Skip to main content

GENERAL COMMENTARY article

Front. Immunol., 20 February 2019
Sec. Microbial Immunology
This article is part of the Research Topic Understanding and Exploiting Host-Commensal Interactions to Combat Pathogens View all 17 articles

Commentary: Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells

  • School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China

A Commentary on
Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells

by Ma, C., Han, M., Heinrich, B., Fu, Q., Zhang, Q., Sandhu, M., et al. (2018). Science 360:eaan5931. doi: 10.1126/science.aan5931

A study by Ma et al. (1) showed that gut microbiome composition in mice closely associates with liver cancer by influencing the immune system. This group provided evidence showing that changing commensal gut bacteria in mice affected the accumulation of hepatic CXCR6+ natural killer T (NKT) cells through mediation of CXCL16 expression in liver sinusoidal endothelial cells. CXCL16 is the only ligand for the chemokine receptor CXCR6, which mediates NKT cell survival and accumulation in the liver (2, 3). The accumulation of CXCR6 in hepatic NKT cells enhances the production of interferon-γ upon antigen stimulation, which contributes to the inhibition of tumor growth. The accumulation of NKT cells is known to be mainly regulated by a type of Clostridium species that metabolizes primary bile acids to secondary bile acids because depletion of Clostridium by vancomycin increases hepatic NKT cells and colonization of C. scindens induces a rapid decrease in liver NKT cells (1). This evidence highlighted the significant contribution of the gut microbiome to regulating anti-tumor immunity in liver and hepatic cancers.

Human microbiota plays a critical role in maintaining metabolic and immune homeostasis and protecting the host against pathogens (4, 5). The gut microbiota provides a prominent benefit to the host; however, there is also increasing evidence of the involvement of the gut microbiota in human disease (6). The liver is closely linked to the gut because of its anatomical connection via the portal vein. The liver is the first system to acquire nutrient-rich blood via a portal vein from the gastrointestinal tract. Accordingly, the liver is also the first target of metabolites from the gut microbiota, including bile acids, choline, short-chain fatty acids, indole derivatives, and lipopolysaccharides (7). Bile acids can be classified into primary bile acids and secondary bile acids, which are synthesized by the liver and by bacterial metabolism in the colon, respectively. Recently, emerging evidence has also indicated direct associations between obesity, gut microbiota, secondary bile acids, and hepatocellular carcinoma (HCC) (8, 9). Dietary obesity induces a clear expansion of gram-positive gut microbiota, especially Clostridium clusters XI and XVIa, in mice with a high-fat diet (8, 9). The elevation in the strains increased the levels of deoxycholic acid (DCA), a secondary bile acid, and lipoteichoic acid (LTA), a major cell wall component in gram-positive bacteria. The accumulation of the two molecules in the livers of HFD mice treated by chemical carcinogen cooperatively enhanced the Toll-like receptor 2 (TLR2)-mediated signals by the upregulation of the receptor, which induced overexpression of cyclooxygenase2 (COX2), catalyzing the production of prostaglandin E2 (PGE2). Accumulation of PGE2 suppressed anti-tumor immunity through a PTGER4 receptor on CD8 cells, thereby contributing to HCC progression (9).

Compared with previous studies, this study contributes to advances in the related field in the following ways. First, the results from Ma et al. (1) indicated that altering the gut microbiome caused the accumulation of both CD8 cells and NKT cells; however, depleting CD8 cells alone had minor effects on the tumor inhibition caused by elimination of commensal gut bacteria, and antibiotic treatment of tumor-bearing mice lacking NKT cells did not reduce liver tumor size. These results suggest that NKT cells are critical for effects on hepatic tumor growth induced by alterations in the gut microbiome. Second, Ma et al. (1) also provided evidence that increasing primary bile acids increased hepatic NKT cells and enhanced tumor inhibition but that increasing secondary bile acids had opposing effects. These analyses ascertained the beneficial effect of primary bile acids functioning as a regulator to enhance tumor inhibition. These findings indicate an axis of bile acids and CXCL16, CXCR6, and NKT cells that regulate live cancer. Third, DCA, a secondary bile acid, has always been speculated to be a promoter of liver cancer (10). This study showed that other secondary bile acids also played important roles. For example, ω-muricholic acid (ω-MCA) but not DCA decreased cxcl16 mRNA expression. Increasing ω-MCA expression by feeding was shown to inhibit the activation of liver sinusoidal endothelial cells. These findings present new knowledge of the function of different secondary bile acids.

