Journal List > J Bacteriol Virol > v.49(4) > 1141089

Seo, Bae, Kim, Jeong, and Baek: Analysis of Intestinal Mucosal Microbiome Changes before and after Chemoradiation in Locally Advanced Rectal Cancer Patients

Abstract

Purpose

Dysbiosis of gut microbiota has been reported to participate in the pathogenesis of colorectal cancer, but changes in microbiota due to radiotherapy have not been studied. In this study, we tried to elucidate the changes in the microbiome in rectal cancer after chemoradiotherapy using RNA sequencing analysis.

Materials and methods

We included 11 pairs of human rectal cancer tissues before and after irradiation between August 2016 and December 2017 and performed RNA sequencing analysis. Mapped reads to human reference genomes were used for pair-wise transcriptome comparisons, and unmapped (non-human) reads were then mapped to bacterial marker genes using PathSeq.

Results

At microbiome level, interindividual variability of mucosal microbiota was greater than the change in microbial composition during radiotherapy. This indicates that rapid homeostatic recovery of the mucosal microbial composition takes place short after radiotherapy. At single microbe level, Prevotella and Fusobacterium, which were identified as important causative microbes of the initiation and progression of rectal cancer were decreased by radiotherapy. Moreover, changes in Prevotella were associated with changes in the human transcriptome of rectal cancer. We also found that there was a gene cluster that increased and decreased in association with changes in microbial composition by chemoradiation.

Conclusion

This study revealed changes in tumor-associated microbial community by irradiation in rectal cancer. These findings can be used to develop a new treatment strategy of neoadjuvant therapy for locally advanced rectal cancer by overcoming radio-resistance or facilitating radio-sensitivity.

Figures and Tables

Figure 1

Microbial composition before and after chemoradiation in rectal cancer patients. The relative abundance of OTU is shown for each taxonomic rank. The paired bar plots on the right show the microbial composition before and after chemoradiation in the same patients. The numbers on the bar plot represent the patient identification numbers. The skyline plots on the left show the bar plot in ascending order based on the most abundant OTU. The y-axis of both charts indicates the relative abundance (%). “PRE” and “PRERT” represent samples before chemoradiation therapy. “POST” and “POSTRT” represent samples after chemoradiation therapy. OTUs that differ in ANCOM analysis are marked with an asterisk and the W-statistics are shown in parentheses. (A) Phylum (B) Class (C) Order and (D) Family.

jbv-49-162-g001
Figure 2

Principal component analysis of the microbiota before and after chemoradiation in rectal cancer patients. The numbers represent the patient identification numbers. “PRERT” and “POSTRT” represent samples before and after chemoradiation therapy, respectively. (A) Phylum (B) Class (C) Order (D) Family.

jbv-49-162-g002
Figure 3

Changes in the abundance of Prevotellaceae family after chemoradiation and concomitant changes in the human transcriptome. (A) Box plot shows the relative abundance of Prevotellaceae before and after chemoradiotherapy. “PRERT” and “POSTRT” represent samples before and after chemoradiation therapy, respectively. (B) Bubble chart shows the functional annotation of host gene expression highly correlated with changes in relative abundance of Prevotellaceae before and after chemoradiotherapy. Gene-Enrichment and Functional Annotation Analysis was performed using DAVID. The y-axis represents the functional term defined in GOTERM_MF_DIRECT. The numbers in parentheses are the identification numbers of the Gene Ontology. Fold enrichment, ratio of the number of genes involved the function term to the total number of inputs genes, and p-value are represented by x-axis, bubble size, and bubble color, respectively.

jbv-49-162-g003
Table 1

Patient characteristics

jbv-49-162-i001

IDa: patient identification number, BMIb: body mass index, Diffc: differentiation (1 = well and moderately differentiated, 2 = poorly differentiated), Budd: presence of tumor budding, PNIe: presence of perineural invasion, ETDf: presence of extranodal tumor deposit, MSIg: microsatellite instability (H: high, L: low), KRASh: presence of KRAS exon 2 mutation, BRAFi: presence of BRAF mutation, TRGj: Mandard tumor regression grade (NR = non-responder, R = responder), pTk: primary tumor categories, pNl: regional lymph node categories.

Table 2

Read count statistics

jbv-49-162-i002

The numbers in parentheses, excluding the “PathSeq mapped” column, represent the percentage of read count in the previous step. The number in parentheses in the “PathSeq mapped” column is the percentage of non-human read counts which unmapped to hg38. IDa: sample identification number (The number before hyphen indicates patient identification number. “PRE” and “POST” represent samples before and after chemoradiation therapy.), Rawb: raw read counts, Trimmedc: read counts after quality control. STAR hg38 mappedd: read counts mapped to hg38 human reference genome using STAR aligner. PathSeq mappede: read counts mapped to bacterial marker genes using PathSeq.

Table 3

PERMANOVA results testing the effect of chemoradiation on microbial composition

jbv-49-162-i003

Three distance metrics were used to test the differences in microbial composition before and after chemoradiation therapy in rectal adenocarcinoma patients. PERMANOVAa: permutational multivariate analysis of variance.

