Same Exposure but Two Radically Different Responses to Antibiotics: Resilience of the Salivary Microbiome versus Long-Term Microbial Shifts in Feces

Egija Zaura, Bernd W Brandt, M Joost Teixeira de Mattos, Mark J Buijs, Martien P M Caspers, Mamun-Ur Rashid, Andrej Weintraub, Carl Erik Nord, Ann Savell, Yanmin Hu, Antony R Coates, Mike Hubank, David A Spratt, Michael Wilson, Bart J F Keijser, Wim Crielaard, Egija Zaura, Bernd W Brandt, M Joost Teixeira de Mattos, Mark J Buijs, Martien P M Caspers, Mamun-Ur Rashid, Andrej Weintraub, Carl Erik Nord, Ann Savell, Yanmin Hu, Antony R Coates, Mike Hubank, David A Spratt, Michael Wilson, Bart J F Keijser, Wim Crielaard

Abstract

Due to the spread of resistance, antibiotic exposure receives increasing attention. Ecological consequences for the different niches of individual microbiomes are, however, largely ignored. Here, we report the effects of widely used antibiotics (clindamycin, ciprofloxacin, amoxicillin, and minocycline) with different modes of action on the ecology of both the gut and the oral microbiomes in 66 healthy adults from the United Kingdom and Sweden in a two-center randomized placebo-controlled clinical trial. Feces and saliva were collected at baseline, immediately after exposure, and 1, 2, 4, and 12 months after administration of antibiotics or placebo. Sequences of 16S rRNA gene amplicons from all samples and metagenomic shotgun sequences from selected baseline and post-antibiotic-treatment sample pairs were analyzed. Additionally, metagenomic predictions based on 16S rRNA gene amplicon data were performed using PICRUSt. The salivary microbiome was found to be significantly more robust, whereas the antibiotics negatively affected the fecal microbiome: in particular, health-associated butyrate-producing species became strongly underrepresented. Additionally, exposure to different antibiotics enriched genes associated with antibiotic resistance. In conclusion, healthy individuals, exposed to a single antibiotic treatment, undergo considerable microbial shifts and enrichment in antibiotic resistance in their feces, while their salivary microbiome composition remains unexpectedly stable. The health-related consequences for the gut microbiome should increase the awareness of the individual risks involved with antibiotic use, especially in a (diseased) population with an already dysregulated microbiome. On the other hand, understanding the mechanisms behind the resilience of the oral microbiome toward ecological collapse might prove useful in combating microbial dysbiosis elsewhere in the body.

Importance: Many health care professionals use antibiotic prophylaxis strategies to prevent infection after surgery. This practice is under debate since it enhances the spread of antibiotic resistance. Another important reason to avoid nonessential use of antibiotics, the impact on our microbiome, has hardly received attention. In this study, we assessed the impact of antibiotics on the human microbial ecology at two niches. We followed the oral and gut microbiomes in 66 individuals from before, immediately after, and up to 12 months after exposure to different antibiotic classes. The salivary microbiome recovered quickly and was surprisingly robust toward antibiotic-induced disturbance. The fecal microbiome was severely affected by most antibiotics: for months, health-associated butyrate-producing species became strongly underrepresented. Additionally, there was an enrichment of genes associated with antibiotic resistance. Clearly, even a single antibiotic treatment in healthy individuals contributes to the risk of resistance development and leads to long-lasting detrimental shifts in the gut microbiome.

Copyright © 2015 Zaura et al.

Figures

FIG 1
FIG 1
Comparison of baseline microbiome profiles from both types of samples, saliva and feces (A), and per sample type, feces (B) and saliva (C), by study site, KI (Sweden) and HP (United Kingdom). The PCA plot is based on randomly subsampled and log2-transformed OTU data. The data set included 37 saliva-feces baseline sample pairs from the HP study and 29 from the KI study. The red ellipse highlights the two “types” of fecal samples—Prevotella- and Bacteroides-dominated samples, respectively.
FIG 2
FIG 2
Effects of antibiotics on microbiome profiles of feces (A) and saliva (B) from the KI study and feces (C) and saliva (D) from the HP study. The PCA plot is based on log2-transformed OTU data. Different colors indicate different time points; different symbols indicate different treatment groups. Outliers in the KI (A) and HP (C) fecal data sets are highlighted with the respective subject number.
FIG 3
FIG 3
Similarity in microbiome profiles between the baseline (BL) and the other visits (W1, week 1; M1, month 1; M2, month 2; M4, month 4; M12, month 12). The horizontal bar indicates the mean value; the error bar indicates the 95% confidence interval. Bray-Curtis similarities were calculated between the log2-transformed microbiome profiles of the baseline and each of the other time point samples of the respective individual. Brackets connect statistically significantly different groups within each visit pair (P < 0.05; one-way analysis of variance, Games-Howell post hoc test).
FIG 4
FIG 4
Relative abundance of the predicted KEGG orthologous groups (KOs) in the fecal (A) and salivary (B) samples from the clindamycin group plotted against the samples from the KI placebo group per individual time point. Error lines indicate standard deviations. No significant differences were observed in saliva, while in feces, 3 KOs at 1 week post-antibiotic treatment and 512 of the 4,606 predicted KOs at 1 month post-antibiotic treatment were significantly different in their proportions from the placebo group (P < 0.005, Welch’s t test, Welch’s inverted confidence interval method, and Storey FDR correction for multiple comparisons).
FIG 5
FIG 5
Most significantly affected KOs (26 of 520) in the predicted metagenomes from clindamycin-exposed feces at month 1 compared to the respective placebo group samples. FAD, flavin adenine dinucleotide; SEPHS, selenophosphate synthetase.

