Atherosclerosis amelioration by allicin in raw garlic through gut microbiota and trimethylamine-N-oxide modulation

Suraphan Panyod, Wei-Kai Wu, Pei-Chen Chen, Kent-Vui Chong, Yu-Tang Yang, Hsiao-Li Chuang, Chieh-Chang Chen, Rou-An Chen, Po-Yu Liu, Ching-Hu Chung, Huai-Syuan Huang, Angela Yu-Chen Lin, Ting-Chin David Shen, Kai-Chien Yang, Tur-Fu Huang, Cheng-Chih Hsu, Chi-Tang Ho, Hsien-Li Kao, Alexander N Orekhov, Ming-Shiang Wu, Lee-Yan Sheen, Suraphan Panyod, Wei-Kai Wu, Pei-Chen Chen, Kent-Vui Chong, Yu-Tang Yang, Hsiao-Li Chuang, Chieh-Chang Chen, Rou-An Chen, Po-Yu Liu, Ching-Hu Chung, Huai-Syuan Huang, Angela Yu-Chen Lin, Ting-Chin David Shen, Kai-Chien Yang, Tur-Fu Huang, Cheng-Chih Hsu, Chi-Tang Ho, Hsien-Li Kao, Alexander N Orekhov, Ming-Shiang Wu, Lee-Yan Sheen

Abstract

Cardiovascular disease (CVD) is strongly associated with the gut microbiota and its metabolites, including trimethylamine-N-oxide (TMAO), formed from metaorganismal metabolism of ʟ-carnitine. Raw garlic juice, with allicin as its primary compound, exhibits considerable effects on the gut microbiota. This study validated the benefits of raw garlic juice against CVD risk via modulation of the gut microbiota and its metabolites. Allicin supplementation significantly decreased serum TMAO in ʟ-carnitine-fed C57BL/6 J mice, reduced aortic lesions, and altered the fecal microbiota in carnitine-induced, atherosclerosis-prone, apolipoprotein E-deficient (ApoE-/-) mice. In human subjects exhibiting high-TMAO production, raw garlic juice intake for a week reduced TMAO formation, improved gut microbial diversity, and increased the relative abundances of beneficial bacteria. In in vitro and ex vivo studies, raw garlic juice and allicin inhibited γ-butyrobetaine (γBB) and trimethylamine production by the gut microbiota. Thus, raw garlic juice and allicin can potentially prevent cardiovascular disease by decreasing TMAO production via gut microbiota modulation.

