DOI QR코드

DOI QR Code

Microbiota of Breast Tissue and Its Potential Association with Regional Recurrence of Breast Cancer in Korean Women

  • Kim, Hyo-Eun (Department of Life Science, Multidisciplinary Genome Institute, Hallym University) ;
  • Kim, Jongjin (Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center) ;
  • Maeng, Sejung (Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center) ;
  • Oh, Bumjo (Department of Family Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center) ;
  • Hwang, Ki-Tae (Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center) ;
  • Kim, Bong-Soo (Department of Life Science, Multidisciplinary Genome Institute, Hallym University)
  • Received : 2021.06.14
  • Accepted : 2021.09.23
  • Published : 2021.12.28

Abstract

Recent studies have reported dysbiosis of the microbiome in breast tissue collected from patients with breast cancer and the association between the microbiota and disease progression. However, the role of the microbiota in breast tissue remains unclear, possibly due to the complexity of breast cancer and various factors, including racial and geographical differences, influencing microbiota in breast tissue. Here, to determine the potential role of microbiota in breast tumor tissue, we analyzed 141 tissue samples based on three different tissue types (tumor, adjacent normal, and lymph node tissues) from the same patients with breast cancer in Korea. The microbiota was not simply distinguishable based on tissue types. However, the microbiota could be divided into two cluster types, even within the same tissue type, and the clinicopathologic factors were differently correlated in the two cluster types. Risk of regional recurrence was also significantly different between the microbiota cluster types (p = 0.014). In predicted function analysis, the pentose and glucuronate interconversions were significantly different between the cluster types (q < 0.001), and Enterococcus was the main genus contributing to these differences (q < 0.01). Results showed that the microbiota of breast tissue could interact with the host and influence the risk of regional recurrence. Although further studies would be recommended to validate our results, this study could expand our understanding on the breast tissue microbiota, and the results might be applied to develop novel prediction methods and treatments for patients with breast cancer.

Keywords

Acknowledgement

This study was supported by Seoul National University Hospital Research Fund [grant number 05-2017-0030 (2017-1048)] and Hallym University Research Fund, 2020 (HRF-202004-012). The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

