Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Longitudinal changes in the nasopharyngeal resistome of South African infants using shotgun metagenomic sequencing

  • Rendani I. Manenzhe ,

    Roles Data curation, Formal analysis, Investigation, Validation, Visualization, Writing – original draft, Writing – review & editing

    manenzhe.r.i@gmail.com

    Affiliation Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa

  • Felix S. Dube,

    Roles Conceptualization, Data curation, Funding acquisition, Investigation, Methodology, Supervision, Writing – original draft, Writing – review & editing

    Affiliations Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa, Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Cape Town, South Africa

  • Meredith Wright,

    Roles Data curation, Formal analysis, Software, Validation, Visualization, Writing – review & editing

    Affiliation J. Craig Venter Institute, Rockville, MD, United States of America

  • Katie Lennard,

    Roles Data curation, Formal analysis, Software, Validation, Visualization, Writing – review & editing

    Affiliation Division of Computational Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa

  • Heather J. Zar,

    Roles Conceptualization, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Writing – review & editing

    Affiliation Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital and MRC unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa

  • Stephanie Mounaud,

    Roles Data curation, Formal analysis, Software, Validation, Visualization, Writing – review & editing

    Affiliation J. Craig Venter Institute, Rockville, MD, United States of America

  • William C. Nierman,

    Roles Conceptualization, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Writing – review & editing

    Affiliation J. Craig Venter Institute, Rockville, MD, United States of America

  • Mark P. Nicol,

    Roles Conceptualization, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing – review & editing

    Affiliations Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa, Division of Infection and Immunity, University of Western Australia, Perth, Australia

  • Clinton Moodley

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing – review & editing

    Affiliations Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa, National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa

Abstract

Introduction

Nasopharyngeal (NP) colonization with antimicrobial-resistant bacteria is a global public health concern. Antimicrobial-resistance (AMR) genes carried by the resident NP microbiota may serve as a reservoir for transfer of resistance elements to opportunistic pathogens. Little is known about the NP antibiotic resistome. This study longitudinally investigated the composition of the NP antibiotic resistome in Streptococcus-enriched samples in a South African birth cohort.

Methods

As a proof of concept study, 196 longitudinal NP samples were retrieved from a subset of 23 infants enrolled as part of broader birth cohort study. These were selected on the basis of changes in serotype and antibiogram over time. NP samples underwent short-term enrichment for streptococci prior to total nucleic acid extraction and whole metagenome shotgun sequencing (WMGS). Reads were assembled and aligned to pneumococcal reference genomes for the extraction of streptococcal and non-streptococcal bacterial reads. Contigs were aligned to the Antibiotic Resistance Gene-ANNOTation database of acquired AMR genes.

Results

AMR genes were detected in 64% (125/196) of the samples. A total of 329 AMR genes were detected, including 36 non-redundant genes, ranging from 1 to 14 genes per sample. The predominant AMR genes detected encoded resistance mechanisms to beta-lactam (52%, 172/329), macrolide-lincosamide-streptogramin (17%, 56/329), and tetracycline antibiotics (12%, 38/329). MsrD, ermB, and mefA genes were only detected from streptococcal reads. The predominant genes detected from non- streptococcal reads included blaOXA-60, blaOXA-22, and blaBRO-1. Different patterns of carriage of AMR genes were observed, with only one infant having a stable carriage of mefA, msrD and tetM over a long period.

Conclusion

This study demonstrates that WMGS can provide a broad snapshot of the NP resistome and has the potential to provide a comprehensive assessment of resistance elements present in this niche.

Introduction

Infection with antibiotic-resistant bacteria is a major public health concern due to the limited availability of new treatment options [1]. Increasing antibiotic resistance has been noted in respiratory tract bacterial pathogens which are capable of causing life-threatening infections [2,3]. The upper respiratory tract, including the nasopharynx, is the reservoir for many respiratory pathogens and may also serve as a source for the transfer of antimicrobial-resistance (AMR) genes from non-pathogenic to pathogenic bacteria [4].

Pathogens which commonly colonize the upper airways include Streptococcus pneumoniae (the pneumococcus), Staphylococcus aureus, Haemophilus influenzae, Neisseria meningitidis, and several Gram-negative bacilli [57]. The pneumococcus and H. influenzae are among the leading causes of bacterial respiratory tract infections in young children [8,9]. Asymptomatic NP carriage of pneumococci is prevalent among infants and often precedes the development of disease [4,10]. Drug-resistant pneumococci may cause difficult-to-treat infections, associated with increased morbidity and mortality [4,10]. In many cases, antibiotic resistance results from horizontal gene transfer (HGT) of a mobile genetic element, or uptake of free DNA from the surrounding environment [11,12], which is of particular importance as pneumococci are naturally competent [13].

