Influence of gastrectomy for gastric cancer treatment on faecal microbiome and metabolome profiles

Pande Putu Erawijantari, Sayaka Mizutani, Hirotsugu Shiroma, Satoshi Shiba, Takeshi Nakajima, Taku Sakamoto, Yutaka Saito, Shinji Fukuda, Shinichi Yachida, Takuji Yamada, Pande Putu Erawijantari, Sayaka Mizutani, Hirotsugu Shiroma, Satoshi Shiba, Takeshi Nakajima, Taku Sakamoto, Yutaka Saito, Shinji Fukuda, Shinichi Yachida, Takuji Yamada

Abstract

Objective: Recent evidence points to the gut microbiome's involvement in postoperative outcomes, including after gastrectomy. Here, we investigated the influence of gastrectomy for gastric cancer on the gut microbiome and metabolome, and how it related to postgastrectomy conditions.

Design: We performed shotgun metagenomics sequencing and capillary electrophoresis time-of-flight mass spectrometry-based metabolomics analyses on faecal samples collected from participants with a history of gastrectomy for gastric cancer (n=50) and compared them with control participants (n=56).

Results: The gut microbiota in the gastrectomy group showed higher species diversity and richness (p<0.05), together with greater abundance of aerobes, facultative anaerobes and oral microbes. Moreover, bile acids such as genotoxic deoxycholic acid and branched-chain amino acids were differentially abundant between the two groups (linear discriminant analysis (LDA) effect size (LEfSe): p<0.05, q<0.1, LDA>2.0), as were also Kyoto Encyclopedia of Genes and Genomes modules involved in nutrient transport and organic compounds biosynthesis (LEfSe: p<0.05, q<0.1, LDA>2.0).

Conclusion: Our results reveal alterations of gut microbiota after gastrectomy, suggesting its association with postoperative comorbidities. The multi-omic approach applied in this study could complement the follow-up of patients after gastrectomy.

Keywords: gastrectomy; gastric cancer; gutmicrobiome; metabolome; metagenome.

Conflict of interest statement

Competing interests: SF and TY are founders of Metabologenomics. The company is focused on the design and control of the gut environment for human health. The company had no control over the interpretation, writing or publication of this work. The terms of these arrangements are being managed by Keio University and Tokyo Institute of Technology in accordance with its conflict of interest policies.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Community structure of the faecal microbiome and metabolome in postgastrectomy and control participants. Principal coordinates analysis (PCoA) with Bray-Curtis distance was performed to assess the community structure of species’ relative abundance obtained by mOTU (A) and MetaPhlAn2 (B) in the gastrectomy group (n=50) (orange) and in the control group (n=56) (blue). The PCoA trend was confirmed by significantly lower microbial structure dissimilarity (Bray-Curtis) within groups (p=5.53×10−7) (C). PCoA was performed also on faecal metabolite concentrations in the gastrectomy group (n=44) (orange) and the control group (n=54) (blue) (D).
Figure 2
Figure 2
Microbiome diversity in gastrectomy and control groups. Species richness was measured using the Chao1 index calculated from the species annotated by mOTU (A) and MetaPhlAn2 (B). Species alpha-diversity was measured using the Shannon-Wiener index based on mOTU (C) and MetaPhlAn2 (D) annotation. Species alpha-diversity was measured for each major phylum using mOTU (E) and MetaPhlAn2 (F) annotation.
Figure 3
Figure 3
Differential enrichment of microbes in gastrectomy and control groups. Cladogram of species annotated by mOTU (A) and MetaPhlAn2 (B). Each dot represents a taxonomic hierarchy. Dots are marked for significant (LEfSe: p3.0 are highlighted and labelled accordingly. The summed relative abundances of oral microbes were compared between the gastrectomy (n=50) and control (n=56) groups based on the species annotated by mOTU (C) and MetaPhlAn2 (D). The summed relative abundances of aerobes (E) and facultative anaerobes (F) were compared between the two groups.
Figure 4
Figure 4
Different trends of functional modules and metabolites from the faecal microbiomes of gastrectomy and control groups. Relative abundance and linear discriminant analysis (LDA) score (log10) of Kyoto Encyclopedia of Genes and Genomes (KEGG) modules annotated by the Human Microbiome Project Unified Metabolic Analysis Network 2 (HUMAnN2) and overlapping with those annotated by our in-house pipeline (linear discriminant analysis effect size (LEfSe): p<0.05, q<0.1, LDA>2.0) (A). Richness (Chao1) and alpha-diversity (Shannon-Wiener) of contributor species were estimated. Three modules (marked by an asterisk (*) in A) contributed by significantly more diverse and richer microbes (two-sided Mann-Whitney U test (MWU): p<0.001) in the gastrectomy group, in spite of their enrichment in the control group (B). The modules were M00196, M00535 and M00122 (two-sided MWU: p=5.31×10−4, 3.17×10−5, 7.14×10−6, for richness, respectively, and p=1.12×10−4, 2.22×10−4, 6.10×10−4, for alpha-diversity, respectively). KEGG modules’ relative abundance is represented by the top value of each stack of bars. Samples were subsequently sorted according to the dominant contributor to a module and then grouped as either gastrectomy or control (sample order differs between panels). Different trends in metabolites were also observed between the two groups (LEfSe: p<0.05, q<0.1, LDA>3.0) (C). Bile acid reaction consisted of deconjugation of conjugated bile acids (glycocholate and taurocholate) into their primary form (cholate) and amino acids (glycine and taurine) followed by 7-α/β-dehydroxylation to form secondary bile acids (deoxycholic acid (DCA)). The colours highlight enrichment in the control (blue) and gastrectomy (orange) groups (D).
Figure 5
Figure 5
Genus-genus and genus-metabolite correlations. Co-occurrence (red) and co-excluding (green) relationships between genera (SparCC: −0.22.0) and metabolites (LEfSe: p3.0) from gastrectomy (n=44) and control (n=54) participants (C). The matrices were derived from Euclidean distance-based bi-clustering of Spearman’s RANK correlation matrices. Correlation coefficients in each square represent positive (red) and negative (blue) relationships. Colours are proportional to the absolute value of Spearman’s RANK correlations (see legend in the figure). Statistically significant correlations (p

