Distinct impact of antibiotics on the gut microbiome and resistome: a longitudinal multicenter cohort study

Matthias Willmann, Maria J G T Vehreschild, Lena M Biehl, Wichard Vogel, Daniela Dörfel, Axel Hamprecht, Harald Seifert, Ingo B Autenrieth, Silke Peter, Matthias Willmann, Maria J G T Vehreschild, Lena M Biehl, Wichard Vogel, Daniela Dörfel, Axel Hamprecht, Harald Seifert, Ingo B Autenrieth, Silke Peter

Abstract

Background: The selection pressure exercised by antibiotic drugs is an important consideration for the wise stewardship of antimicrobial treatment programs. Treatment decisions are currently based on crude assumptions, and there is an urgent need to develop a more quantitative knowledge base that can enable predictions of the impact of individual antibiotics on the human gut microbiome and resistome.

Results: Using shotgun metagenomics, we quantified changes in the gut microbiome in two cohorts of hematological patients receiving prophylactic antibiotics; one cohort was treated with ciprofloxacin in a hospital in Tübingen and the other with cotrimoxazole in a hospital in Cologne. Analyzing this rich longitudinal dataset, we found that gut microbiome diversity was reduced in both treatment cohorts to a similar extent, while effects on the gut resistome differed. We observed a sharp increase in the relative abundance of sulfonamide antibiotic resistance genes (ARGs) by 148.1% per cumulative defined daily dose of cotrimoxazole in the Cologne cohort, but not in the Tübingen cohort treated with ciprofloxacin. Through multivariate modeling, we found that factors such as individual baseline microbiome, resistome, and plasmid diversity; liver/kidney function; and concurrent medication, especially virostatic agents, influence resistome alterations. Strikingly, we observed different effects on the plasmidome in the two treatment groups. There was a substantial increase in the abundance of ARG-carrying plasmids in the cohort treated with cotrimoxazole, but not in the cohort treated with ciprofloxacin, indicating that cotrimoxazole might contribute more efficiently to the spread of resistance.

Conclusions: Our study represents a step forward in developing the capability to predict the effect of individual antimicrobials on the human microbiome and resistome. Our results indicate that to achieve this, integration of the individual baseline microbiome, resistome, and mobilome status as well as additional individual patient factors will be required. Such personalized predictions may in the future increase patient safety and reduce the spread of resistance.

Trial registration: ClinicalTrials.gov, NCT02058888 . Registered February 10 2014.

Keywords: Antibiotic impact prediction; Antimicrobial resistance; Metagenomics study; Plasmid expansion; Resistome analysis.

Conflict of interest statement

MJGTV is a consultant to Alb Fils Kliniken GmbH, Berlin Chemie, MSD/Merck, and Astellas Pharma; has served at the speakers’ bureau of Astellas Pharma, Basilea, Gilead Sciences, Merck/MSD, Organobalance, and Pfizer; received research funding from 3M, Astellas Pharma, DaVolterra, Gilead Sciences, Merck/MSD, Morphochem, Organobalance, and Seres Therapeutics.

The other authors declare that they have no competing interests.