References

  1. Siegel RL, Miller KD, Jemal A. 2019. Cancer statistics, 2019. CA Cancer J. Clin. 69: 7-34. https://doi.org/10.3322/caac.21551
  2. Barnes BB, Steindorf K, Hein R, Flesch-Janys D, Chang-Claude J. 2011. Population attributable risk of invasive postmenopausal breast cancer and breast cancer subtypes for modifiable and non-modifiable risk factors. Cancer Epidemiol. 35: 345-352. https://doi.org/10.1016/j.canep.2010.11.003
  3. Sampson JN, Falk RT, Schairer C, Moore SC, Fuhrman BJ, Dallal CM, et al. 2017. Association of estrogen metabolism with breast cancer risk in different cohorts of postmenopausal women. Cancer Res. 77: 918-925. https://doi.org/10.1158/0008-5472.CAN-16-1717
  4. Qiu J, Du Z, Liu J, Zhou Y, Liang F, Lu Q. 2018. Association between polymorphisms in estrogen metabolism genes and breast cancer development in Chinese women: A prospective case-control study. Medicine (Baltimore) 97: e13337. https://doi.org/10.1097/md.0000000000013337
  5. Human Microbiome Project C. 2012. Structure, function and diversity of the healthy human microbiome. Nature 486: 207-214. https://doi.org/10.1038/nature11234
  6. Selber-Hnatiw S, Rukundo B, Ahmadi M, Akoubi H, Al-Bizri H, Aliu AF, et al. 2017. Human gut microbiota: Toward an ecology of disease. Front. Microbiol. 8: 1265. https://doi.org/10.3389/fmicb.2017.01265
  7. Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, et al. 2018. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 360: eaan5931. https://doi.org/10.1126/science.aan5931
  8. Yan X, Yang M, Liu J, Gao R, Hu J, Li J, et al. 2015. Discovery and validation of potential bacterial biomarkers for lung cancer. Am. J. Cancer Res. 5: 3111-3122.
  9. Yu Y, Champer J, Beynet D, Kim J, Friedman AJ. 2015. The role of the cutaneous microbiome in skin cancer: lessons learned from the gut. J. Drugs Dermatol. 14: 461-465.
  10. DeStefano Shields CE, Van Meerbeke SW, Housseau F, Wang H, Huso DL, Casero RA, Jr., et al. 2016. Reduction of murine colon tumorigenesis driven by enterotoxigenic Bacteroides fragilis using cefoxitin treatment. J. Infect. Dis. 214: 122-129. https://doi.org/10.1093/infdis/jiw069
  11. Goedert JJ, Jones G, Hua X, Xu X, Yu G, Flores R, et al. 2015. Investigation of the association between the fecal microbiota and breast cancer in postmenopausal women: a population-based case-control pilot study. J. Natl. Cancer Inst. 107: djv147.
  12. Xuan C, Shamonki JM, Chung A, DiNome ML, Chung M, Sieling PA, et al. 2014. Microbial dysbiosis is associated with human breast cancer. PLoS One 9: e83744. https://doi.org/10.1371/journal.pone.0083744
  13. Kwa M, Plottel CS, Blaser MJ, Adams S. 2016. The intestinal microbiome and estrogen receptor-positive female breast cancer. J. Natl. Cancer Inst. 108: djw029.
  14. Urbaniak C, Burton JP, Reid G. 2012. Breast, milk and microbes: a complex relationship that does not end with lactation. Womens Health (Lond) 8: 385-398. https://doi.org/10.2217/WHE.12.23
  15. Meng S, Chen B, Yang J, Wang J, Zhu D, Meng Q, et al. 2018. Study of microbiomes in aseptically collected samples of human breast tissue using needle biopsy and the potential role of in situ tissue microbiomes for promoting malignancy. Front. Oncol. 8: 318. https://doi.org/10.3389/fonc.2018.00318
  16. Wang H, Altemus J, Niazi F, Green H, Calhoun BC, Sturgis C, et al. 2017. Breast tissue, oral and urinary microbiomes in breast cancer. Oncotarget 8: 88122-88138. https://doi.org/10.18632/oncotarget.21490
  17. Urbaniak C, Cummins J, Brackstone M, Macklaim JM, Gloor GB, Baban CK, et al. 2014. Microbiota of human breast tissue. Appl. Environ. Microbiol. 80: 3007-3014. https://doi.org/10.1128/AEM.00242-14
  18. Hieken TJ, Chen J, Hoskin TL, Walther-Antonio M, Johnson S, Ramaker S, et al. 2016. The microbiome of aseptically collected human breast tissue in benign and malignant disease. Sci. Rep. 6: 30751. https://doi.org/10.1038/srep30751
  19. Urbaniak C, Gloor GB, Brackstone M, Scott L, Tangney M, Reid G. 2016. The microbiota of breast tissue and its association with breast cancer. Appl. Environ. Microbiol. 82: 5039-5048. https://doi.org/10.1128/AEM.01235-16
  20. Chan AA, Bashir M, Rivas MN, Duvall K, Sieling PA, Pieber TR, et al. 2016. Characterization of the microbiome of nipple aspirate fluid of breast cancer survivors. Sci. Rep. 6: 28061. https://doi.org/10.1038/srep28061