The findings from Ma et al. shed light on the prevention and treatment of liver cancer by targeting the gut microbiota in clinical application. The data directly indicated that elimination of gram-positive bacteria by vancomycin from the gut prevents tumorigenesis (1). The data from the study also solidified the evidences of influence of liver health by diets, probiotics, and antibiotics, which affect the composition of the human gut microbiota. This research cautioned that Clostridium colonization in gut promotes tumor growth, on the other hand, the commensal Bifidobacterium can enhance antitumor immunity and regulate the therapy efficacy by blocking programmed cell death 1 ligand 1 (PD-L1) (11). Because there is individual variability in response to diets, endobiotics, and xenobiotics (12), the studies of precision editing of the gut microbiota are needed to prevent live cancer. Furthermore, the results from Ma et al. also raised the questions on the influence of gut microbiota on the monoclonal antibodies therapies by PD-L1 or PD-1 (programmed cell death 1) blockade. Nivolumab, an anti-PD-1 monoclonal antibody, has been approved by the FDA for liver cancer in 2017 (13). Recent studies showed that gut microbiomes modulate the efficacy of immunotherapies against melanoma and epithelial tumors (14, 15). On the basis of this study, further studies should be performed to assess the effect of gut microbiomes on the immunotherapies to cure liver cancer in clinical trials.

This study provided a comprehensive analysis of the relationship among the gut microbiome, the immune system, and liver cancer. However, this research invokes three related questions. First, the mechanism of bile acids regulating Cxcl16 expression is still unclear. CXCL16 is a small cytokine with a C-X-C motif with an O-glycosylated mucin-like stalk, a transmembrane helix and a cytoplasmic domain with a potential tyrosine phosphorylation site. These features allow CXCL16 to be expressed as a soluble chemokine as well as a cell surface-bound molecule (16). Further analysis should be performed to elicit if CXCL16 bind bile acids directly or through other molecules. Second, which secondary bile acids did contribute significantly to liver cancer? It has been reported that DCA induced liver cancer and nodules in rats in 1991 (17). Yoshimoto and Loo further showed that DCA was one of the factors facilitating liver cancer development (8, 9). The current research indicated that ω-MCA should be one of the critical players to promote liver cancer. ωMCA is a transformed from primary bile acid βMCA by three strains in a cooperative way, including one Eubacterium lentum strain and two Fusobacterium sp. strains (18). While DCA is transformed from cholic acid by Clostridium clusters XI and XVIa. Then it is critical to elucidate the contributions of different bacteria and secondary bile acids to promote live cancer. Third, can the findings be applied to humans? Approximately 1% of hepatic lymphocytes are NKT cells in humans; however, the cells constitute up to 40% of hepatic lymphocytes in mice. Promisingly, it was shown that primary bile acid CDCA levels in human samples were correlated with CXCL16 expression, whereas secondary bile acid glycolithocholate (GLCA) levels were inversely correlated. Furthermore, mucosal-associated invariant T (MAIT) cells, which are prevalent in human liver, can also express CXCR6 that can bind CXCL16. This evidence suggests that the current study could be translated into clinical practice. cancer. However, comprehensive analysis of human liver tissue is necessary for clinical application considering the differences between humans and mice.

Author Contributions

BJ wrote the commentary.