Figures

Fig. 1.
Fig. 1.
Antibiotic impact on the gut microbiome. Trajectories of richness, Shannon diversity, and Simpson’s evenness before treatment (T0) and at the end of the observation period (T3) are shown on phylum rank (a) and species rank (b) for both antibiotic treatments. Pink data points are measurements at T0, purple data points at T3. Boxplots indicate the distribution of data. The connecting magenta line shows the means at each time point and their development under treatment. The p value is displayed at the top of each box and indicates statistical significant differences between T0 and T3 within each treatment cohort (paired t-test). Under ciprofloxacin treatment, richness and Shannon diversity decrease significantly while Simpson’s evenness remains stable. In contrast, under cotrimoxazole, loss of richness and diversity is less pronounced and only significant on the phylum rank. c Violin plots illustrate the differences in baseline values between those patients with a positive baseline-endpoint comparison (BEC, green color) and those with a negative (orange color). The group size is displayed in the respective colors. Baseline species Shannon diversity was higher in the group of patients that lost diversity under cotrimoxazole, while patients with no decline or even an increase in diversity had a lower baseline diversity. The same was observed for species Simpson’s evenness under ciprofloxacin. d Based on multivariate regression modeling, the average percentage change per defined daily dose (DDD) is illustrated for each treatment cohort. Under both antibiotics, a loss in diversity was observed. However, no statistically significant difference was detected between both antibiotics. If an additional impact of concurrent medication was detected beside antibiotics in the multivariate models, this has been illustrated by different filling pattern. e Mean cumulative dose for antimicrobial agents in DDDs for the ciprofloxacin cohort and the cotrimoxazole cohort at each sampling time point (T0–T3). The colors indicate the drug classes, administered in either the ciprofloxacin or cotrimoxazole cohort (illustrated in brackets). The cumulative dose of ciprofloxacin was higher than the dose of cotrimoxazole. f Mean emergence and disappearance of species under antibiotic treatment in percentage compared to the species count at baseline. Frequent potentially pathogenic species are displayed. The number of patients with an emergence or disappearance of these species is shown in brackets
Fig. 2.
Fig. 2.
Antibiotic impact on the gut resistome. a Stacked bar chart of summed length-corrected relative abundances (LCRA) of major antimicrobial resistance gene (ARG) classes at baseline (T0) and over the treatment period (T1–T3). The following ARG classes are depicted: aminoglycosides (AGly), beta-lactamases (Bla), fluoroquinolones (Flq), glycopeptides (Gly), macrolide-lincosamide-streptogramin (MLS), nitroimidazoles (Ntmdz), phenicols (Phe), sulfonamides (Sul), tetracyclines (tet), and trimethoprim (Tmt). b Trajectories of antimicrobial resistance genes quantification by LCRA before treatment (T0) and at the end of the observation period (T3) are shown for both antibiotic treatments. Pink data points are measurements at T0, purple data points at T3. Boxplots indicate the distribution of data. The connecting magenta line shows the means at each time point and their development under treatment. The p value is displayed at the top of each box and indicates statistical significant differences between T0 and T3 within each treatment cohort (paired t-test). Trends for LCRA changes are prominent but do not reach statistical significance. c Two-dimensional kernel estimation density of square root transformed LCRA values of sulfonamide and trimethoprim ARG classes in relation to the administered cumulative antibiotic dose in defined daily doses (DDD). ARG LCRA rises significantly with increasing doses of cotrimoxazole, but not under ciprofloxacin. d Based on multivariate regression modeling, the average percentage change of ARG class LCRA per defined daily dose (DDD) is illustrated for each treatment cohort. Bonferroni-corrected statistically significant differences between both antibiotics (LR p < 0.002) are presented by single asterisks. Significant differences in antimicrobial selection pressure were observed for aminoglycoside, CTX-M, glycopeptide, MLS, nitroimidazole, phenicol, sulfonamide, and trimethoprim ARGs. If an additional impact of concurrent medication was detected beside antibiotics in the multivariate models, this has been illustrated by different filling pattern. e Fluoroquinolone resistance-mediating mutation frequencies increase under ciprofloxacin exposure in patient 512 comparing baseline (T0) and endpoint (T3)
