Alterations of the Human Lung and Gut Microbiomes in Non-Small Cell Lung Carcinomas and Distant Metastasis

Hui Lu, Na L Gao, Fan Tong, Jiaojiao Wang, Huanhuan Li, Ruiguang Zhang, Hong Ma, Nong Yang, Yongchang Zhang, Ye Wang, Zhiwen Liang, Hao Zeng, Wei-Hua Chen, Xiaorong Dong, Hui Lu, Na L Gao, Fan Tong, Jiaojiao Wang, Huanhuan Li, Ruiguang Zhang, Hong Ma, Nong Yang, Yongchang Zhang, Ye Wang, Zhiwen Liang, Hao Zeng, Wei-Hua Chen, Xiaorong Dong

Abstract

Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. Although dysbiosis of the lung and gut microbiota have been associated with NSCLC, their relative contributions are unclear; in addition, their roles in distant metastasis (DM) are still illusive. We recruited in total 121 participants, including 87 newly diagnosed treatment-naive NSCLC patients of various stages and 34 healthy volunteers, and surveyed their fecal and sputum microbiota. We compared the microbial profiles between groups, identified microbial biomarkers, and generated machine learning models for distinguishing healthy individuals from patients with NSCLC and patients of various stages. We found significant perturbations of gut and sputum microbiota in patients with NSCLC and DM. A machine learning model combining both microbiota (combined model) performed better than an individual data set in patient stratification, with the highest area under the curve (AUC) value of 0.896. Sputum and gut microbiota both contributed to the combined model; in most cases, sputum-only models performed similar to the combined models. Several microbial biomarkers were shared by both microbiotas, indicating their similar roles at distinct body sites. Microbial biomarkers of distinct disease stages were mostly shared, suggesting biomarkers for DM could be acquired early. Furthermore, Pseudomonas aeruginosa, a species previously associated with wound infections, was significantly more abundant in brain metastasis, indicating that distinct types of DMs could have different microbes. Our results indicate that alterations of the sputum microbiota have stronger relationships with NSCLC and DM than the gut and strongly support the feasibility of metagenome-based noninvasive disease diagnosis and risk evaluation. (This study has been registered at ClinicalTrials.gov under registration no. NCT03454685). IMPORTANCE Our survey on gut and sputum microbiota revealed that both were significantly disturbed in non-small cell lung cancer (NSCLC) and associated with distant metastasis (DM) while only the sputum microbiota was associated with non-DM NSCLC. The lung microbiota could therefore have a stronger association with (and thus may contribute more to) disease development than the gut microbiota. Mathematic models using both microbiotas performed better in patient stratification than using individual microbiota. Sputum models, however, performed similar to the combined models, suggesting a convenient, noninvasive diagnostic for NSCLC. Microbial biomarkers of distinct disease stages were mostly shared, suggesting that the same set of microbes were underlying disease progression, and the signals for distant metastasis could be acquired at early stages of the disease. Our results strongly support the feasibility of noninvasive diagnosis of NSCLC, including distant metastasis, are of clinical importance, and should warrant further research on the underlying molecular mechanisms.

Keywords: NSCLC; brain metastasis; distant metastasis; gut microbiota; lung microbiota; machine learning; patient stratification.

Figures

FIG 1
FIG 1
Sputum and gut microbiota differed significantly in terms of alpha- and beta-diversities. (A) Numbers of sputum (red) and gut (blue) samples collected in this study and their distributions in healthy controls and distinct disease stage groups; CON, healthy controls; I_III, patients with stages of I to III; DM, patients with distant metastasis (also referred to as stage IV). Disease stages were assigned according to the 8th American Joint Committee on Cancer guidelines. (B) Comparisons of alpha-diversity and beta-diversity between the sputum and gut in healthy controls. Richness index (alpha-diversity; left) at the genus level was significantly lower in feces; principal-coordinate analysis (PCoA; right) based on Bray-Curtis distance at the genus level showed that the overall microbiota composition was different between fecal and sputum samples. Wilcoxon rank sum tests were used to compare between groups. (C) Comparisons of alpha-diversity (left) and beta-diversity (right) between sputum and gut in NSCLC patients (stages I to IV). Level of significance: ***, P < 0.001; **, P < 0.01; *, P < 0.05; NS, P ≥ 0.05.