  21. Yazdi HR, Movafagh A, Fallah F, Alizadeh Shargh S, Mansouri N, Heidary Pour A, et al. 2016. Evaluation of Methylobacterium radiotolerance and Sphyngomonas yanoikoaie in sentinel lymph nodes of breast cancer cases. Asian Pac. J. Cancer Prev. 17: 279-285. https://doi.org/10.7314/APJCP.2016.17.S3.279
  22. Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, et al. 2020. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368: 973-980. https://doi.org/10.1126/science.aay9189
  23. Schwabe RF, Jobin C. 2013. The microbiome and cancer. Nat. Rev. Cancer 13: 800-812. https://doi.org/10.1038/nrc3610
  24. Xavier JB, Young VB, Skufca J, Ginty F, Testerman T, Pearson AT, et al. 2020. The cancer microbiome: Distinguishing direct and indirect effects requires a systemic view. Trends Cancer 6: 192-204. https://doi.org/10.1016/j.trecan.2020.01.004
  25. Smith A, Pierre JF, Makowski L, Tolley E, Lyn-Cook B, Lu L, et al. 2019. Distinct microbial communities that differ by race, stage, or breast-tumor subtype in breast tissues of non-Hispanic Black and non-Hispanic White women. Sci. Rep. 9: 11940. https://doi.org/10.1038/s41598-019-48348-1
  26. Grice EA, Segre JA. 2012. The human microbiome: our second genome. Annu. Rev. Genomics Hum. Genet. 13: 151-170. https://doi.org/10.1146/annurev-genom-090711-163814
  27. Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. 2010. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J. Clin. Oncol. 28: 2784-2795. https://doi.org/10.1200/JCO.2009.25.6529
  28. Singh K, Tantravahi U, Lomme MM, Pasquariello T, Steinhoff M, Sung CJ. 2016. Updated 2013 College of American Pathologists/American Society of Clinical Oncology (CAP/ASCO) guideline recommendations for human epidermal growth factor receptor 2 (HER2) fluorescent in situ hybridization (FISH) testing increase HER2 positive and HER2 equivocal breast cancer cases; retrospective study of HER2 FISH results of 836 invasive breast cancers. Breast Cancer Res. Treat. 157: 405-411. https://doi.org/10.1007/s10549-016-3824-x
  29. Park JU, Oh B, Lee JP, Choi MH, Lee MJ, Kim BS. 2019. Influence of microbiota on diabetic foot wound in comparison with adjacent normal skin based on the clinical features. Biomed. Res. Int. 2019: 7459236.
  30. Lluch J, Servant F, Paisse S, Valle C, Valiere S, Kuchly C, et al. 2015. The characterization of novel tissue microbiota using an optimized 16S metagenomic sequencing pipeline. PLoS One 10: e0142334. https://doi.org/10.1371/journal.pone.0142334
  31. Bukin YS, Galachyants YP, Morozov IV, Bukin SV, Zakharenko AS, Zemskaya TI. 2019. The effect of 16S rRNA region choice on bacterial community metabarcoding results. Sci. Data 6: 190007. https://doi.org/10.1038/sdata.2019.7
  32. Kameoka S, Motooka D, Watanabe S, Kubo R, Jung N, Midorikawa Y, et al. 2021. Benchmark of 16S rRNA gene amplicon sequencing using Japanese gut microbiome data from the V1-V2 and V3-V4 primer sets. BMC Genomics 22: 527. https://doi.org/10.1186/s12864-021-07746-4
  33. Lee MJ, Kang MJ, Lee SY, Lee E, Kim K, Won S, et al. 2018. Perturbations of gut microbiome genes in infants with atopic dermatitis according to feeding type. J. Allergy Clin. Immunol. 141: 1310-1319. https://doi.org/10.1016/j.jaci.2017.11.045
  34. Kim HE, Lee JJ, Lee MJ, Kim BS. 2019. Analysis of microbiome in raw chicken meat from butcher shops and packaged products in South Korea to detect the potential risk of foodborne illness. Food Res. Int. 122: 517-527. https://doi.org/10.1016/j.foodres.2019.05.032
  35. Vetrovsky T, Baldrian P. 2013. The variability of the 16S rRNA gene in bacterial genomes and its consequences for bacterial community analyses. PLoS One 8: e57923. https://doi.org/10.1371/journal.pone.0057923
  36. Edgar RC. 2013. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat. Methods 10: 996-998. https://doi.org/10.1038/nmeth.2604
  37. Yoon SH, Ha SM, Kwon S, Lim J, Kim Y, Seo H, et al. 2017. Introducing EzBioCloud: a taxonomically united database of 16S rRNA gene sequences and whole-genome assemblies. Int. J. Syst. Evol. Microbiol. 67: 1613-1617. https://doi.org/10.1099/ijsem.0.001755
  38. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. 2009. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 75: 7537-7541. https://doi.org/10.1128/AEM.01541-09
