Resveratrol Attenuates Trimethylamine-N-Oxide (TMAO)-Induced Atherosclerosis by Regulating TMAO Synthesis and Bile Acid Metabolism via Remodeling of the Gut Microbiota

Ming-liang Chen, Long Yi, Yong Zhang, Xi Zhou, Li Ran, Jining Yang, Jun-dong Zhu, Qian-yong Zhang, Man-tian Mi, Ming-liang Chen, Long Yi, Yong Zhang, Xi Zhou, Li Ran, Jining Yang, Jun-dong Zhu, Qian-yong Zhang, Man-tian Mi

Abstract

The gut microbiota is found to be strongly associated with atherosclerosis (AS). Resveratrol (RSV) is a natural phytoalexin with anti-AS effects; however, its mechanisms of action remain unclear. Therefore, we sought to determine whether the anti-AS effects of RSV were related to changes in the gut microbiota. We found that RSV attenuated trimethylamine-N-oxide (TMAO)-induced AS in ApoE(-/-) mice. Meanwhile, RSV decreased TMAO levels by inhibiting commensal microbial trimethylamine (TMA) production via gut microbiota remodeling in mice. Moreover, RSV increased levels of the genera Lactobacillus and Bifidobacterium, which increased the bile salt hydrolase activity, thereby enhancing bile acid (BA) deconjugation and fecal excretion in C57BL/6J and ApoE(-/-) mice. This was associated with a decrease in ileal BA content, repression of the enterohepatic farnesoid X receptor (FXR)-fibroblast growth factor 15 (FGF15) axis, and increased cholesterol 7a-hydroxylase (CYP7A1) expression and hepatic BA neosynthesis. An FXR antagonist had the same effect on FGF15 and CYP7A1 expression as RSV, while an FXR agonist abolished RSV-induced alterations in FGF15 and CYP7A1 expression. In mice treated with antibiotics, RSV neither decreased TMAO levels nor increased hepatic BA synthesis. Additionally, RSV-induced inhibition of TMAO-caused AS was also markedly abolished by antibiotics. In conclusion, RSV attenuated TMAO-induced AS by decreasing TMAO levels and increasing hepatic BA neosynthesis via gut microbiota remodeling, and the BA neosynthesis was partially mediated through the enterohepatic FXR-FGF15 axis.

Importance: Recently, trimethylamine-N-oxide (TMAO) has been identified as a novel and independent risk factor for promoting atherosclerosis (AS) partially through inhibiting hepatic bile acid (BA) synthesis. The gut microbiota plays a key role in the pathophysiology of TMAO-induced AS. Resveratrol (RSV) is a natural phytoalexin with prebiotic benefits. A growing body of evidence supports the hypothesis that phenolic phytochemicals with poor bioavailability are possibly acting primarily through remodeling of the gut microbiota. The current study showed that RSV attenuated TMAO-induced AS by decreasing TMAO levels and increasing hepatic BA neosynthesis via gut microbiota remodeling. And RSV-induced hepatic BA neosynthesis was partially mediated through downregulating the enterohepatic farnesoid X receptor-fibroblast growth factor 15 axis. These results offer new insights into the mechanisms responsible for RSV's anti-AS effects and indicate that the gut microbiota may become an interesting target for pharmacological or dietary interventions to decrease the risk of developing cardiovascular diseases.

Copyright © 2016 Chen et al.

Figures

FIG 1
FIG 1
RSV inhibited TMAO synthesis in C57BL/6J mice. (A and B) Eight-week-old female C57BL/6J mice were administered choline (400 mg/kg of body weight, n = 10) (A) or TMA (40 mg/kg, n = 10) (B). Blood samples were collected at the indicated times. Serum TMA and TMAO levels were determined by LC/MS. (C and D) Eight-week-old female C57BL/6J mice were fed a chow diet with or without RSV (0.4%) in the presence or absence of choline (1%) or Abs. After 30 days, several mice of the chow-and-RSV-fed group were administered choline (400 mg/kg, n = 10) (C) or TMA (40 mg/kg, n = 10) (D). At 4 h after choline was given, or 1 h after TMA was given, the mice were euthanized and blood was collected. Serum TMA and TMAO levels were determined by LC/MS. (E and F) The other mice were also euthanized, and blood was collected. Serum TMA and TMAO levels (E) and RSV levels (F) were determined by LC/MS. Values are presented as means ± SD (n = 10). a, P < 0.05; b, P < 0.01 (versus vehicle-treated control group); d, P < 0.01 (versus choline-treated group); #, P < 0.05 (versus group cotreated with choline and RSV).