Conflict of interest statement

The authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Allicin reduced serum TMA, TMAO,…
Fig. 1. Allicin reduced serum TMA, TMAO, and γBB levels, and ʟ-carnitine principally changed the fecal microbiome composition in ʟ-carnitine-fed male C57BL/6 J mice (n = 10 per group).
a Experimental design; b serum TMA; c serum TMAO; d serum γBB; e principal coordinate analysis (PCoA) plot with Bray−Curtis dissimilarity; f observed OTUs α-diversity indices; and g relative abundance of fecal microbiota at family level. Dot plots are expressed as the mean ± SD or median; statistical analyses of serum bacterial metabolites were performed using the Wilcoxon signed-rank test Con vs. Con + A group, Con vs. Carn group; Kruskal–Wallis tests with Dunn’s multiple comparisons, Carn vs. Carn + A group, Carn vs. Carn + D group, and Carn + A vs. Carn + D group. Gut microbiota ɑ-diversity was determined using the unpaired two-tailed Student’s t-test; one-way ANOVA with Tukey’s range test. The statistical analyses of the relative abundance were performed using the unpaired Wilcoxon signed-rank test with the false discovery rate (FDR), Con vs. Con + A group (#P < 0.1); Con vs. Carn group (*P < 0.1); Carn vs. Carn + A group (†P < 0.1); Carn + D group (‡P < 0.1).
Fig. 2. Raw garlic juice reduced plasma…
Fig. 2. Raw garlic juice reduced plasma TMAO formation ability, increased plasma γBB level in healthy TMAO producers, and shaped fecal microbiota composition via increasing the evenness, ɑ-diversity, and relative abundance of specific beneficial bacterial taxa.
a Experimental design, healthy participants (n = 9) were screened for TMAO production using oral carnitine challenge test (OCCT), the criterion for categorization as high-TMAO producers was plasma TMAOMAX > 10 μM. High-TMAO producers (n = 7) subsequently received an intervention of garlic juice (55 mL, equivalent to 48 mg of allicin/day) for 1 week, followed by OCCT; b plasma and c urine TMAO, TMAOMAX, and TMAOAUC; d plasma γBB and γBBAUC. e Principal coordinate analysis (PCoA) plot with Bray−Curtis dissimilarity; f Shannon α-diversity index; g family-level composition of fecal microbiome; h volcano plot of fecal microbiota before and after garlic juice intervention; and i heatmap of the relative abundances of fecal microbiota with a significant difference using the Wilcoxon signed-rank test (P < 0.05). Data are expressed as the mean ± SEM; statistical analyses were performed by using the two-tailed paired Student’s t-test (*P < 0.05; **P < 0.01; and ***P < 0.001). The relative abundance bar plot statistical analysis was performed using a paired Wilcoxon signed-rank test (*P < 0.05). Volcano plot: red points indicate OTUs with a P-value < 0.05 and log2 fold-change > 1; orange points indicate OTUs with a P-value < 0.05; and blue points indicate log2 fold-change > 1.
Fig. 3. Allicin reduced aortic lesions through…
Fig. 3. Allicin reduced aortic lesions through the reduction of TMA and TMAO formation and changed the fecal microbiome composition in ʟ-carnitine-induced atherosclerosis female ApoE−/− mice (n = 9 per group).
a experimental design; b representative image of oil red-stained enface aorta, scale bar is 0.5 cm; c percentage of aortic lesion; d d9-TMA level according to oral carnitine challenge test (OCCT) and its AUC; e d9-TMAO level of OCCT and its AUC; f d9-γBB level according to OCCT and its AUC. g α-diversity indices, observed OTUs, Shannon index, and Chao1 index; h Principal coordinate analysis (PCoA) plot with Bray−Curtis dissimilarity; and i heatmap of the relative abundances of fecal microbiota with a significant difference using the Kruskal–Wallis test with false discovery rate (FDR) (P < 0.001). OCCT curves are expressed as the mean ± SEM, and dot plots are expressed as the mean ± SD; Statistical analyses were performed using an unpaired two-tailed Student’s t-test Con vs. Con + A group, Con vs. Carn group; one-way ANOVA with Tukey’s range test for comparisons Carn vs. Carn + A group, Carn vs. Carn + D group, and Carn + A vs. Carn + D group.
Fig. 4. Garlic-juice and allicin inhibited the…
Fig. 4. Garlic-juice and allicin inhibited the formation of d9-γBB and d9-TMA in vitro after inoculation of TMA/γBB-producing bacteria and ex vivo after inoculation of human feces from high-TMAO producers.
a inhibitory effect of garlic juice and allicin on the bacteria converting carnitine → γBB (co-culture of Proteus penneri, Escherichia fergusonii, and Edwardsiella tarda) in Wilkins–Chalgren (WC) broth supplemented with d9-carnitine; b inhibitory effect of garlic juice and allicin on the bacteria converting γBB to TMA (Emergencia timonensis) in WC broth supplemented with d9-γBB. c Inhibitory effect of garlic juice and allicin on the high-TMAO-producing gut microbiota in WC broth supplemented with d9-carnitine and d d9-γBB. The utilization of d9-carnitine/d9-γBB and the production of d9-γBB/d9-TMA were measured at 0, 6, 12, and 24 h. Half-maximal inhibitory concentration (IC50) values were determined using nonlinear regression (see supplementary Fig. 13).
Fig. 5. Effect of allicin and raw…
Fig. 5. Effect of allicin and raw garlic juice on CVD prevention and atherosclerosis amelioration through gut microbiota and TMAO modulation.
(1) Allicin decreased the gut microbiota- and host-derived TMA and TMAO levels in mice supplemented with ʟ-carnitine in drinking water; (2) Raw garlic juice reduced the plasma and urine TMAO levels and increased the beneficial gut bacterial abundance in high-TMAO producing humans; (3) Allicin ameliorated aortic lesions by reducing the TMA/TMAO production and altered the gut microbiome in ʟ-carnitine-induced atherosclerotic ApoE−/− mice; (4) Raw garlic juice and allicin inhibited microbial pathway of carnitine → γBB → TMA conversion in specific bacteria and high-TMAO producer’s gut microbiota.