References

    1. Cantas L, Shah SQA, Cavaco LM, Manaia CM, Walsh F, Popowska M, Garelick H, Bürgmann H, Sørum H. 2013. A brief multi-disciplinary review on antimicrobial resistance in medicine and its linkage to the global environmental microbiota. Front Microbiol 4:96. doi:10.3389/fmicb.2013.00096.
    1. Alanis AJ. 2005. Resistance to antibiotics: are we in the post-antibiotic era? Arch Med Res 36:697–705. doi:10.1016/j.arcmed.2005.06.009.
    1. Clemente J, Ursell L, Parfrey L, Knight R. 2012. The impact of the gut microbiota on human health: an integrative view. Cell 148:1258–1270. doi:10.1016/j.cell.2012.01.035.
    1. Modi SR, Collins JJ, Relman DA. 2014. Antibiotics and the gut microbiota. J Clin Invest 124:4212–4218. doi:10.1172/JCI72333.
    1. Munyaka PM, Khafipour E, Ghia J. 2014. External influence of early childhood establishment of gut microbiota and subsequent health implications. Front Pediatr 2:109. doi:10.3389/fped.2014.00109.
    1. Roberts SE, Wotton CJ, Williams JG, Griffith M, Goldacre MJ. 2011. Perinatal and early life risk factors for inflammatory bowel disease. World J Gastroenterol 17:743–749. doi:10.3748/wjg.v17.i6.743.
    1. Ferreira CM, Vieira AT, Vinolo MAR, Oliveira FA, Curi R, Martins FDS. 2014. The central role of the gut microbiota in chronic inflammatory diseases. J Immunol Res 2014:689492. doi:10.1155/2014/689492.
    1. Willing BP, Russell SL, Finlay BB. 2011. Shifting the balance: antibiotic effects on host–microbiota mutualism. Nat Rev Microbiol 9:233–243. doi:10.1038/nrmicro2536.
    1. Abdulah R. 2012. Antibiotic abuse in developing countries. Pharm Regul Aff 1:e106. doi:10.4172/2167-7689.1000e106.
    1. Widayati A, Suryawati S, de Crespigny C, Hiller JE. 2011. Self medication with antibiotics in Yogyakarta City Indonesia: a cross sectional population-based survey. BMC Res Notes 4:491. doi:10.1186/1756-0500-4-491.
    1. ESPAUR Writing Committee 2014. English surveillance programme for antimicrobial utilization and resistance (ESPAUR). Report 2014 Public Health England, London, United Kingdom: .
    1. European Centre for Disease Prevention and Control. 2014. Surveillance of antimicrobial consumption in Europe 2012. ECDC, European Centre for Disease Prevention and Control, Stockholm, Sweden.
    1. Fernandez y Mostajo M, Zaura E, Crielaard W, Beertsen W. 2011. Does routine analysis of subgingival microbiota in periodontitis contribute to patient benefit? Eur J Oral Sci 119:259–264. doi:10.1111/j.1600-0722.2011.00828.x.
    1. Keenan JR, Veitz-Keenan A. 2015. Antibiotic prophylaxis for dental implant placement? Evid Based Dent 16:52–53. doi:10.1038/sj.ebd.6401097.
    1. Oomens MAE, Forouzanfar T. 2012. Antibiotic prophylaxis in third molar surgery: a review. Oral Surg Oral Med Oral Pathol Oral Radiol 114:e5–e12. doi:10.1016/j.oooo.2011.10.023.
    1. Dethlefsen L, Huse S, Sogin ML, Relman DA. 2008. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol 6:e280. doi:10.1371/journal.pbio.0060280.
    1. Dethlefsen L, Relman DA. 2011. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A 108:4554–4561. doi:10.1073/pnas.1000087107.
    1. Rashid M-, Zaura E, Buijs MJ, Keijser BJF, Crielaard W, Nord CE, Weintraub A. 2015. Determining the long-term effect of antibiotic administration on the human normal intestinal microbiota using culture and pyrosequencing methods. Clin Infect Dis 60:S77–S84. doi:10.1093/cid/civ137.
    1. Canu A, Leclercq R. 2009. Macrolides and lincosamides, p 211–221. In Mayers DL. (ed), Antimicrobial drug resistance, vol 1 Mechanisms of drug resistance. Humana Press, Springer, New York, NY.
    1. Guay D. 2007. Update on clindamycin in the management of bacterial, fungal and protozoal infections. Expert Opin Pharmacother 8:2401–2444. doi:10.1517/14656566.8.14.2401.
    1. Moudgal VV, Kaatz GW. 2009. Fluoroquinolone resistance in bacteria, p 195–205. In Mayers DL. (ed), Antimicrobial drug resistance, vol 1 Mechanisms of drug resistance. Humana Press, Springer, New York, NY.