FIG 2
FIG 2
RSV inhibited TMAO synthesis by decreasing TMA generation via remodeling microbiota in C57BL/6J mice. Eight-week-old female C57BL/6J mice (n = 5 per group) were fed chow with or without RSV (0.4%) for 30 days. ACTB, β-actin. (A) Western blotting detection of FMO3 in the liver. (B) Expression levels of FMO3 gene mRNAs were quantified using qPCR assays. (C) Liver FMO activity was assessed as described in Materials and Methods. (D) 16S rRNA gene sequencing analysis of cecal content at the phylum level. (E) Heat map of 16S rRNA gene sequencing analysis of cecal content at the genus level. The scale reflects the data as follows: red indicates high values whereas blue indicates low values for the percentages of reads that were classified at that rank. (F) Linear discriminant analysis (LDA) coupled with effect size measurements identifies the taxons most differentially abundant between the chow and RSV diets at the genus level. RSV-diet-enriched taxa are indicated with a positive LDA score (green), and taxa enriched in the normal chow diet have a negative score (red). Only taxa meeting an LDA significant threshold value of >2 are shown. (G) Correlation heat map demonstrating the association between the indicated microbiota taxonomic genera and TMA and TMAO concentrations (all reported as means ± SD in micromoles) from mice grouped by dietary status (chow and RSV). Red denotes a positive association, blue a negative association, and white no association. A single asterisk indicates a significant FDR-adjusted association at P values of ≤0.05, and a double asterisk indicates a significant FDR-adjusted association at P values of ≤0.01. (H) The production of TMA from choline by cecal content in vitro. (I) Cecal RSV content. (J) The same 8-week-old female C57BL/6J mice (n = 5 per group) were administered choline (400 mg/kg) at visit 1 (baseline), visit 2 (RSV treatment for 1 month), and visit 3 (washout for 1 month). Four hours after choline was given at each visit, blood samples were obtained from the tail veins. Serum TMA and TMAO levels were determined by LC/MS. Values are presented as means ± SD (n = 5). a, P < 0.05; b, P < 0.01 (versus vehicle-treated control group); &, P < 0.01 (versus RSV-treated group).
FIG 3
FIG 3
RSV enhanced BA deconjugation and fecal excretion in C57BL/6J mice. Eight-week-old female C57BL/6J mice (n = 10 per group) were fed chow with or without RSV (0.4%) in the presence or absence of Abs for 30 days. Fecal samples were collected. (A) Fecal BA excretion was measured by a total BA test (Wako). Fecal BA composition was determined by LC/MS as described in Materials and Methods. (B) CA/DCA ratio. (C) Conjugated/unconjugated (conj/unconj) BA ratio in fecal samples. (D) BSH activity. Values are expressed as means ± SD (n = 10). b, P < 0.01 (versus vehicle-treated control group); &, P < 0.01 (versus RSV-treated group).
FIG 4
FIG 4
RSV induced hepatic BA synthesis in C57BL/6J mice. Eight-week-old female C57BL/6J mice (n = 10 per group) were fed chow with or without RSV (0.4%) in the presence or absence of Abs for 30 days. (A) Liver cholesterol content. (B) The BA pool size was determined for the total BA content of gallbladder bile, liver, and the SI lumenal. (C to F) Total biliary BA content (C) and total BA content in SI lumenal (D), serum (E), and liver (F) were detected. (G) Gallbladder bile samples were subjected to LC/MS, and TCA/TβMCA ratios were calculated. (H) Relative expression levels of the indicated mRNAs in the liver were determined by qPCR. (I) Expression of CYP7A1 was analyzed by Western blotting. Values are expressed as means ± SD (n = 10). a, P < 0.05; b, P < 0.01 (versus vehicle-treated control group); &, P < 0.01 (versus RSV-treated group).
