mahmoudi M, Halperin F, Wu JC, Pakpour S. Gut microbiota and cardiovascular disease: opportunities and challenges. Microbiome. 2020;8(1):36. doi:10.1186/s40168-020-00821-0
  • Levi M. Role of bile acid-regulated nuclear receptor FXR and G protein-coupled receptor TGR5 in regulation of cardiorenal syndrome (Cardiovascular Disease and Chronic Kidney Disease). Hypertension. 2016;67(6):1080–1084. doi:10.1161/HYPERTENSIONAHA.115.06417
  • Porez G, Prawitt J, Gross B, Staels B. Bile acid receptors as targets for the treatment of dyslipidemia and cardiovascular disease. J Lipid Res. 2012;53(9):1723–1737. doi:10.1194/jlr.R024794
  • Comeglio P, Morelli A, Adorini L, Maggi M, Vignozzi L. Beneficial effects of bile acid receptor agonists in pulmonary disease models. Expert Opin Investig Drugs. 2017;26(11):1215–1228. doi:10.1080/13543784.2017.1385760
  • Nemet I, Saha PP, Gupta N, et al. A cardiovascular disease-linked gut microbial metabolite acts via adrenergic receptors. Cell. 2020;180(5):862–877.e822. doi:10.1016/j.cell.2020.02.016
  • Chen W, Zhang S, Wu J, et al. Butyrate-producing bacteria and the gut-heart axis in atherosclerosis. Clin Chim Acta. 2020;507:236–241. doi:10.1016/j.cca.2020.04.037
  • Huynh K. Novel gut microbiota-derived metabolite promotes platelet thrombosis via adrenergic receptor signalling. Nat Rev Cardiol. 2020;17(5):265. doi:10.1038/s41569-020-0367-y
  • Fang C, Zuo K, Fu Y, et al. Dysbiosis of gut microbiota and metabolite phenylacetylglutamine in coronary artery disease patients with stent stenosis. Front Cardiovasc Med. 2022;9:832092. doi:10.3389/fcvm.2022.832092
  • Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. 2018;23(6):716–724. doi:10.1016/j.chom.2018.05.003
  • Liu JR, Miao H, Deng DQ, Vaziri ND, Li P, Zhao YY. Gut microbiota-derived tryptophan metabolism mediates renal fibrosis by aryl hydrocarbon receptor signaling activation. Cell Mol Life Sci. 2021;78(3):909–922. doi:10.1007/s00018-020-03645-1
  • Niinisalo P, Oksala N, Levula M, et al. Activation of indoleamine 2,3-dioxygenase-induced tryptophan degradation in advanced atherosclerotic plaques: Tampere vascular study. Ann Med. 2010;42(1):55–63. doi:10.3109/07853890903321559
  • Song P, Ramprasath T, Wang H, Zou MH. Abnormal kynurenine pathway of tryptophan catabolism in cardiovascular diseases. Cell Mol Life Sci. 2017;74(16):2899–2916. doi:10.1007/s00018-017-2504-2
  • Sanchez-Gimenez R, Ahmed-Khodja W, Molina Y, et al. Gut microbiota-derived metabolites and cardiovascular disease risk: a systematic review of prospective cohort studies. Nutrients. 2022;14(13):2654. doi:10.3390/nu14132654
  • Barreto FC, Barreto DV, Liabeuf S, et al. Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol. 2009;4(10):1551–1558. doi:10.2215/CJN.03980609
  • Koike H, Morita T, Tatebe J, et al. The relationship between serum indoxyl sulfate and the renal function after catheter ablation of atrial fibrillation in patients with mild renal dysfunction. Heart Vessels. 2019;34(4):641–649. doi:10.1007/s00380-018-1288-0
  • Dou L, Jourde-Chiche N, Faure V, et al. The uremic solute indoxyl sulfate induces oxidative stress in endothelial cells. J Thromb Haemost. 2007;5(6):1302–1308. doi:10.1111/j.1538-7836.2007.02540.x
  • Huć T, Nowinski A, Drapala A, Konopelski P, Ufnal M. Indole and indoxyl sulfate, gut bacteria metabolites of tryptophan, change arterial blood pressure via peripheral and central mechanisms in rats. Pharmacol Res. 2018;130:172–179. doi:10.1016/j.phrs.2017.12.025
  • Imazu M, Fukuda H, Kanzaki H, et al. Plasma indoxyl sulfate levels predict cardiovascular events in patients with mild chronic heart failure. Sci Rep. 2020;10(1):16528. doi:10.1038/s41598-020-73633-9
  • Metghalchi S, Ponnuswamy P, Simon T, et al. Indoleamine 2,3-Dioxygenase fine-tunes immune homeostasis in atherosclerosis and colitis through repression of Interleukin-10 production. Cell Metab. 2015;22(3):460–471. doi:10.1016/j.cmet.2015.07.004
  • Sanchez-Rodriguez E, Egea-Zorrilla A, Plaza-Díaz J, et al. The gut microbiota and its implication in the development of atherosclerosis and related cardiovascular diseases. Nutrients. 2020;12(3):605. doi:10.3390/nu12030605