FIG 2
FIG 2
Global alteration of the sputum microbiota was associated with NSCLC and distant metastasis (A, B), while the fecal microbiota was only significantly associated with the latter (C, D). (A) Significant differences were found in alpha-diversity between healthy controls and individuals with NSCLC. Richness index, Shannon index, evenness index and Simpson index at the genus level were significantly lower in patients than in healthy controls. A Wilcoxon rank sum test was used to compare between groups. Level of significance: ***, P < 0.001; **, P < 0.01; *, P < 0.05; NS, P ≥ 0.05. (B) Alpha-diversity of sputum dysbiosis in pairwise comparisons. The richness index (top left), Shannon index (top right), and evenness index (bottom left) are shown. The Shannon index and richness index were significantly lower in patients than in healthy controls. An ANOVA with a post hoc Tukey HSD test was used to compare between groups. Level of significance: ***, P < 0.001; **, P < 0.01; *, P < 0.05; NS, P ≥ 0.05. (C) Significant differences were found in beta-diversity between controls and individuals with NSCLC as well as between controls and I_III, controls and DM, and I_III and DM groups, indicating that dysbiosis of the sputum microbiota was associated with lung cancer development and metastasis. Conversely, applying similar analyses to fecal samples, no differences in alpha-diversities were apparent (D), but the beta-diversity in controls versus DM and I_III versus DM were different (P = 0.07), suggesting that fecal microbiota dysbiosis was associated with distal metastasis but not NSCLC.
FIG 3
FIG 3
Shared and distinct microbial biomarkers between subject groups in sputum (red) and fecal (blue) microbiota. Differentially abundant microbial biomarkers between subject groups were identified using LEfSe analyses; red and blue bar plots indicate LEfSe results for sputum and fecal microbiota, respectively. The relative abundance of 18 and 9 genera was significantly different between NSCLC and control groups in sputum (A) and feces (B), respectively. To identify biomarkers for specific disease stages, we compared neighboring groups along the disease progression in sputum (C) and feces (D). LDA, linear discriminant analysis.
FIG 4
FIG 4
The top-ranked genera of the mixed models shown in Table 2 for each disease stage. The genera were ranked by the robustness of 1,000 repeats; therefore, box plots were used here to demonstrate the means and distributions of these values. (A) Control versus NSCLC. (B) Control versus I_III. (C) I_III versus DM. (D) Control versus DM. Red boxes represent sputum-derived genera, and blue boxes represent gut-derived genera. Please consult Table 2 for details on the model performance. F, feces; S, sputum.
FIG 5
FIG 5
Patients with brain metastasis differed significantly from other distant metastasis in microbial profiles of the sputum and feces. (A) Numbers of sputum (red) and gut (blue) brain metastasis samples (left); BM, NSCLC patients in stage IV with brain metastasis; non-BM, stage IV NSCLC patients without brain metastasis. A principal-coordinate analysis showed differences in beta-diversity between BM and non-BM in sputum (middle) but not in the gut (right). LEfSe (left) analysis and Wilcoxon rank sum test (right) of differentially abundant microbial biomarkers between BM and non-BM in sputum (B) and the gut (C). Level of significance: ***, P < 0.001; **, P < 0.01; *, P < 0.05; NS, P ≥ 0.05. The star indicates that the genus Pseudomonas was significantly different in abundance.
FIG 6
FIG 6
Brain metastasis classification based on taxonomic profiles of the sputum, gut, and both. (A) The classification performance using the relative abundance of genera as the area under the ROC curve between BM and non-BM. (B) The top 20 genera important to the sputum model ranked by the median values of 1,000 repeats. Box plots were used here to demonstrate the medians and distributions of these values. The star indicates that the genus Pseudomonas was significantly different in abundance using LEfSe and Wilcoxon analysis.

References

    1. Siegel RL, Miller KD, Jemal A. 2018. Cancer statistics, 2018. CA Cancer J Clin 68:7–30. doi:10.3322/caac.21442.
    1. Shen H, Zhu M, Wang C. 2019. Precision oncology of lung cancer: genetic and genomic differences in Chinese population. NPJ Precis Oncol 3:14. doi:10.1038/s41698-019-0086-1.