FIG 5
FIG 5
The enterohepatic FXR-FGF15 axis played a key role in RSV-induced BA synthesis. Eight-week-old female C57BL/6J mice (n = 10 per group) were fed chow with or without RSV (0.4%) for 30 days. Z-Gug (100 mg/kg body weight) or GW4064 (75 mg/kg body weight) was given 7 days prior to surgical procedures. (A) Relative expression levels of ileal FXR and FGF15 gene mRNAs. (B) Western blotting was used to detect ileal FXR and FGF15 expression. (C) Expression of the indicated proteins in ileal tissues. (D) Liver CYP7A1 expression was detected by Western blotting. (E) FGF15 expression in ileal tissues. (F) CYP7A1 expression in liver samples. Values are expressed as means ± SD (n = 10). b, P < 0.01 (versus vehicle-treated control group); &, P < 0.01 (versus RSV-treated group).
FIG 6
FIG 6
RSV protected ApoE−/− mice from TMAO-induced AS. Eight-week-old female ApoE−/− mice (n = 10 per group) were given RSV (0.4%) with or without choline (1%) in the absence or presence of Abs for 4 months. The control group was fed with a chow diet. (A) Ultrasound B-mode images of the aortic sinus and quantification. Arrows indicate the regions of interest. RV, right ventricle; RA, right atrium; LA, left atrium. (B) Oil red O-stained aortic roots (counterstained with hematoxylin) and quantification. (C) Oil red O staining of whole aortas, including the aortic arch, thoracic, and abdominal regions, and their quantitation. (D) Total cholesterol content in the thoracic and abdominal aorta. Values are expressed as means ± SD (n = 10). a, P < 0.05; b, P < 0.01 (versus vehicle-treated control group); c, P < 0.05; d, P < 0.01 (versus choline-treated group); #, P < 0.05 (versus group cotreated with RSV and choline).
FIG 7
FIG 7
RSV reduced TMAO levels by decreasing TMA generation via remodeling microbiota in ApoE−/− mice. Eight-week-old female ApoE−/− mice (n = 5 per group) were fed chow, chow with RSV (0.4%), chow with choline (1%), or chow with choline (1%) plus RSV (0.4%) in the absence or presence of Abs for 4 months. (A) Serum TMA and TMAO levels were measured by LC/MS. (B) Western blotting detection of FMO3 expression in the liver. (C) Expression levels of FMO3 gene mRNAs were quantified using qPCR assays. (D) Liver FMO activity was assessed as described in Materials and Methods. (E) 16S rRNA gene sequencing analysis of cecal content at the phylum level. (F) Heat map of 16S rRNA gene sequencing analysis of cecal content at the genus level. The scale reflects the data as follows: red indicates high values whereas blue indicates low values for the spercentage of reads that were classified at that rank. (G) Linear discriminant analysis (LDA) coupled with effect size measurements identifies the most differentially abundant taxons at the genus level between the choline diet and the RSV-plus-choline diet. RSV-plus-choline-diet-enriched taxa are indicated with a positive LDA score (green), and taxa enriched in the choline diet have a negative score (red). Only taxa meeting an LDA significance threshold of >2 are shown. (H) Correlation heat map demonstrating the association between the indicated microbiota taxonomic genera and TMA and TMAO concentrations (all reported as means ± SD in micromoles) from mice grouped by dietary status (choline and RSV plus choline). Red denotes a positive association, blue a negative association, and white no association. A single asterisk indicates a significant FDR-adjusted association of P values of ≤0.05, and a double asterisk indicates a significant FDR-adjusted association of P values of ≤0.01. (I) Production of TMA from choline by cecal content in vitro. Values are expressed as means ± SD (n = 5). a, P < 0.05; b, P < 0.01 (versus vehicle-treated control group); c, P < 0.05; d, P < 0.01 (versus choline-treated group); #, P < 0.05 (versus group cotreated with RSV and choline).