Figure 6

Data integration-derived schematic hypothesis. Schematic…

Figure 6

Data integration-derived schematic hypothesis. Schematic hypothesis of gut microbiome and metabolite alterations after…

Figure 6
Data integration-derived schematic hypothesis. Schematic hypothesis of gut microbiome and metabolite alterations after gastrectomy. The scheme is divided into three parts denoting physiological changes (grey), findings from our study (blue) and the possible consequences (red). Solid lines show the links confirmed by previous studies. Dashed lines show possible connections that can be inferred from our findings. The higher or lower levels observed in postgastrectomy patients are shown in comparison with those of control participants. BCAA, branched-chain amino acids.
Figure 6
Figure 6
Data integration-derived schematic hypothesis. Schematic hypothesis of gut microbiome and metabolite alterations after gastrectomy. The scheme is divided into three parts denoting physiological changes (grey), findings from our study (blue) and the possible consequences (red). Solid lines show the links confirmed by previous studies. Dashed lines show possible connections that can be inferred from our findings. The higher or lower levels observed in postgastrectomy patients are shown in comparison with those of control participants. BCAA, branched-chain amino acids.

References

    1. Guyton K, Alverdy JC. The gut microbiota and gastrointestinal surgery. Nat Rev Gastroenterol Hepatol 2017;14:43–54. 10.1038/nrgastro.2016.139
    1. Lederer A-K, Pisarski P, Kousoulas L, et al. . Postoperative changes of the microbiome: are surgical complications related to the gut flora? A systematic review. BMC Surg 2017;17:125 10.1186/s12893-017-0325-8
    1. Gutiérrez-Repiso C, Moreno-Indias I, de Hollanda A, et al. . Gut microbiota specific signatures are related to the successful rate of bariatric surgery. Am J Transl Res 2019;11:942–52.
    1. Mondot S, Lepage P, Seksik P, et al. . Structural robustness of the gut mucosal microbiota is associated with Crohn's disease remission after surgery. Gut 2016;65:954–62. 10.1136/gutjnl-2015-309184
    1. Bennett JMH, Mehta S, Rhodes M. Surgery for morbid obesity. Postgrad Med J 2007;83:8–15. 10.1136/pgmj.2006.048868
    1. Penna M, Allum W. New treatments for gastric cancer: are they changing clinical practice? Clinical Practice 2013;10:649–59. 10.2217/cpr.13.56
    1. Zhang H, DiBaise JK, Zuccolo A, et al. . Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci U S A 2009;106:2365–70. 10.1073/pnas.0812600106
    1. Graessler J, Qin Y, Zhong H, et al. . Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: correlation with inflammatory and metabolic parameters. Pharmacogenomics J 2013;13:514–22. 10.1038/tpj.2012.43
    1. Palleja A, Kashani A, Allin KH, et al. . Roux-En-Y gastric bypass surgery of morbidly obese patients induces swift and persistent changes of the individual gut microbiota. Genome Med 2016;8:67 10.1186/s13073-016-0312-1
    1. Aron-Wisnewsky J, Prifti E, Belda E, et al. . Major microbiota dysbiosis in severe obesity: fate after bariatric surgery. Gut 2019;68:70–82. 10.1136/gutjnl-2018-316103
    1. Tseng C-H, Lin J-T, Ho HJ, et al. . Gastric microbiota and predicted gene functions are altered after subtotal gastrectomy in patients with gastric cancer. Sci Rep 2016;6 10.1038/srep20701
    1. Lin X-H, Huang K-H, Chuang W-H, et al. . The long term effect of metabolic profile and microbiota status in early gastric cancer patients after subtotal gastrectomy. PLoS One 2018;13:e0206930 10.1371/journal.pone.0206930