Fig. 3.
Fig. 3.
Antibiotic impact on the gut plasmidome. a Trajectories of total plasmid abundance, plasmid abundance from proteobacteria, plasmid Shannon diversity, and plasmid Simpson’s evenness before treatment (T0) and at the end of the observation period (T3) are shown for both antibiotic treatments. Pink data points are measurements at T0, purple data points at T3. Boxplots indicate the distribution of data. The connecting magenta line shows the means at each time point and their development under treatment. The p value is displayed at the top of each box and indicates statistical significant differences between T0 and T3 within each treatment cohort (paired t-test). Total plasmid abundance, plasmid abundance from Proteobacteria, and plasmid diversity decreased significantly under ciprofloxacin treatment while plasmid evenness remained stable. In contrast, plasmids were not strongly affected by cotrimoxazole. b Based on multivariate regression modeling, the average percentage change of plasmid characteristics per defined daily dose (DDD) is illustrated for each treatment cohort. Bonferroni-corrected statistically significant differences between both antibiotics (LR p < 0.002) are presented by single asterisks. If an additional impact of concurrent medication was detected beside antibiotics in the multivariate models, this has been illustrated by a different filling pattern (checkerboard pattern = virostatic agents, horizontal stripes = antifungal agents, vertical stripes = virostatic and antifungal agents). Trends for plasmid evenness were significantly different, with a slight increase under ciprofloxacin and moderate decrease under cotrimoxazole. c, d The co-occurrence network displays the relationship between ARG-carrying plasmids from certain taxonomic origins and the ARG classes located on these plasmids at each sample collection time point for the ciprofloxacin cohort (c) and the cotrimoxazole cohort (d). The total plasmid-ARG content is expressed by the line width between plasmid origin and ARG class. The bar on the upper right part of each network row displays the scale of the total plasmid-ARG content (range 1–27). The diagrams in the lower right parts illustrate the Proteobacteria plasmid-ARG content for aminoglycoside, sulfonamide, trimethoprim ARGs, and beta-lactamase A enzymes. The y-axis ranges from 1 to 27 and displays the respective plasmid-ARG content. The ARG classes in the diagrams correspond to the colors of the networks and the legend at the bottom of the graph. Plasmids harboring ARGs from Proteobacteria expanded under cotrimoxazole, while ARG-containing plasmids from all origins declined under ciprofloxacin
Fig. 4.
Fig. 4.
Links between baseline gut microbiome and resistome alteration under antibiotic pressure. a Spearman’s rank correlation matrix revealed a positive correlation between the resistance score (indicating more positive antibiotic resistance gene selection in patients) and baseline microbiome and plasmid diversity. Pink-colored edgings indicate statistically significant correlation coefficients (p ≤ 0.05). b Scatter graphs with detailed illustration of the relation between baseline microbiome and plasmid diversity as well as between resistance score and baseline microbiome and plasmid diversity
Fig. 5.
Fig. 5.
Independent contributors that shape the gut resistome along with antibiotic treatment. The graph summarizes the concept of additional independent variables that impact the alterations of the gut resistome under antimicrobial selection pressure caused by antibiotic treatment