FIG 8
FIG 8
RSV induced hepatic BA synthesis in choline-treated ApoE−/− mice. Eight-week-old female ApoE−/− mice (n = 10 per group) were fed chow, chow with RSV (0.4%), chow with choline (1%), or chow with choline (1%) plus RSV (0.4%) in the absence or presence of Abs for 4 months. (A) Serum TC levels were measured enzymatically using commercially available kits. (B) Liver cholesterol content. (C) The BA pool size was determined for the total BA content of gallbladder bile, liver, and the SI lumenal. BW, body weight. (D to F) Total biliary BA content (D) and total BA content in SI lumenal (E) and liver (F) were detected. (G) Relative expression levels of the indicated mRNAs in the liver were determined by qPCR. (H) Expression of CYP7A1 was analyzed by Western blotting. Values are expressed as means ± SD (n = 10). a, P < 0.05; b, P < 0.01 (versus vehicle-treated control group); c, P < 0.05; d, P < 0.01 (versus choline-treated group); #, P < 0.05 (versus choline and RSV-cotreated group).
FIG 9
FIG 9
RSV downregulated the enterohepatic FXR-FGF15 axis in choline-treated ApoE−/− mice. Eight-week-old female ApoE−/− mice (n = 10 per group) were fed chow, chow with RSV (0.4%), chow with choline (1%), or chow with choline (1%) plus RSV (0.4%) in the absence or presence of Abs for 4 months. (A) Expression of CYP7A1 was analyzed by Western blotting. (B and C) Relative expression levels of the indicated mRNAs in the liver (B) and ileal tissue (C) were determined by qPCR. (D) Western blotting was used to detect ileal FXR and FGF15 expression. (E and F) BA content in SI tissue (E) and fecal samples (F). b, P < 0.01 (versus vehicle-treated control group); d, P < 0.01 (versus choline-treated group); #, P < 0.05 (versus group cotreated with choline and RSV).
FIG 10
FIG 10
A model depicting use of RSV to attenuate AS by targeting gut microbiota. Dietary RSV can alter the composition of gut flora. On the one hand, this can result in reduced levels of gut microbial TMA production, subsequently leading to decreased TMAO synthesis in the liver and, ultimately, to inhibition of AS. On the other hand, RSV increases BSH activity by gut microbiota remodeling, which promotes the generation of unconjugated BAs from conjugated BAs and enhances fecal BA loss. RSV-induced fecal BA loss leads to a decrease in ileal BA content, thereby inhibiting the ileal FXR-FGF15 axis, and then increases the expression levels of CYP7A1 in the liver, subsequently inducing hepatic BA neosynthesis which contributes to cholesterol homeostasis, finally attenuating AS. Red indicates the effect of RSV, purple indicates downregulation by RSV, green indicates upregulation by RSV, and blue indicates the gut flora. LCA, lithocholic acid.

References

    1. Murray CJ, Lopez AD. 1997. Alternative projections of mortality and disability by cause 1990–2020: Global Burden of Disease Study. Lancet 349:1498–1504. doi:10.1016/S0140-6736(96)07492-2.
    1. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. 2011. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472:57–63. doi:10.1038/nature09922.
    1. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL. 2013. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 368:1575–1584. doi:10.1056/NEJMoa1109400.
    1. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH. 2013. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19:576–585. doi:10.1038/nm.3145.
    1. Sayin SI, Wahlström A, Felin J, Jäntti S, Marschall HU, Bamberg K, Angelin B, Hyötyläinen T, Orešič M, Bäckhed F. 2013. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab 17:225–235. doi:10.1016/j.cmet.2013.01.003.
    1. Jones ML, Tomaro-Duchesneau C, Prakash S. 2014. The gut microbiome, probiotics, bile acids axis, and human health. Trends Microbiol 22:306–308. doi:10.1016/j.tim.2014.04.010.