    1. Eom BW, Lee H-J, Yoo M-W, et al. . Synchronous and metachronous cancers in patients with gastric cancer. J Surg Oncol 2008;98:106–10. 10.1002/jso.21027
    1. Ikeda Y, Saku M, Kawanaka H, et al. . Features of second primary cancer in patients with gastric cancer. Oncology 2003;65:113–7. 10.1159/000072335
    1. Yachida S, Mizutani S, Shiroma H, et al. . Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med 2019;25:968–76. 10.1038/s41591-019-0458-7
    1. Wirbel J, Pyl PT, Kartal E, et al. . Meta-Analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med 2019;25:679–89. 10.1038/s41591-019-0406-6
    1. Thomas AM, Manghi P, Asnicar F, et al. . Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat Med 2019;25:667–78. 10.1038/s41591-019-0405-7
    1. Nishimoto Y, Mizutani S, Nakajima T, et al. . High stability of faecal microbiome composition in guanidine thiocyanate solution at room temperature and robustness during colonoscopy. Gut 2016;65:1574–5. 10.1136/gutjnl-2016-311937
    1. Shin S, Saito E, Sawada N, et al. . Dietary patterns and colorectal cancer risk in middle-aged adults: A large population-based prospective cohort study. Clinical Nutrition 2018;37:1019–26. 10.1016/j.clnu.2017.04.015
    1. Soga T, Ohashi Y, Ueno Y, et al. . Quantitative metabolome analysis using capillary electrophoresis mass spectrometry. J Proteome Res 2003;2:488–94. 10.1021/pr034020m
    1. Sunagawa S, Mende DR, Zeller G, et al. . Metagenomic species profiling using universal phylogenetic marker genes. Nat Methods 2013;10:1196–9. 10.1038/nmeth.2693
    1. Truong DT, Franzosa EA, Tickle TL, et al. . MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods 2015;12:902–3. 10.1038/nmeth.3589
    1. Chen T, Yu W-H, Izard J, et al. . The human oral microbiome database: a web accessible resource for investigating oral microbe taxonomic and genomic information. Database 2010;2010:baq013 10.1093/database/baq013
    1. Langille MGI, Zaneveld J, Caporaso JG, et al. . Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol 2013;31:814–21. 10.1038/nbt.2676
    1. Franzosa EA, McIver LJ, Rahnavard G, et al. . Species-Level functional profiling of metagenomes and metatranscriptomes. Nat Methods 2018;15:962–8. 10.1038/s41592-018-0176-y
    1. Kanehisa M, Goto S. Kegg: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res 2000;28:27–30. 10.1093/nar/28.1.27
    1. Segata N, Izard J, Waldron L, et al. . Metagenomic biomarker discovery and explanation. Genome Biol 2011;12:R60 10.1186/gb-2011-12-6-r60
    1. Morgan XC, Tickle TL, Sokol H, et al. . Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012;13:R79 10.1186/gb-2012-13-9-r79
    1. Friedman J, Alm EJ. Inferring correlation networks from genomic survey data. PLoS Comput Biol 2012;8:e1002687 10.1371/journal.pcbi.1002687
    1. Noecker C, Eng A, Srinivasan S, et al. . Metabolic model-based integration of microbiome taxonomic and metabolomic profiles elucidates mechanistic links between ecological and metabolic variation. mSystems 2016;1 10.1128/mSystems.00013-15
    1. Radigan AE. Post-Gastrectomy: managing the nutrition fallout. Pract Gastroenterol 2004;28:63–79.
    1. Ilhan ZE, DiBaise JK, Isern NG, et al. . Distinctive microbiomes and metabolites linked with weight loss after gastric bypass, but not gastric banding. Isme J 2017;11:2047–58. 10.1038/ismej.2017.71
    1. Kong L-C, Tap J, Aron-Wisnewsky J, et al. . Gut microbiota after gastric bypass in human obesity: increased richness and associations of bacterial genera with adipose tissue genes. Am J Clin Nutr 2013;98:16–24. 10.3945/ajcn.113.058743
    1. Röttjers L, Faust K. From hairballs to hypotheses–biological insights from microbial networks. FEMS Microbiol Rev 2018;42:761–80. 10.1093/femsre/fuy030