    1. Li Y, Xiao X, Han Y, Gorlova O, Qian D, Leighl N, Johansen JS, Barnett M, Chen C, Goodman G, Cox A, Taylor F, Woll P, Wichmann H-E, Manz J, Muley T, Risch A, Rosenberger A, Arnold SM, Haura EB, Bolca C, Holcatova I, Janout V, Kontic M, Lissowska J, Mukeria A, Ognjanovic S, Orlowski TM, Scelo G, Swiatkowska B, Zaridze D, Bakke P, Skaug V, Zienolddiny S, Duell EJ, Butler LM, Houlston R, Soler Artigas M, Grankvist K, Johansson M, Shepherd FA, Marcus MW, Brunnström H, Manjer J, Melander O, Muller DC, Overvad K, Trichopoulou A, Tumino R, Liu G, et al. . 2018. Genome-wide interaction study of smoking behavior and non-small cell lung cancer risk in Caucasian population. Carcinogenesis 39:336–346. doi:10.1093/carcin/bgx113.
    1. Czene K, Lichtenstein P, Hemminki K. 2002. Environmental and heritable causes of cancer among 9.6 million individuals in the Swedish Family-Cancer Database. Int J Cancer 99:260–266. doi:10.1002/ijc.10332.
    1. Dai J, Shen W, Wen W, Chang J, Wang T, Chen H, Jin G, Ma H, Wu C, Li L, Song F, Zeng YXin, Jiang Y, Chen J, Wang C, Zhu M, Zhou W, Du J, Xiang Y, Shu X-O, Hu Z, Zhou W, Chen K, Xu J, Jia W, Lin D, Zheng W, Shen H. 2017. Estimation of heritability for nine common cancers using data from genome-wide association studies in Chinese population. Int J Cancer 140:329–336. doi:10.1002/ijc.30447.
    1. Zhuang H, Cheng L, Wang Y, Zhang Y-K, Zhao M-F, Liang G-D, Zhang M-C, Li Y-G, Zhao J-B, Gao Y-N, Zhou Y-J, Liu S-L. 2019. Dysbiosis of the gut microbiome in lung cancer. Front Cell Infect Microbiol 9:112. doi:10.3389/fcimb.2019.00112.
    1. Jin C, Lagoudas GK, Zhao C, Bullman S, Bhutkar A, Hu B, Ameh S, Sandel D, Liang XS, Mazzilli S, Whary MT, Meyerson M, Germain R, Blainey PC, Fox JG, Jacks T. 2019. Commensal microbiota promote lung cancer development via γδ T cells. Cell 176:998–1013. doi:10.1016/j.cell.2018.12.040.
    1. Jin Y, Dong H, Xia L, Yang Y, Zhu Y, Shen Y, Zheng H, Yao C, Wang Y, Lu S. 2019. The diversity of gut microbiome is associated with favorable responses to anti-programmed death 1 immunotherapy in Chinese patients with NSCLC. J Thorac Oncol 14:1378–1389. doi:10.1016/j.jtho.2019.04.007.
    1. Lee SH, Sung JY, Yong D, Chun J, Kim SY, Song JH, Chung KS, Kim EY, Jung JY, Kang YA, Kim YS, Kim SK, Chang J, Park MS. 2016. Characterization of microbiome in bronchoalveolar lavage fluid of patients with lung cancer comparing with benign mass like lesions. Lung Cancer 102:89–95. doi:10.1016/j.lungcan.2016.10.016.
    1. Liu H-X, Tao L-L, Zhang J, Zhu Y-G, Zheng Y, Liu D, Zhou M, Ke H, Shi M-M, Qu J-M. 2018. Difference of lower airway microbiome in bilateral protected specimen brush between lung cancer patients with unilateral lobar masses and control subjects. Int J Cancer 142:769–778. doi:10.1002/ijc.31098.
    1. Ren L, Zhang R, Rao J, Xiao Y, Zhang Z, Yang B, Cao D, Zhong H, Ning P, Shang Y, Li M, Gao Z, Wang J. 2018. Transcriptionally active lung microbiome and its association with bacterial biomass and host inflammatory status. mSystems 3:e00199-18. doi:10.1128/mSystems.00199-18.