Conflict of Interest Statement

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu S, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science (2018) 360:eaan5931. doi: 10.1126/science.aan5931

CrossRef Full Text | Google Scholar

2. Geissmann F, Cameron TO, Sidobre S, Manlongat N, Kronenberg M, Briskin MJ, et al. Intravascular immune surveillance by CXCR6+ NKT cells patrolling liver sinusoids. PLoS Biol. (2005) 3:e113. doi: 10.1371/journal.pbio.0030113

CrossRef Full Text | Google Scholar

3. Shekhar S, Joyee AG, Yang X. Dynamics of NKT-cell responses to chlamydial infection. Front Immunol. (2015) 6:233. doi: 10.3389/fimmu.2015.00233

CrossRef Full Text | Google Scholar

4. Shekhar S, Schenck K, Petersen FC. Exploring host-commensal interactions in the respiratory tract. Front Immunol. (2017) 8:1971. doi: 10.3389/fimmu.2017.01971

CrossRef Full Text | Google Scholar

5. Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. (2017) 474:1823–36. doi: 10.1042/BCJ20160510

CrossRef Full Text | Google Scholar

6. Schroeder BO, Backhed F. Signals from the gut microbiota to distant organs in physiology and disease. Nat Med. (2016) 22:4185. doi: 10.1038/nm.4185

CrossRef Full Text | Google Scholar

7. Fu ZD, Cui JY. Remote sensing between liver and intestine: importance of microbial metabolites. Curr Pharmacol Rep. (2017) 3:101–13. doi: 10.1007/s40495-017-0087-0

CrossRef Full Text | Google Scholar

8. Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature (2013) 499:97–101. doi: 10.1038/nature12347

CrossRef Full Text | Google Scholar

9. Loo TM, Kamachi F, Watanabe Y, Yoshimoto S, Kanda H, Arai Y, et al. Gut microbiota promotes obesity-associated liver cancer through PGE2-mediated suppression of antitumor immunity. Cancer Discov. (2017) 7:522–38. doi: 10.1158/2159-8290.cd-16-0932

CrossRef Full Text | Google Scholar

10. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol. (2014) 30:332–8. doi: 10.1097/mog.0000000000000057

CrossRef Full Text | Google Scholar

11. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science (2015) 350:1084–9. doi: 10.1126/science.aac4255

CrossRef Full Text | Google Scholar

12. Jetten MJ, Claessen SM, Dejong CH, Lahoz A, Castell JV, Van Delft JH, et al. Interindividual variation in response to xenobiotic exposure established in precision-cut human liver slices. Toxicology (2014) 323:61–9. doi: 10.1016/j.tox.2014.06.007

CrossRef Full Text | Google Scholar

13. Finkelmeier F, Waidmann O, Trojan J. Nivolumab for the treatment of hepatocellular carcinoma. Expert Rev Anticancer Ther. (2018) 18:1169–75. doi: 10.1080/14737140.2018.1535315

CrossRef Full Text | Google Scholar

14. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiomemodulates response to anti-PD-1 immunotherapy in melanoma patients. Science (2018) 359:97–103. doi: 10.1126/science.aan4236

CrossRef Full Text | Google Scholar

15. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science (2018) 359:91–7. doi: 10.1126/science.aan3706

CrossRef Full Text | Google Scholar

16. Abel S, Hundhausen C, Mentlein R, Schulte A, Berkhout TA, Broadway N, et al. The transmembrane CXC-chemokine ligand 16 is induced by IFN-gamma and TNF-alpha and shed by the activity of the disintegrin-like metalloproteinase ADAM10. J Immunol. (2004) 172:6362–72. doi: 10.4049/jimmunol.172.10.6362

CrossRef Full Text | Google Scholar

17. Armstrong D, Cameron RG. Comparison of liver cancer and nodules induced in rats by deoxycholic acid diet with or without prior initiation. Cancer Lett. (1991) 57:153–7.

Google Scholar

18. Eyssen H, De Pauw G, Stragier J, Verhulst A. Cooperative formation of omega-muricholic acid by intestinal microorganisms. Appl Environ Microbiol. (1983) 45:141–7.

Google Scholar

Keywords: gut microbiome, bile acid, liver cancer, NKT cells, immune regulation

Citation: Jia B (2019) Commentary: Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells. Front. Immunol. 10:282. doi: 10.3389/fimmu.2019.00282

Received: 04 December 2018; Accepted: 04 February 2019;
Published: 20 February 2019.

Edited by:

Sudhanshu Shekhar, University of Oslo, Norway

Reviewed by:

Sunil Joshi, University of Miami, United States

Copyright © 2019 Jia. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Baolei Jia, baoleijia@cau.ac.kr

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.