Table 4

ANCOM analysis between before and after chemoradiotherapy

jbv-49-162-i004

ANCOM: Analysis of composition of microbiomes, Wa: ANCOM W-statistics, Nfeat b: Total number of OTUs detected within the same taxonomic rank, D_0.9c~D_0.6f: ANCOM significance with cut-offs from 0.9 to 0.6.

Table 5

Top 10 genes showing the strongest positive correlation with the abundance of Prevotellaceae

jbv-49-162-i005
Table 6

Top 10 genes showing the strongest negative correlation with the abundance of Prevotellaceae

jbv-49-162-i006

Notes

No potential conflict of interest relevant to this article was reported.

References

1. Swedish Rectal Cancer Trial. Improved survival with preoperative radiotherapy in resectable rectal cancer. N Engl J Med. 1997; 336:980–987.
2. Kapiteijn E, Marijnen CA, Nagtegaal ID, Putter H, Steup WH, Wiggers T, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med. 2001; 345:638–646.
crossref
3. Bosset JF, Collette L, Calais G, Mineur L, Maingon P, Radosevic-Jelic L, et al. Chemotherapy with preoperative radiotherapy in rectal cancer. N Engl J Med. 2006; 355:1114–1123.
crossref
4. Han YD, Kim WR, Park SW, Cho MS, Hur H, Min BS, et al. Predictors of Pathologic Complete Response in Rectal Cancer Patients Undergoing Total Mesorectal Excision After Preoperative Chemoradiation. Medicine (Baltimore). 2015; 94:e1971.
crossref
5. Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004; 58:862–870.
crossref
6. Vatner RE, Cooper BT, Vanpouille-Box C, Demaria S, Formenti SC. Combinations of immunotherapy and radiation in cancer therapy. Front Oncol. 2014; 4:325.
crossref
7. Tuyaerts S, Van Nuffel AMT, Naert E, Van Dam PA, Vuylsteke P, De Caluwé A, et al. PRIMMO study protocol: a phase II study combining PD-1 blockade, radiation and immunomodulation to tackle cervical and uterine cancer. BMC Cancer. 2019; 19:506.
crossref
8. Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer. 2017; 17:271–285.
crossref
9. Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013; 342:967–970.
crossref
10. 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; 350:1084–1089.
crossref
11. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014; 30:2114–2120.
crossref
12. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013; 29:15–21.
crossref
13. Kostic AD, Ojesina AI, Pedamallu CS, Jung J, Verhaak RG, Getz G, et al. PathSeq: software to identify or discover microbes by deep sequencing of human tissue. Nat Biotechnol. 2011; 29:393–396.
crossref
14. Mandal S, Van Treuren W, White RA, Eggesbo M, Knight R, Peddada SD. Analysis of composition of microbiomes: a novel method for studying microbial composition. Microb Ecol Health Dis. 2015; 26:27663.
crossref
15. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15:550.
crossref
16. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009; 4:44–57.
crossref
17. Team RC. R: A language and environment for statistical computing. . Vienna, Austria: R Foundation for Statstical Computing;2018.
18. Wickham H. Ggplot2: elegant graphics for data analysis. New York: Springer-Verlag;2016.
19. Dove WF, Clipson L, Gould KA, Luongo C, Marshall DJ, Moser AR, et al. Intestinal neoplasia in the ApcMin mouse: independence from the microbial and natural killer (beige locus) status. Cancer Res. 1997; 57:812–814.
20. Cuevas-Ramos G, Petit CR, Marcq I, Boury M, Oswald E, Nougayrède JP. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc Natl Acad Sci U S A. 2010; 107:11537–11542.
crossref
21. Toprak NU, Yagci A, Gulluoglu BM, Akin ML, Demirkalem P, Celenk T, et al. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin Microbiol Infect. 2006; 12:782–786.
crossref
22. Gao Z, Guo B, Gao R, Zhu Q, Qin H. Microbiota disbiosis is associated with colorectal cancer. Front Microbiol. 2015; 6:20.
crossref
23. Wu N, Yang X, Zhang R, Li J, Xiao X, Hu Y, et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol. 2013; 66:462–470.
crossref
24. Tjalsma H, Boleij A, Marchesi JR, Dutilh BE. A bacterial driver-passenger model for colorectal cancer: beyond the usual suspects. Nat Rev Microbiol. 2012; 10:575–582.
crossref
25. Hajishengallis G, Darveau RP, Curtis MA. The keystone-pathogen hypothesis. Nat Rev Microbiol. 2012; 10:717–725.
crossref
26. Li S, Konstantinov SR, Smits R, Peppelenbosch MP. Bacterial Biofilms in Colorectal Cancer Initiation and Progression. Trends Mol Med. 2017; 23:18–30.
crossref
27. Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015; 350:1079–1084.
crossref
28. Kim YS, Kim J, Park SJ. High-throughput 16S rRNA gene sequencing reveals alterations of mouse intestinal microbiota after radiotherapy. Anaerobe. 2015; 33:1–7.
crossref
29. Gerassy-Vainberg S, Blatt A, Danin-Poleg Y, Gershovich K, Sabo E, Nevelsky A, et al. Radiation induces proinflammatory dysbiosis: transmission of inflammatory susceptibility by host cytokine induction. Gut. 2018; 67:97–107.
crossref
30. Wang A, Ling Z, Yang Z, Kiela PR, Wang T, Wang C, et al. Gut microbial dysbiosis may predict diarrhea and fatigue in patients undergoing pelvic cancer radiotherapy: a pilot study. PLoS One. 2015; 10:e0126312.
crossref
31. Nam YD, Kim HJ, Seo JG, Kang SW, Bae JW. Impact of pelvic radiotherapy on gut microbiota of gynecological cancer patients revealed by massive pyrosequencing. PLoS One. 2013; 8:e82659.
crossref
32. Manichanh C, Varela E, Martinez C, Antolin M, Llopis M, Doré J, et al. The gut microbiota predispose to the pathophysiology of acute postradiotherapy diarrhea. Am J Gastroenterol. 2008; 103:1754–1761.
crossref
33. Flemer B, Lynch DB, Brown JM, Jeffery IB, Ryan FJ, Claesson MJ, et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut. 2017; 66:633–643.
crossref
34. Zoetendal EG, von Wright A, Vilpponen-Salmela T, Ben-Amor K, Akkermans AD, de Vos WM. Mucosa-associated bacteriain the human gastrointestinal tract are uniformly distributed along the colon and differ from the community recovered from feces. Appl Environ Microbiol. 2002; 68:3401–3407.
crossref
35. Ranawat P, Rawat S. Radiation resistance in thermophiles: mechanisms and applications. World J Microbiol Biotechnol. 2017; 33:112.
crossref
36. Palleja A, Mikkelsen KH, Forslund SK, Kashani A, Allin KH, Nielsen T, et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat Microbiol. 2018; 3:1255–1265.
crossref
37. Jalanka J, Salonen A, Salojärvi J, Ritari J, Immonen O, Marciani L, et al. Effects of bowel cleansing on the intestinal microbiota. Gut. 2015; 64:1562–1568.
crossref
38. Sinha R, Ahn J, Sampson JN, Shi J, Yu G, Xiong X, et al. Fecal Microbiota, Fecal Metabolome, and Colorectal Cancer Interrelations. PLoS One. 2016; 11:e0152126.
crossref
39. Xu K, Jiang B. Analysis of Mucosa-Associated Microbiota in Colorectal Cancer. Med Sci Monit. 2017; 23:4422–4430.
crossref
40. Vogtmann E, Hua X, Zeller G, Sunagawa S, Voigt AY, Hercog R, et al. Colorectal Cancer and the Human Gut Microbiome: Reproducibility with Whole-Genome Shotgun Sequencing. PLoS One. 2016; 11:e0155362.
crossref
41. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013; 14:195–206.
crossref
42. Mima K, Sukawa Y, Nishihara R, Qian ZR, Yamauchi M, Inamura K, et al. Fusobacterium nucleatum and T Cells in Colorectal Carcinoma. JAMA Oncol. 2015; 1:653–661.
crossref
43. Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, et al. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One. 2011; 6:e16393.
crossref
44. Wu D, Wu P, Huang Q, Liu Y, Ye J, Huang J. Interleukin-17: a promoter in colorectal cancer progression. Clin Dev Immunol. 2013; 2013:436307.
crossref
45. Liu J, Duan Y, Cheng X, Chen X, Xie W, Long H, et al. IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun. 2011; 407:348–354.
crossref
46. Wang K. Targeting IL-17 for cancer-associated inflammation and immunity. J Immunol. 2017; 198:66.5.
47. Crawford PA, Gordon JI. Microbial regulation of intestinal radiosensitivity. Proc Natl Acad Sci U S A. 2005; 102:13254–13259.
crossref
48. Ferreira MR, Muls A, Dearnaley DP, Andreyev HJ. Microbiota and radiation-induced bowel toxicity: lessons from inflammatory bowel disease for the radiation oncologist. Lancet Oncol. 2014; 15:e139–e147.
crossref
49. Cui M, Xiao H, Li Y, Zhou L, Zhao S, Luo D, et al. Faecal microbiota transplantation protects against radiation-induced toxicity. EMBO Mol Med. 2017; 9:448–461.
crossref
50. Nejdfors P, Ekelund M, Weström BR, Willén R, Jeppsson B. Intestinal permeability in humans is increased after radiation therapy. Dis Colon Rectum. 2000; 43:1582–1587. discussion 1587-8.
crossref
51. Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest. 2007; 117:2197–2204.
crossref
52. Goubet AG, Daillère R, Routy B, Derosa L, M Roberti P, Zitvogel L. The impact of the intestinal microbiota in therapeutic responses against cancer. C R Biol. 2018; 341:284–289.
crossref
TOOLS
Similar articles