References

    1. Roth GA, et al. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J. Am. Coll. Cardiol. 2017;70:1–25.
    1. Zhao D, Liu J, Wang M, Zhang X, Zhou M. Epidemiology of cardiovascular disease in china: current features and implications. Nat. Rev. Cardiol. 2019;16:203–212.
    1. Noble N, Paul C, Turon H, Oldmeadow C. Which modifiable health risk behaviours are related? A systematic review of the clustering of smoking, nutrition, alcohol and physical activity (‘SNAP’) health risk factors. Prev. Med. 2015;81:16–41.
    1. Clarke G, et al. Minireview: gut microbiota: the neglected endocrine organ. Mol. Endocrinol. 2014;28:1221–1238.
    1. David LA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563.
    1. Jie Z, et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017;8:845.
    1. Brown JM, Hazen SL. The gut microbial endocrine organ: bacterially derived signals driving cardiometabolic diseases. Annu. Rev. Med. 2015;66:343–359.
    1. Gentile CL, Weir TL. The gut microbiota at the intersection of diet and human health. Science. 2018;362:776–780.
    1. Witkowski M, Weeks TL, Hazen SL. Gut microbiota and cardiovascular disease. Circ. Res. 2020;127:553–570.
    1. Koeth RA, et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013;19:576–585.
    1. Tilg H. A gut feeling about thrombosis. New. Engl. J. Med. 2016;374:2494–2496.
    1. Wang Z, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63.
    1. Schiattarella GG, et al. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur. Heart J. 2017;38:2948–294.
    1. Tang WH, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013;368:1575–1584.
    1. Zhu Y, et al. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc. Natl Acad. Sci. USA. 2014;111:4268–4273.
    1. Zhu W, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165:111–124.
    1. Zhu WF, Wang ZN, Tang WHW, Hazen SL. Gut microbe-generated trimethylamine N-oxide from dietary choline is prothrombotic in subjects. Circulation. 2017;135:1671–1673.
    1. Koeth RA, et al. L-carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Invest. 2019;129:373–387.
    1. Koeth RA, et al. γ-butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab. 2014;20:799–812.
    1. Wu WK, et al. Characterization of TMAO productivity from carnitine challenge facilitates personalized nutrition and microbiome signatures discovery. Microbiome. 2020;8:162.
    1. Barabási A-L, Menichetti G, Loscalzo J. The unmapped chemical complexity of our diet. Nat. Food. 2020;1:33–37.
    1. Roberts AB, et al. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat. Med. 2018;24:1407–1417.
    1. Smits, L. P. et al. Effect of vegan fecal microbiota transplantation on carnitine- and choline-derived trimethylamine-N-oxide production and vascular inflammation in patients with metabolic syndrome. J. Am. Heart. Assoc.7, e008342 (2018).
    1. Wang Z, et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163:1585–1595.
    1. Simo C, Garcia-Canas V. Dietary bioactive ingredients to modulate the gut microbiota-derived metabolite TMAO. New opportunities for functional food development. Food Funct. 2020;11:6745–6776.
    1. He Z, et al. Hawthorn fruit extract reduced trimethylamine-N-oxide (TMAO)-exacerbated atherogenesis in mice via anti-inflammation and anti-oxidation. Nutr. Metab. (Lond.) 2021;18:6.
    1. Li X, et al. Berberine attenuates choline-induced atherosclerosis by inhibiting trimethylamine and trimethylamine-N-oxide production via manipulating the gut microbiome. NPJ Biofilms Microbiomes. 2021;7:36.
    1. Panyod, S. et al. Modulation of gut microbiota by foods and herbs to prevent cardiovascular diseases. J. Tradit. Complement. Med. 10.1016/j.jtcme.2021.09.006 (2021).
    1. Petrovska BB, Cekovska S. Extracts from the history and medical properties of garlic. Pharmacogn. Rev. 2010;4:106–110.
    1. Banerjee SK, Maulik SK. Effect of garlic on cardiovascular disorders: a review. Nutr. J. 2002;1:4.
    1. Liperoti R, Vetrano DL, Bernabei R, Onder G. Herbal medications in cardiovascular medicine. J. Am. Coll. Cardiol. 2017;69:1188–1199.
    1. Rahman K, Lowe GM. Garlic and cardiovascular disease: a critical review. J. Nutr. 2006;136:736–740.
    1. Sobenin, I. A. et al. The effects of time-released garlic powder tablets on multifunctional cardiovascular risk in patients with coronary artery disease. Lipids Health Dis. 9, 119 (2010).
    1. Harris JC, Cottrell SL, Plummer S, Lloyd D. Antimicrobial properties of Allium sativum (garlic) Appl. Microbiol. Biotechnol. 2001;57:282–286.
    1. Ankri S, Mirelman D. Antimicrobial properties of allicin from garlic. Microbes Infect. 1999;1:125–129.
    1. Choo S, et al. Review: antimicrobial properties of allicin used alone or in combination with other medications. Folia Microbiol. 2020;65:451–465.