    1. Rang HP, Dale MM. 1991. Pharmacology, 2nd ed, p 804–832. Churchill Livingstone, London, United Kingdom.
    1. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, Beiko RG, Huttenhower C. 2013. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol 31:814–821. doi:10.1038/nbt.2676.
    1. Liu B, Pop M. 2009. ARDB—antibiotic resistance genes database. Nucleic Acids Res 37:D443–D447. doi:10.1093/nar/gkn656.
    1. Vital M, Howe AC, Tiedje JM. 2014. Revealing the bacterial butyrate synthesis pathways by analyzing (meta)genomic data. mBio 5:e00889-14. doi:10.1128/mBio.00889-14.
    1. Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer R-. 2008. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther 27:104–119. doi:10.1111/j.1365-2036.2007.03562.x.
    1. Albenberg LG, Wu GD. 2014. Diet and the intestinal microbiome: associations, functions, and implications for health and disease. Gastroenterology 146:1564–1572. doi:10.1053/j.gastro.2014.01.058.
    1. Mager DL, Ximenez-Fyvie LA, Haffajee AD, Socransky SS. 2003. Distribution of selected bacterial species on intraoral surfaces. J Clin Periodontol 30:644–654. doi:10.1034/j.1600-051X.2003.00376.x.
    1. Li K, Bihan M, Methé BA. 2013. Analyses of the stability and core taxonomic memberships of the human microbiome. PLoS One 8:e63139. doi:10.1371/journal.pone.0063139.
    1. Sommer MOA, Dantas G, Church GM. 2009. Functional characterization of the antibiotic resistance reservoir in the human microflora. Science 325:1128–1131. doi:10.1126/science.1176950.
    1. Mölstad S, Erntell M, Hanberger H, Melander E, Norman C, Skoog G, Lundborg CS, Söderström A, Torell E, Cars O. 2008. Sustained reduction of antibiotic use and low bacterial resistance: 10-year follow-up of the Swedish Strama programme. Lancet Infect Dis 8:125–132. doi:10.1016/S1473-3099(08)70017-3.
    1. Shoemaker NB, Vlamakis H, Hayes K, Salyers AA. 2001. Evidence for extensive resistance gene transfer among Bacteroides spp. and among Bacteroides and other genera in the human colon. Appl Environ Microbiol 67:561–568. doi:10.1128/AEM.67.2.561-568.2001.
    1. Ochman H, Lawrence JG, Groisman EA. 2000. Lateral gene transfer and the nature of bacterial innovation. Nature 405:299–304. doi:10.1038/35012500.
    1. Marsh PD, Moter A, Devine DA. 2011. Dental plaque biofilms: communities, conflict and control. Periodontol 2000 55:16–35. doi:10.1111/j.1600-0757.2009.00339.x.
    1. Abeles SR, Ly M, Santiago-Rodriguez TM, Pride DT. 2015. Effects of long term antibiotic therapy on human oral and fecal viromes. PLoS One 10:e0134941. doi:10.1371/journal.pone.0134941.
    1. Zaura E, Keijser BJ, Huse SM, Crielaard W. 2009. Defining the healthy “core microbiome” of oral microbial communities. BMC Microbiol 9:259. doi:10.1186/1471-2180-9-259.
    1. Kraneveld EA, Buijs MJ, Bonder MJ, Visser M, Keijser BJF, Crielaard W, Zaura E. 2012. The relation between oral Candida load and bacterial microbiome profiles in Dutch older adults. PLoS One 7:e42770. doi:10.1371/journal.pone.0042770.
    1. Koopman JE, Röling WFM, Buijs MJ, Sissons CH, ten Cate JM, Keijser BJF, Crielaard W, Zaura E. 2015. Stability and resilience of oral microcosms toward acidification and Candida outgrowth by arginine supplementation. Microb Ecol 69:422–433. doi:10.1007/s00248-014-0535-x.
    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. Edgar RC. 2010. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26:2460–2461. doi:10.1093/bioinformatics/btq461.
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat B, White O, Kelley ST, Methé B, Schloss PD, Gevers D, Mitreva M, Huttenhower C. 2012. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput Biol 8:e1002358. doi:10.1371/journal.pcbi.1002358.
    1. Hammer O, Harper DAT, Ryan PD. 2001. PAST: paleontological statistics software package for education and data analysis. Palaeontol Electron 4:1–9.
    1. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. 2014. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics 30:3123–3124. doi:10.1093/bioinformatics/btu494.

Source: PubMed

3
購読する