    1. Huang D, Su X, Yuan M, Zhang S, He J, Deng Q, Qiu W, Dong H, Cai S. 2019. The characterization of lung microbiome in lung cancer patients with different clinicopathology. Am J Cancer Res 9:2047–2063.
    1. Peng Z, Zhou J, Tian L. 2020. Pathogenic characteristics of sputum and bronchoalveolar lavage fluid samples from patients with lower respiratory tract infection in a large teaching hospital in China: a retrospective study. BMC Pulm Med 20:233. doi:10.1186/s12890-020-01275-8.
    1. Santoni M, Piva F, Conti A, Santoni A, Cimadamore A, Scarpelli M, Battelli N, Montironi R. 2018. Re: gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Eur Urol 74:521–522. doi:10.1016/j.eururo.2018.05.033.
    1. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M-L, Luke JJ, Gajewski TF. 2018. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359:104–108. doi:10.1126/science.aao3290.
    1. Gajdacs M, Urban E. 2020. The pathogenic role of Actinomyces spp. and related organisms in genitourinary infections: discoveries in the new, modern diagnostic era. Antibiotics (Basel) 9:524. doi:10.3390/antibiotics9080524.
    1. Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi K, Yu G-Y, Osterreicher CH, Hung KE, Datz C, Feng Y, Fearon ER, Oukka M, Tessarollo L, Coppola V, Yarovinsky F, Cheroutre H, Eckmann L, Trinchieri G, Karin M. 2012. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature 491:254–258. doi:10.1038/nature11465.
    1. Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, Agdashian D, Terabe M, Berzofsky JA, Fako V, Ritz T, Longerich T, Theriot CM, McCulloch JA, Roy S, Yuan W, Thovarai V, Sen SK, Ruchirawat M, Korangy F, Wang XW, Trinchieri G, Greten TF. 2018. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 360:eaan5931. doi:10.1126/science.aan5931.
    1. Cogdill AP, Gaudreau PO, Arora R, Gopalakrishnan V, Wargo JA. 2018. The impact of intratumoral and gastrointestinal microbiota on systemic cancer therapy. Trends Immunol 39:900–920. doi:10.1016/j.it.2018.09.007.
    1. Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, Chen Y, Chen H, Hong J, Zou W, Fang J-Y. 2017. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell 170:548–563. doi:10.1016/j.cell.2017.07.008.
    1. Dickson RP, Cox MJ. 2017. Gut microbiota and protection from pneumococcal pneumonia. Gut 66:384. doi:10.1136/gutjnl-2016-311823.
    1. Rami-Porta R, Asamura H, Travis WD, Rusch VW. 2017. Lung cancer—major changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin 67:138–155. doi:10.3322/caac.21390.
    1. Chen Y, Yang F, Lu H, Wang B, Chen Y, Lei D, Wang Y, Zhu B, Li L. 2011. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology 54:562–572. doi:10.1002/hep.24423.
    1. Durack J, Christian LS, Nariya S, Gonzalez J, Bhakta NR, Ansel KM, Beigelman A, Castro M, Dyer A-M, Israel E, Kraft M, Martin RJ, Mauger DT, Peters SP, Rosenberg SR, Sorkness CA, Wechsler ME, Wenzel SE, White SR, Lynch SV, Boushey HA, Huang YJ, National Heart, Lung, and Blood Institute’s”AsthmaNet“ . 2020. Distinct associations of sputum and oral microbiota with atopic, immunologic, and clinical features in mild asthma. J Allergy Clin Immunol 146:1016–1026. doi:10.1016/j.jaci.2020.03.028.
    1. Zheng L, Sun R, Zhu Y, Li Z, She X, Jian X, Yu F, Deng X, Sai B, Wang L, Zhou W, Wu M, Li G, Tang J, Jia W, Xiang J. 2021. Lung microbiome alterations in NSCLC patients. Sci Rep 11:11736. doi:10.1038/s41598-021-91195-2.
    1. Clavel T, Lepage P, Charrier C. 2014. The Family Coriobacteriaceae, p 201–238. In Rosenberg E, DeLong EF, Lory S, Stackebrandt E, Thompson F (ed), The Prokaryotes. Springer, Berlin, Germany.