    1. Reiter, J. et al. Diallylthiosulfinate (allicin), a volatile antimicrobial from garlic (Allium sativum), kills human lung pathogenic bacteria, including MDR strains, as a vapor. Molecules 12, 22 (2017).
    1. Panyod S, Sheen LY. Beneficial effects of Chinese herbs in the treatment of fatty liver diseases. J. Tradit. Complement. Med. 2020;10:260–267.
    1. Panyod S, et al. Allicin modifies the composition and function of the gut microbiota in alcoholic hepatic steatosis mice. J. Agric. Food Chem. 2020;68:3088–3098.
    1. Wu WK, et al. Dietary allicin reduces transformation of L-carnitine to TMAO through impact on gut microbiota. J. Funct. Foods. 2015;15:408–417.
    1. Wu W-K, 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.
    1. Kasahara K, et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat. Microbiol. 2018;3:1461–1471.
    1. Li J, Lin S, Vanhoutte PM, Woo CW, Xu A. Akkermansia muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in Apoe−/− mice. Circulation. 2016;133:2434–2446.
    1. Chen, K. et al. Preventive effects and mechanisms of garlic on dyslipidemia and gut microbiome dysbiosis. Nutrients 11, 1225 (2019).
    1. O’Gara EA, Hill DJ, Maslin DJ. Activities of garlic oil, garlic powder, and their diallyl constituents against Helicobacter pylori. Appl. Environ. Microb. 2000;66:2269–2273.
    1. Naganawa R, et al. Inhibition of microbial growth by ajoene, a sulfur-containing compound derived from garlic. Appl. Environ. Microb. 1996;62:4238–4242.
    1. Casella S, Leonardi M, Melai B, Fratini F, Pistelli L. The role of diallyl sulfides and dipropyl sulfides in the in vitro antimicrobial activity of the essential oil of garlic, Allium sativum L., and leek, Allium porrum L. Phytother. Res. 2013;27:380–383.
    1. Avato P, Tursi F, Vitali C, Miccolis V, Candido V. Allylsulfide constituents of garlic volatile oil as antimicrobial agents. Phytomedicine. 2000;7:239–243.
    1. Rajakovich, L. J., Fu, B., Bollenbach, M. & Balskus, E. P. Elucidation of an anaerobic pathway for metabolism of L-carnitine-derived γ-butyrobetaine to trimethylamine in human gut bacteria. Proc. Natl. Acad. Sci. USA118, e2101498118 (2021).
    1. Zhang C, et al. Allicin-induced host-gut microbe interactions improves energy homeostasis. FASEB J. 2020;34:10682–10698.
    1. Everard A, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl Acad. Sci. USA. 2013;110:9066–9071.
    1. Chelakkot, C. et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp. Mol. Med. 50, e450 (2018).
    1. Yoon HS, et al. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat. Microbiol. 2021;6:563–573.
    1. Sokol H, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl Acad. Sci. USA. 2008;105:16731–16736.
    1. Panyod S, et al. Diet supplementation with allicin protects against alcoholic fatty liver disease in mice by improving anti-inflammation and antioxidative functions. J. Agric. Food Chem. 2016;64:7104–7113.
    1. Robinet P, et al. Consideration of sex differences in design and reporting of experimental arterial pathology studies-statement from ATVB council. Arterioscler. Thromb. Vasc. Biol. 2018;38:292–303.
    1. Bennett BJ, et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013;17:49–60.
    1. Beattie JH, Duthie SJ, Kwun IS, Ha TY, Gordon MJ. Rapid quantification of aortic lesions in ApoE−/− mice. J. Vasc. Res. 2009;46:347–352.
    1. Lai YS, et al. Garlic essential oil protects against obesity-triggered nonalcoholic fatty liver disease through modulation of lipid metabolism and oxidative stress. J. Agric. Food Chem. 2014;62:5897–5906.
    1. Goodman AL, et al. Extensive personal human gut microbiota culture collections characterized and manipulated in gnotobiotic mice. Proc. Natl Acad. Sci. USA. 2011;108:6252–6257.
    1. Krautkramer KA, et al. Diet-microbiota interactions mediate global epigenetic programming in multiple host tissues. Mol. Cell. 2016;64:982–992.
    1. Mortele, O. et al. Optimization of an in vitro gut microbiome biotransformation platform with chlorogenic acid as model compound: from fecal sample to biotransformation product identification. J. Pharmaceut. Biomed. Anal. 175, 112768 (2019).
    1. Romano KA, Vivas EI, Amador-Noguez D, Rey FE. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. mBio. 2015;6:e02481.
    1. Wu WK, et al. Optimization of fecal sample processing for microbiome study—the journey from bathroom to bench. J. Formos. Med. Assoc. 2019;118:545–555.
    1. Comeau, A. M., Douglas, G. M. & Langille, M. G. Microbiome helper: a custom and streamlined workflow for microbiome research. mSystems10.1128/mSystems.00127-16 (2017).
    1. Rognes T, Flouri T, Nichols B, Quince C, Mahe F. Vsearch: a versatile open source tool for metagenomics. PeerJ. 2016;4:e2584.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods. 2010;7:335–336.

Source: PubMed

3
Abonneren