References

    1. Versporten A, Zarb P, Caniaux I, Gros MF, Drapier N, Miller M, Jarlier V, Nathwani D, Goossens H. Global PPSn: antimicrobial consumption and resistance in adult hospital inpatients in 53 countries: results of an internet-based global point prevalence survey. Lancet Glob Health. 2018;6:e619–e629. doi: 10.1016/S2214-109X(18)30186-4.
    1. Tacconelli E, Sifakis F, Harbarth S, Schrijver R, van Mourik M, Voss A, Sharland M, Rajendran NB, Rodriguez-Bano J, Group EP-NC-M Surveillance for control of antimicrobial resistance. Lancet Infect Dis. 2018;18:e99–e106. doi: 10.1016/S1473-3099(17)30485-1.
    1. Tackling drug-resistant infections globally: final report and recommendations []. Accessed 3 Aug 2018.
    1. Coyne MJ, Zitomersky NL, McGuire AM, Earl AM, Comstock LE. Evidence of extensive DNA transfer between bacteroidales species within the human gut. MBio. 2014;5:e01305–e01314. doi: 10.1128/mBio.01305-14.
    1. van Schaik W. The human gut resistome. Philos Trans R Soc Lond Ser B Biol Sci. 2015;370:20140087. doi: 10.1098/rstb.2014.0087.
    1. Willmann M, Peter S. Translational metagenomics and the human resistome: confronting the menace of the new millennium. J Mol Med (Berl) 2017;95:41–51. doi: 10.1007/s00109-016-1478-0.
    1. Buelow E, Bello Gonzalez TDJ, Fuentes S, de Steenhuijsen Piters WAA, Lahti L, Bayjanov JR, Majoor EAM, Braat JC, van Mourik MSM, Oostdijk EAN, et al. Comparative gut microbiota and resistome profiling of intensive care patients receiving selective digestive tract decontamination and healthy subjects. Microbiome. 2017;5:88. doi: 10.1186/s40168-017-0309-z.
    1. Buelow E, Gonzalez TB, Versluis D, Oostdijk EA, Ogilvie LA, van Mourik MS, Oosterink E, van Passel MW, Smidt H, D'Andrea MM, et al. Effects of selective digestive decontamination (SDD) on the gut resistome. J Antimicrob Chemother. 2014;69:2215–2223. doi: 10.1093/jac/dku092.
    1. Gibson MK, Wang B, Ahmadi S, Burnham CA, Tarr PI, Warner BB, Dantas G. Developmental dynamics of the preterm infant gut microbiota and antibiotic resistome. Nat Microbiol. 2016;1:16024. doi: 10.1038/nmicrobiol.2016.24.
    1. Willmann M, El-Hadidi M, Huson DH, Schutz M, Weidenmaier C, Autenrieth IB, Peter S. Antibiotic selection pressure determination through sequence-based metagenomics. Antimicrob Agents Chemother. 2015;59:7335–7345. doi: 10.1128/AAC.01504-15.
    1. Imhann F, Bonder MJ, Vich Vila A, Fu J, Mujagic Z, Vork L, Tigchelaar EF, Jankipersadsing SA, Cenit MC, Harmsen HJ, et al. Proton pump inhibitors affect the gut microbiome. Gut. 2016;65:740–748. doi: 10.1136/gutjnl-2015-310376.
    1. Hamprecht A, Rohde AM, Behnke M, Feihl S, Gastmeier P, Gebhardt F, Kern WV, Knobloch JK, Mischnik A, Obermann B, et al. Colonization with third-generation cephalosporin-resistant Enterobacteriaceae on hospital admission: prevalence and risk factors. J Antimicrob Chemother. 2016;71:2957–2963. doi: 10.1093/jac/dkw216.
    1. Gupta SK, Padmanabhan BR, Diene SM, Lopez-Rojas R, Kempf M, Landraud L, Rolain JM. ARG-ANNOT, a new bioinformatic tool to discover antibiotic resistance genes in bacterial genomes. Antimicrob Agents Chemother. 2014;58:212–220. doi: 10.1128/AAC.01310-13.
    1. Redgrave LS, Sutton SB, Webber MA, Piddock LJ. Fluoroquinolone resistance: mechanisms, impact on bacteria, and role in evolutionary success. Trends Microbiol. 2014;22:438–445. doi: 10.1016/j.tim.2014.04.007.
    1. Faron ML, Ledeboer NA, Buchan BW. Resistance mechanisms, epidemiology, and approaches to screening for vancomycin-resistant Enterococcus in the health care setting. J Clin Microbiol. 2016;54:2436–2447. doi: 10.1128/JCM.00211-16.
    1. Smillie CS, Smith MB, Friedman J, Cordero OX, David LA, Alm EJ. Ecology drives a global network of gene exchange connecting the human microbiome. Nature. 2011;480:241–244. doi: 10.1038/nature10571.
    1. Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, Mujagic Z, Vila AV, Falony G, Vieira-Silva S, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352:565–569. doi: 10.1126/science.aad3369.