  39. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. 2011. Metagenomic biomarker discovery and explanation. Genome Biol. 12: R60. https://doi.org/10.1186/gb-2011-12-6-r60
  40. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. 2014. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics 30: 3123-3124. https://doi.org/10.1093/bioinformatics/btu494
  41. Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO, Moffatt MF, et al. 2014. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 12: 87. https://doi.org/10.1186/s12915-014-0087-z
  42. DeSantis CE, Fedewa SA, Goding Sauer A, Kramer JL, Smith RA, Jemal A. 2016. Breast cancer statistics, 2015: Convergence of incidence rates between black and white women. CA Cancer J. Clin. 66: 31-42. https://doi.org/10.3322/caac.21320
  43. Alcantara VS, Lim GH, Lim SH, Sultana R, Lee JA. 2017. Incidence and prognosis of non-metastatic triple negative breast cancer (TNBC) among different races in Southeast Asia. J. Surg. Oncol. 115: 523-537. https://doi.org/10.1002/jso.24559
  44. Rosario SR, Long MD, Affronti HC, Rowsam AM, Eng KH, Smiraglia DJ. 2018. Pan-cancer analysis of transcriptional metabolic dysregulation using the cancer genome atlas. Nat. Commun. 9: 5330. https://doi.org/10.1038/s41467-018-07232-8
  45. Chaudhary K, Poirion OB, Lu L, Garmire LX. 2018. Deep learning-based multi-omics integration robustly predicts survival in liver cancer. Clin. Cancer Res. 24: 1248-1259. https://doi.org/10.1158/1078-0432.CCR-17-0853
  46. Zeng F, Fu J, Hu F, Tang Y, Fang X, Zeng F, et al. 2018. Identification of key pathways and genes in response to trastuzumab treatment in breast cancer using bioinformatics analysis. Oncotarget 9: 32149-32160. https://doi.org/10.18632/oncotarget.24605
  47. Arias CA, Murray BE. 2012. The rise of the Enterococcus: beyond vancomycin resistance. Nat. Rev. Microbiol. 10: 266-278. https://doi.org/10.1038/nrmicro2761
  48. Kristich CJ, Rice LB, Arias CA. 2014. Enterococcal infection-treatment and antibiotic resistance, pp. 87-134. In Gilmore MS, Clewell DB, Ike Y, Shankar N (eds.), Enterococci: From commensals to leading causes of drug resistant infection, Ed. Massachusetts Eye and Ear Infirmary, Boston, USA.
  49. Hassan Z, Mustafa S, Rahim RA, Isa NM. 2016. Anti-breast cancer effects of live, heat-killed and cytoplasmic fractions of Enterococcus faecalis and Staphylococcus hominis isolated from human breast milk. In Vitro Cell Dev. Biol. Anim. 52: 337-348. https://doi.org/10.1007/s11626-015-9978-8
  50. Daillere R, Vetizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, et al. 2016. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity 45: 931-943. https://doi.org/10.1016/j.immuni.2016.09.009
  51. Huycke MM, Abrams V, Moore DR. 2002. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 23: 529-536. https://doi.org/10.1093/carcin/23.3.529
  52. Fearon ER. 2011. Molecular genetics of colorectal cancer. Annu. Rev. Pathol. 6: 479-507. https://doi.org/10.1146/annurev-pathol-011110-130235
  53. Oikari S, Kettunen T, Tiainen S, Hayrinen J, Masarwah A, Sudah M, et al. 2018. UDP-sugar accumulation drives hyaluronan synthesis in breast cancer. Matrix Biol. 67: 63-74. https://doi.org/10.1016/j.matbio.2017.12.015
  54. Arnold JM, Gu F, Ambati CR, Rasaily U, Ramirez-Pena E, Joseph R, et al. 2019. UDP-glucose 6-dehydrogenase regulates hyaluronic acid production and promotes breast cancer progression. Oncogene 39: 3089-3101.
  55. Fuster MM, Esko JD. 2005. The sweet and sour of cancer: glycans as novel therapeutic targets. Nat. Rev. Cancer 5: 526-542. https://doi.org/10.1038/nrc1649
  56. Connelly MA, Gruppen EG, Otvos JD, Dullaart RPF. 2016. Inflammatory glycoproteins in cardiometabolic disorders, autoimmune diseases and cancer. Clin. Chim. Acta 459: 177-186. https://doi.org/10.1016/j.cca.2016.06.012
  57. Furukawa K, Ohmi Y, Ohkawa Y, Bhuiyan RH, Zhang P, Tajima O, et al. 2019. New era of research on cancer-associated glycosphingolipids. Cancer Sci. 110: 1544-1551. https://doi.org/10.1111/cas.14005
  58. Tarp MA, Clausen H. 2008. Mucin-type O-glycosylation and its potential use in drug and vaccine development. Biochim. Biophys. Acta 1780: 546-563. https://doi.org/10.1016/j.bbagen.2007.09.010
  59. Prodan A, Tremaroli V, Brolin H, Zwinderman AH, Nieuwdorp M, Levin E. 2020. Comparing bioinformatic pipelines for microbial 16S rRNA amplicon sequencing. PLoS One 15: e0227434. https://doi.org/10.1371/journal.pone.0227434