    1. Li-Hawkins J, Gåfvels M, Olin M, Lund EG, Andersson U, Schuster G, Björkhem I, Russell DW, Eggertsen G. 2002. Cholic acid mediates negative feedback regulation of bile acid synthesis in mice. J Clin Invest 110:1191–1200. doi:10.1172/JCI16309.
    1. Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. 2009. Bile acids as regulatory molecules. J Lipid Res 50:1509–1520. doi:10.1194/jlr.R900007-JLR200.
    1. Miyata M, Takamatsu Y, Kuribayashi H, Yamazoe Y. 2009. Administration of ampicillin elevates hepatic primary bile Acid synthesis through suppression of ileal fibroblast growth factor 15 expression. J Pharmacol Exp Ther 331:1079–1085. doi:10.1124/jpet.109.160093.
    1. Degirolamo C, Rainaldi S, Bovenga F, Murzilli S, Moschetta A. 2014. Microbiota modification with probiotics induces hepatic bile acid synthesis via downregulation of the FXR-FGF15 axis in mice. Cell Rep 7:12–18. doi:10.1016/j.celrep.2014.02.032.
    1. Miyata M, Sakaida Y, Matsuzawa H, Yoshinari K, Yamazoe Y. 2011. Fibroblast growth factor 19 treatment ameliorates disruption of hepatic lipid metabolism in farnesoid X receptor (FXR)-null mice. Biol Pharm Bull 34:1885–1889. doi:10.1248/bpb.34.1885.
    1. Li F, Jiang CT, Krausz KW, Li YF, Albert I, Hao HP, Fabre KM, Mitchell JB, Patterson AD, Gonzalez FJ. 2013. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat Commun 4:2384. doi:10.1038/ncomms3384.
    1. Chung JH, Manganiello V, Dyck JR. 2012. Resveratrol as a calorie restriction mimetic: therapeutic implications. Trends Cell Biol 22:546–554. doi:10.1016/j.tcb.2012.07.004.
    1. Subramanian L, Youssef S, Bhattacharya S, Kenealey J, Polans AS, van Ginkel PR. 2010. Resveratrol: challenges in translation to the clinic—a critical discussion. Clin Cancer Res 16:5942–5948. doi:10.1158/1078-0432.CCR-10-1486.
    1. Asensi M, Medina I, Ortega A, Carretero J, Baño MC, Obrador E, Estrela JM. 2002. Inhibition of cancer growth by resveratrol is related to its low bioavailability. Free Radic Biol Med 33:387–398. doi:10.1016/S0891-5849(02)00911-5.
    1. Shin NR, Lee JC, Lee HY, Kim MS, Whon TW, Lee MS, Bae JW. 2014. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63:727–735. doi:10.1136/gutjnl-2012-303839.
    1. Anhê FF, Roy D, Pilon G, Dudonné S, Matamoros S, Varin TV, Garofalo C, Moine Q, Desjardins Y, Levy E, Marette A. 2015. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 64:872–883 doi:10.1136/gutjnl-2014-307142.
    1. Larrosa M, Yañéz-Gascón MJ, Selma MV, González-Sarrías A, Toti S, Cerón JJ, Tomás-Barberán F, Dolara P, Espín JC. 2009. Effect of a low dose of dietary resveratrol on colon microbiota, inflammation and tissue damage in a DSS-induced colitis rat model. J Agric Food Chem 57:2211–2220. doi:10.1021/jf803638d.
    1. Jung CM, Heinze TM, Schnackenberg LK, Mullis LB, Elkins SA, Elkins CA, Steele RS, Sutherland JB. 2009. Interaction of dietary resveratrol with animal-associated bacteria. FEMS Microbiol Lett 297:266–273. doi:10.1111/j.1574-6968.2009.01691.x.
    1. Queipo-Ortuño MI, Boto-Ordóñez M, Murri M, Gomez-Zumaquero JM, Clemente-Postigo M, Estruch R, Cardona Diaz F, Andrés-Lacueva C, Tinahones FJ. 2012. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am J Clin Nutr 95:1323–1334. doi:10.3945/ajcn.111.027847.