    1. Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, Brochado AR, Fernandez KC, Dose H, Mori H, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555:623–628. doi: 10.1038/nature25979.
    1. Imhann F, Vich Vila A, Bonder MJ, Lopez Manosalva AG, Koonen DPY, Fu J, Wijmenga C, Zhernakova A, Weersma RK. The influence of proton pump inhibitors and other commonly used medication on the gut microbiota. Gut Microbes. 2017;8:351–358. doi: 10.1080/19490976.2017.1284732.
    1. Taur Y, Jenq RR, Perales MA, Littmann ER, Morjaria S, Ling L, No D, Gobourne A, Viale A, Dahi PB, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124:1174–1182. doi: 10.1182/blood-2014-02-554725.
    1. Weber D, Jenq RR, Peled JU, Taur Y, Hiergeist A, Koestler J, Dettmer K, Weber M, Wolff D, Hahn J, et al. Microbiota disruption induced by early use of broad-spectrum antibiotics is an independent risk factor of outcome after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2017;23:845–852. doi: 10.1016/j.bbmt.2017.02.006.
    1. Zarrinpar A, Chaix A, Xu ZZ, Chang MW, Marotz CA, Saghatelian A, Knight R, Panda S. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism. Nat Commun. 2018;9:2872. doi: 10.1038/s41467-018-05336-9.
    1. van Vliet MJ, Tissing WJ, Dun CA, Meessen NE, Kamps WA, de Bont ES, Harmsen HJ. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin Infect Dis. 2009;49:262–270. doi: 10.1086/599346.
    1. Gillings MR. Integrons: past, present, and future. Microbiol Mol Biol Rev. 2014;78:257–277. doi: 10.1128/MMBR.00056-13.
    1. Abeles SR, Jones MB, Santiago-Rodriguez TM, Ly M, Klitgord N, Yooseph S, Nelson KE, Pride DT. Microbial diversity in individuals and their household contacts following typical antibiotic courses. Microbiome. 2016;4:39. doi: 10.1186/s40168-016-0187-9.
    1. Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40:373–383. doi: 10.1016/0021-9681(87)90171-8.
    1. Teasdale G, Jennett B. Assessment of coma and impaired consciousness. A practical scale. Lancet. 1974;2:81–84. doi: 10.1016/S0140-6736(74)91639-0.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, Lesin VM, Nikolenko SI, Pham S, Prjibelski AD, et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19:455–477. doi: 10.1089/cmb.2012.0021.
    1. Menzel P, Ng KL, Krogh A. Fast and sensitive taxonomic classification for metagenomics with kaiju. Nat Commun. 2016;7:11257. doi: 10.1038/ncomms11257.
    1. Buchfink B, Xie C, Huson DH. Fast and sensitive protein alignment using DIAMOND. Nat Methods. 2015;12:59–60. doi: 10.1038/nmeth.3176.
    1. Bush K, Jacoby GA. Updated functional classification of beta-lactamases. Antimicrob Agents Chemother. 2010;54:969–976. doi: 10.1128/AAC.01009-09.
    1. Hooper DC, Jacoby GA. Mechanisms of drug resistance: quinolone resistance. Ann N Y Acad Sci. 2015;1354:12–31. doi: 10.1111/nyas.12830.
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Krawczyk PS, Lipinski L, Dziembowski A. PlasFlow: predicting plasmid sequences in metagenomic data using genome signatures. Nucleic Acids Res. 2018;46:e35. doi: 10.1093/nar/gkx1321.
    1. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics. 2014;30:2068–2069. doi: 10.1093/bioinformatics/btu153.
    1. Fu L, Niu B, Zhu Z, Wu S, Li W. CD-HIT: accelerated for clustering the next-generation sequencing data. Bioinformatics. 2012;28:3150–3152. doi: 10.1093/bioinformatics/bts565.
    1. Camacho C, Coulouris G, Avagyan V, Ma N, Papadopoulos J, Bealer K, Madden TL. BLAST+: architecture and applications. BMC Bioinformatics. 2009;10:421. doi: 10.1186/1471-2105-10-421.
    1. Willmann M, Vehreschild MJGT, Biehl LM, Vogel W, Dörfel D, Hamprecht A, Seifert H, Autenrieth IB, Peter S. Impact of antibiotics on the human gut microbiome and resistome. European Nucleotide Archive study accession number PRJEB28058 . Accessed 16 Aug 2019.

Source: PubMed

3
Sottoscrivi