    1. Kim K-S, Rowlinson M-C, Bennion R, Liu C, Talan D, Summanen P, Finegold SM. 2010. Characterization of Slackia exigua isolated from human wound infections, including abscesses of intestinal origin. J Clin Microbiol 48:1070–1075. doi:10.1128/JCM.01576-09.
    1. Chavez de Paz LE, Molander A, Dahlen G. 2004. Gram-positive rods prevailing in teeth with apical periodontitis undergoing root canal treatment. Int Endod J 37:579–587. doi:10.1111/j.1365-2591.2004.00845.x.
    1. Marchesi JR, Dutilh BE, Hall N, Peters WHM, Roelofs R, Boleij A, Tjalsma H. 2011. Towards the human colorectal cancer microbiome. PLoS One 6:e20447. doi:10.1371/journal.pone.0020447.
    1. Tjalsma H, Boleij A, Marchesi JR, Dutilh BE. 2012. A bacterial driver-passenger model for colorectal cancer: beyond the usual suspects. Nat Rev Microbiol 10:575–582. doi:10.1038/nrmicro2819.
    1. Grenda A, Krawczyk P. 2021. Cancer trigger or remedy: two faces of the human microbiome. Appl Microbiol Biotechnol 105:1395–1405. doi:10.1007/s00253-021-11125-0.
    1. Valour F, Sénéchal A, Dupieux C, Karsenty J, Lustig S, Breton P, Gleizal A, Boussel L, Laurent F, Braun E, Chidiac C, Ader F, Ferry T. 2014. Actinomycosis: etiology, clinical features, diagnosis, treatment, and management. Infect Drug Resist 7:183–197.
    1. Schmidt TS, Hayward MR, Coelho LP, Li SS, Costea PI, Voigt AY, Wirbel J, Maistrenko OM, Alves RJ, Bergsten E, de Beaufort C, Sobhani I, Heintz-Buschart A, Sunagawa S, Zeller G, Wilmes P, Bork P. 2019. Extensive transmission of microbes along the gastrointestinal tract. eLife 8:e42693. doi:10.7554/eLife.42693.
    1. Zych KWJ, Essex M, Breuer K, Karcher N, Costea PI, Sunagawa S, Bork P, Zeller G. 2018. SIAMCAT: statistical inference of associations between microbial communities and host phenotypes. . Accessed 11 May 2020.
    1. Galanis E. 2007. Campylobacter and bacterial gastroenteritis. CMAJ 177:570–571. doi:10.1503/cmaj.070660.
    1. Fitzgerald C. 2015. Campylobacter. Clin Lab Med 35:289–298. doi:10.1016/j.cll.2015.03.001.
    1. Liu F, Ma R, Wang Y, Zhang L. 2018. The clinical importance of Campylobacter concisus and other human hosted Campylobacter species. Front Cell Infect Microbiol 8:243. doi:10.3389/fcimb.2018.00243.
    1. Douglas GM, Maffei VJ, Zaneveld JR, Yurgel SN, Brown JR, Taylor CM, Huttenhower C, Langille MGI. 2020. PICRUSt2 for prediction of metagenome functions. Nat Biotechnol 38:685–688. doi:10.1038/s41587-020-0548-6.
    1. Wu S, Sun C, Li Y, Wang T, Jia L, Lai S, Yang Y, Luo P, Dai D, Yang Y-Q, Luo Q, Gao NL, Ning K, He L-j, Zhao X-M, Chen W-H. 2020. GMrepo: a database of curated and consistently annotated human gut metagenomes. Nucleic Acids Res 48:D545–D553. doi:10.1093/nar/gkz764.
    1. Yum H-K, Park I-N, Shin B-M, Choi S-J. 2014. Recurrent Pseudomonas aeruginosa infection in chronic lung diseases: relapse or reinfection? Tuberc Respir Dis (Seoul) 77:172–177. doi:10.4046/trd.2014.77.4.172.
    1. White L, Mirrani G, Grover M, Rollason J, Malin A, Suntharalingam J. 2012. Outcomes of Pseudomonas eradication therapy in patients with non-cystic fibrosis bronchiectasis. Respir Med 106:356–360. doi:10.1016/j.rmed.2011.11.018.