    1. Qiao Y, Sun J, Xia S, Tang X, Shi Y, Le G. 2014. Effects of resveratrol on gut microbiota and fat storage in a mouse model with high-fat-induced obesity. Food Funct 5:1241–1249. doi:10.1039/c3fo60630a.
    1. Etxeberria U, Arias N, Boqué N, Macarulla MT, Portillo MP, Martínez JA, Milagro FI. 2015. Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J Nutr Biochem 26:651–660. doi:10.1016/j.jnutbio.2015.01.002.
    1. Bennett BJ, de Aguiar Vallim TQ, Wang Z, Shih DM, Meng Y, Gregory J, Allayee H, Lee R, Graham M, Crooke R, Edwards PA, Hazen SL, Lusis AJ. 2013. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab 17:49–60. doi:10.1016/j.cmet.2012.12.011.
    1. Ridlon JM, Kang DJ, Hylemon PB. 2006. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 47:241–259. doi:10.1194/jlr.R500013-JLR200.
    1. Zhang Y, Limaye PB, Lehman-McKeeman LD, Klaassen CD. 2012. Dysfunction of organic anion transporting polypeptide 1a1 alters intestinal bacteria and bile acid metabolism in mice. PLoS One 7:e34522. doi:10.1371/journal.pone.0034522.
    1. Alrefai WA, Gill RK. 2007. Bile acid transporters: structure, function, regulation and pathophysiological implications. Pharm Res 24:1803–1823. doi:10.1007/s11095-007-9289-1.
    1. Herrema H, Meissner M, van Dijk TH, Brufau G, Boverhof R, Oosterveer MH, Reijngoud DJ, Müller M, Stellaard F, Groen AK, Kuipers F. 2010. Bile salt sequestration induces hepatic de novo lipogenesis through farnesoid X receptor- and liver X receptor alpha-controlled metabolic pathways in mice. Hepatology 51:806–816. doi:10.1002/hep.23408.
    1. Out C, Hageman J, Bloks VW, Gerrits H, Sollewijn Gelpke MD, Bos T, Havinga R, Smit MJ, Kuipers F, Groen AK. 2011. Liver receptor homolog-1 is critical for adequate up-regulation of Cyp7a1 gene transcription and bile salt synthesis during bile salt sequestration. Hepatology 53:2075–2085. doi:10.1002/hep.24286.
    1. Chiang JY. 2009. Bile acids: regulation of synthesis. J Lipid Res 50:1955–1966. doi:10.1194/jlr.R900010-JLR200.
    1. Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, Gonzalez FJ. 2000. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 102:731–744. doi:10.1016/S0092-8674(00)00062-3.
    1. Inagaki T, Choi M, Moschetta A, Peng L, Cummins CL, McDonald JG, Luo G, Jones SA, Goodwin B, Richardson JA, Gerard RD, Repa JJ, Mangelsdorf DJ, Kliewer SA. 2005. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab 2:217–225. doi:10.1016/j.cmet.2005.09.001.
    1. Lu TT, Makishima M, Repa JJ, Schoonjans K, Kerr TA, Auwerx J, Mangelsdorf DJ. 2000. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell 6:507–515. doi:10.1016/S1097-2765(00)00050-2.
    1. Xiao D, Singh SV. 2008. z-Guggulsterone, a constituent of ayurvedic medicinal plant Commiphora mukul, inhibits angiogenesis in vitro and in vivo. Mol Cancer Ther 7:171–180. doi:10.1158/1535-7163.MCT-07-0491.
    1. Kemper JK, Xiao Z, Ponugoti B, Miao J, Fang S, Kanamaluru D, Tsang S, Wu SY, Chiang CM, Veenstra TD. 2009. FXR acetylation is normally dynamically regulated by p300 and SIRT1 but constitutively elevated in metabolic disease states. Cell Metab 10:392–404. doi:10.1016/j.cmet.2009.09.009.