    1. Dai Z-L, Wu G, Zhu W-Y. Amino acid metabolism in intestinal bacteria: links between gut ecology and host health. Front Biosci 2011;16:1768–86. 10.2741/3820
    1. Aron-Wisnewsky J, Doré J, Clement K. The importance of the gut microbiota after bariatric surgery. Nat Rev Gastroenterol Hepatol 2012;9:590–8. 10.1038/nrgastro.2012.161
    1. Quercia I, Dutia R, Kotler DP, et al. . Gastrointestinal changes after bariatric surgery. Diabetes Metab 2014;40:87–94. 10.1016/j.diabet.2013.11.003
    1. Celiker H. A new proposed mechanism of action for gastric bypass surgery: air hypothesis. Med Hypotheses 2017;107:81–9. 10.1016/j.mehy.2017.08.012
    1. Lim J, Yoo M-W, Kang SY, et al. . Long-Term changes in the metabolic and nutritional parameters after gastrectomy in early gastric cancer patients with overweight. Asian J Surg 2019;42:386–93. 10.1016/j.asjsur.2018.06.011
    1. Jun J-H, Yoo JE, Lee JA, et al. . Anemia after gastrectomy in long-term survivors of gastric cancer: a retrospective cohort study. International Journal of Surgery 2016;28:162–8. 10.1016/j.ijsu.2016.02.084
    1. Degnan PH, Taga ME, Goodman AL. Vitamin B 12 as a modulator of gut microbial ecology. Cell Metab 2014;20:769–78. 10.1016/j.cmet.2014.10.002
    1. Liu H, Hu C, Zhang X, et al. . Role of gut microbiota, bile acids and their cross-talk in the effects of bariatric surgery on obesity and type 2 diabetes. J Diabetes Investig 2018;9:13–20. 10.1111/jdi.12687
    1. Ridlon JM, Kang D-J, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006;47:241–59. 10.1194/jlr.R500013-JLR200
    1. Yoshimoto S, Loo TM, Atarashi K, et al. . Obesity-Induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013;499:97–101. 10.1038/nature12347
    1. Ajouz H, Mukherji D, Shamseddine A. Secondary bile acids: an underrecognized cause of colon cancer. World J Surg Oncol 2014;12:164 10.1186/1477-7819-12-164
    1. Hull MA, Markar SR, Morris EJA. Cancer risk after bariatric surgery — is colorectal cancer a special case? Nat Rev Gastroenterol Hepatol 2018;15:653–4. 10.1038/s41575-018-0070-1
    1. Mackenzie H, Markar SR, Askari A, et al. . Obesity surgery and risk of cancer. Br J Surg 2018;105:1650–7. 10.1002/bjs.10914
    1. Rubinstein MR, Wang X, Liu W, et al. . Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-Cadherin/β-Catenin signaling via its FadA adhesin. Cell Host Microbe 2013;14:195–206. 10.1016/j.chom.2013.07.012
    1. Kostic AD, Chun E, Robertson L, et al. . Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013;14:207–15. 10.1016/j.chom.2013.07.007
    1. Kasai C, Sugimoto K, Moritani I, et al. . Comparison of human gut microbiota in control subjects and patients with colorectal carcinoma in adenoma: terminal restriction fragment length polymorphism and next-generation sequencing analyses. Oncol Rep 2016;35:325–33. 10.3892/or.2015.4398
    1. Yu J, Feng Q, Wong SH, et al. . Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut 2017;66:70–8. 10.1136/gutjnl-2015-309800
    1. Chan AW, et al. Potential role of metabolomics in diagnosis and surveillance of gastric cancer. WJG 2014;20:12874–82. 10.3748/wjg.v20.i36.12874
    1. Denkert C, Budczies J, Weichert W, et al. . Metabolite profiling of human colon carcinoma – deregulation of TCA cycle and amino acid turnover. Mol Cancer 2008;7:72 10.1186/1476-4598-7-72
    1. Le Chatelier E, Nielsen T, Qin J, et al. . Richness of human gut microbiome correlates with metabolic markers. Nature 2013;500:541–6. 10.1038/nature12506
    1. Shan B, Shan L, Morris D, et al. . Systematic review on quality of life outcomes after gastrectomy for gastric carcinoma. J Gastrointest Oncol 2015;6:544–60.

Source: PubMed

3
구독하다