    1. Kiu R, Caim S, Alcon-Giner C, Belteki G, Clarke P, Pickard D, Dougan G, Hall LJ. 2017. Preterm infant-associated Clostridium tertium, Clostridium cadaveris, and Clostridium paraputrificum strains: genomic and evolutionary insights. Genome Biol Evol 9:2707–2714. doi:10.1093/gbe/evx210.
    1. Seale AC, Baker CJ, Berkley JA, Madhi SA, Ordi J, Saha SK, Schrag SJ, Sobanjo-ter Meulen A, Vekemans J. 2019. Vaccines for maternal immunization against Group B Streptococcus disease: WHO perspectives on case ascertainment and case definitions. Vaccine 37:4877–4885. doi:10.1016/j.vaccine.2019.07.012.
    1. Stratton WSJE. 2015. Neisseria meningitidis, p 1729–1750. In Tan Y-W, Sussman M, Liu D, Poxton I, Schwartzman J (ed), Molecular medical microbiology, 2nd ed, vol 3. Academic Press, Cambridge, MA.
    1. Lau SK, Woo PC, Fung AM, Chan K-M, Woo GK, Yuen K-Y. 2004. Anaerobic, non-sporulating, Gram-positive bacilli bacteraemia characterized by 16S rRNA gene sequencing. J Med Microbiol 53:1247–1253. doi:10.1099/jmm.0.45803-0.
    1. Wang K, Huang Y, Zhang Z, Liao J, Ding Y, Fang X, Liu L, Luo J, Kong J. 2019. A preliminary study of microbiota diversity in saliva and bronchoalveolar lavage fluid from patients with primary bronchogenic carcinoma. Med Sci Monit 25:2819–2834. doi:10.12659/MSM.915332.
    1. Rodriguez J, Hiel S, Delzenne NM. 2018. Metformin: old friend, new ways of action—implication of the gut microbiome? Curr Opin Clin Nutr Metab Care 21:294–301. doi:10.1097/MCO.0000000000000468.
    1. Jiang P, Wu S, Luo Q, Zhao X-m, Chen W-H. 2021. Metagenomic analysis of common intestinal diseases reveals relationships among microbial signatures and powers multidisease diagnostic models. mSystems 6:e00112-21. doi:10.1128/mSystems.00112-21.
    1. Sun L, Zhang X, Zhang Y, Zheng K, Xiang Q, Chen N, Chen Z, Zhang N, Zhu J, He Q. 2019. Antibiotic-induced disruption of gut microbiota alters local metabolomes and immune responses. Front Cell Infect Microbiol 9:99. doi:10.3389/fcimb.2019.00099.
    1. Wu S, Jiang P, Zhao X-M, Chen W-H. 2021. Treatment regimens may compromise gut-microbiome-derived signatures for liver cirrhosis. Cell Metab 33:455–456. doi:10.1016/j.cmet.2021.02.012.
    1. Hosgood HD, III, Sapkota AR, Rothman N, Rohan T, Hu W, Xu J, Vermeulen R, He X, White JR, Wu G, Wei F, Mongodin EF, Lan Q. 2014. The potential role of lung microbiota in lung cancer attributed to household coal burning exposures. Environ Mol Mutagen 55:643–651. doi:10.1002/em.21878.
    1. Kim J, Lee H, An J, Song Y, Lee C-K, Kim K, Kong H. 2019. Alterations in gut microbiota by statin therapy and possible intermediate effects on hyperglycemia and hyperlipidemia. Front Microbiol 10:1947. doi:10.3389/fmicb.2019.01947.
    1. Bolger AM, Lohse M, Usadel B. 2014. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30:2114–2120. doi:10.1093/bioinformatics/btu170.
    1. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. 2016. DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods 13:581–583. doi:10.1038/nmeth.3869.
    1. Allard G, Ryan FJ, Jeffery IB, Claesson MJ. 2015. SPINGO: a rapid species-classifier for microbial amplicon sequences. BMC Bioinformatics 16:324. doi:10.1186/s12859-015-0747-1.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, Huttenhower C. 2011. Metagenomic biomarker discovery and explanation. Genome Biol 12:R60. doi:10.1186/gb-2011-12-6-r60.
    1. Bauer DF. 1972. Constructing confidence sets using rank statistics. J Am Stat Assoc 67:687–690. doi:10.1080/01621459.1972.10481279.

Source: PubMed

3
Iratkozz fel