    1. Purushotham A, Xu Q, Lu J, Foley JF, Yan X, Kim DH, Kemper JK, Li X. 2012. Hepatic deletion of SIRT1 decreases hepatocyte nuclear factor 1 alpha/farnesoid X receptor signaling and induces formation of cholesterol gallstones in mice. Mol Cell Biol 32:1226–1236. doi:10.1128/MCB.05988-11.
    1. Kulkarni SS, Cantó C. 2015. The molecular targets of resveratrol. Biochim Biophys Acta 1852:1114–1123. doi:10.1016/j.bbadis.2014.10.005.
    1. El-Mowafy AM, El-Mesery ME, Salem HA, Al-Gayyar MM, Darweish MM. 2010. Prominent chemopreventive and chemoenhancing effects for resveratrol: unraveling molecular targets and the role of C-reactive protein. Chemotherapy 56:60–65. doi:10.1159/000298821.
    1. Zhang Y, Chen ML, Zhou Y, Yi L, Gao YX, Ran L, Chen SH, Zhang T, Zhou X, Zou D, Wu B, Wu Y, Chang H, Zhu JD, Zhang QY, Mi MT. 2015. Resveratrol improves hepatic steatosis by inducing autophagy through the cAMP signaling pathway. Mol Nutr Food Res 59:1443–1457. doi:10.1002/mnfr.201500016.
    1. Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, Gu X, Huang Y, Zamanian-Daryoush M, Culley MK, DiDonato AJ, Fu X, Hazen JE, Krajcik D, DiDonato JA, Lusis AJ, Hazen SL. 2015. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 163:1585–1595. doi:10.1016/j.cell.2015.11.055.
    1. Swann JR, Want EJ, Geier FM, Spagou K, Wilson ID, Sidaway JE, Nicholson JK, Holmes E. 2011. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc Natl Acad Sci U S A 108:4523–4530. doi:10.1073/pnas.1006734107.
    1. Begley M, Hill C, Gahan CGM. 2006. Bile salt hydrolase activity in probiotics. Appl Environ Microbiol 72:1729–1738. doi:10.1128/AEM.72.3.1729-1738.2006.
    1. Joyce SA, MacSharry J, Casey PG, Kinsella M, Murphy EF, Shanahan F, Hill C, Gahan CG. 2014. Regulation of host weight gain and lipid metabolism by bacterial bile acid modification in the gut. Proc Natl Acad Sci U S A 111:7421–7426. doi:10.1073/pnas.1323599111.
    1. Chen Q, Wang E, Ma L, Zhai P. 2012. Dietary resveratrol increases the expression of hepatic 7alpha-hydroxylase and ameliorates hypercholesterolemia in high-fat fed C57BL/6J mice. Lipids Health Dis 11:56. doi:10.1186/1476-511X-11-56.
    1. Chothe PP, Swaan PW. 2014. Resveratrol promotes degradation of the human bile acid transporter ASBT (SLC10A2). Biochem J 459:301–312. doi:10.1042/BJ20131428.
    1. De Aguiar Vallim TQ, Tarling EJ, Edwards PA. 2013. Pleiotropic roles of bile acids in metabolism. Cell Metab 17:657–669. doi:10.1016/j.cmet.2013.03.013.
    1. Sayin SI, Wahlström A, Felin J, Jäntti S, Marschall HU, Bamberg K, Angelin B, Hyötyläinen T, Orešič M, Bäckhed F. 2013. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab 17:225–235. doi:10.1016/j.cmet.2013.01.003.
    1. Liberles SD, Buck LB. 2006. A second class of chemosensory receptors in the olfactory epithelium. Nature 442:645–650. doi:10.1038/nature05066.
    1. Bai C, Biwersi J, Verkman AS, Matthay MA. 1998. A mouse model to test the in vivo efficacy of chemical chaperones. J Pharmacol Toxicol Methods 40:39–45. doi:10.1016/S1056-8719(98)00034-3.
    1. Mello CC, Barrick D. 2003. Measuring the stability of partly folded proteins using TMAO. Protein Sci 12:1522–1529. doi:10.1110/ps.0372903.

Source: PubMed

3
Abonneren