Culture-based methods only allow for the detection of certain AMR genes in viable, culturable bacteria, and are therefore unable to completely characterise the resistome in a particular niche [14]. An alternative approach for detection of AMR genes is whole metagenome shotgun sequencing (WMGS) of DNA extracted directly from samples [15,16]. The majority of WMGS studies of the antibiotic resistome have focused on the human gut resistome [1620]. To our knowledge there are no published studies of the NP antibiotic resistome.

We have previously reported changes in NP pneumococcal antibiotic-resistance in infants studied longitudinally over the first year of life, using culture-based susceptibility testing [21]. Here we further characterise a subset of these samples using WMGS to demonstrate proof of concept for resistome analysis of upper respiratory tract samples.

Material and methods

This study was nested within a birth-cohort study which investigates the within-host micro-evolution of naturally acquired pneumococci in 800 infants. NP swabs were longitudinally collected fortnightly from birth until twelve months, in this high-carriage African setting [22]. As a proof of concept, we selected obtained 196 NP swabs from 23 infants, on the basis of longitudinal changes in serotype and antibiogram over the first year of life, for shotgun metagenomic sequencing [21]. These NP swabs were stored in 1 ml skim milk-tryptone-glucose-glycerol (STGG) medium as previously described [23]. The study was approved by the Faculty of Health Sciences Human Research Ethics Committee of the University of Cape Town (reference numbers: 235/2016 and 401/2009) and written informed consent obtained from all parents or legal guardians at enrolment.

The NP-STGG samples were enriched as previously described, with minor modifications [24]. Briefly, 200 μl of an NP-STGG sample was transferred to 6 ml Todd-Hewitt Broth (without antibiotics), containing 0.5% yeast extract and 17% fetal bovine serum. The broth was incubated at 37°C with 5% CO2, without shaking for 6 hours, then centrifuged at 9000 rpm for 10 minutes at 4°C. Total nucleic acid extraction was performed on the collected pellet using the QIAsymphony SP automated platform (Qiagen, Hilden, Germany) with the QIAsymphony Virus/Bacteria Mini Kit (Cat. No. 931036) following the manufacturer’s instructions. Nucleic acid concentrations and purity were determined by UV spectrophotometry using the NanoDrop® ND-100 (Thermo Fishers Scientific, Waltham, USA).

Total nucleic acid was subjected to shotgun sequencing on the MiSeq platform using the MiSeq Reagent Kit v3 (600-cycle) (Illumina, San Diego, USA) at the J. Craig Venter Institute, Rockville, USA. Metagenomic DNA sequencing and assembly protocols have previously been described [24]. Reads were assembled using metaSPAdes [25], and aligned to a database containing Streptococcus pneumoniae complete genomes in order to re-construct the pneumococcal genomes and extract all the streptococcal contigs (genes identified using pneumococcal references may have come from other streptococcus species due to high level of genetic relatedness thus referring to these as streptococcal contigs). Bacterial contigs not mapping to pneumococcal genomes were regarded as non-streptococcal contigs and were separately extracted for further analysis [24].

Screening for AMR genes present in the selected NP samples was performed on the assembled contigs for both streptococcal and non- streptococcal contig datasets. Contigs were aligned to the Antibiotic Resistance Gene-ANNOTation (ARG-ANNOT) database of acquired AMR genes. To assess the reproducibility of the resistome analysis, all bacterial contigs as well as the streptococcal and non-streptococcal contigs were separately aligned against the ARG-ANNOT database. To increase sensitivity for identifying novel genes or genotypes with low levels of similarity to the reference genes, less stringent criteria were used [26]. A sequence with ≥ 90% identity [27], with an alignment coverage length of ≥ 25% to the reference gene sequence was designated as an AMR gene [26]. The AMR genes were manually confirmed.

Statistical analyses were performed using STATA (Stata Corporation, College Station, TX). Chi-square and Fisher’s exact tests were used to compare the differences in the proportion of samples with AMR genes. A p-value of <0.05 was considered statistically significant.

Results

Participants and metagenomic sample characteristics

A total of 196 longitudinal NP samples were selected from 23 infants, with an average of 9 selected NP samples per infant (range, 4–21 samples). The age at which the NP samples were collected spanned the first year of life with an average age of 15 weeks. Four of the 23 infants were born via caesarean section (Table 1). Eight infants were born to HIV infected mothers, but none of the infants were infected. Antibiotics were administered to 6 out of 7 infants who had severe or non-severe lower respiratory tract infection (LRTI) during the first year of life (ages, 0–52 weeks) (Table 1).

Nasopharyngeal resistome characteristics

The average depth of coverage of the detected AMR genes from all contigs was 26X (range 1 – 862X). A total of 329 AMR genes were detected in 64% (125/196) of the selected NP samples. Among these, 57% (188/329) were detected at ≥ 90% identity and ≥ 25% gene coverage while only 30% (97/329) were detected at the more stringent cut-off of ≥ 90% identity and ≥ 80% gene coverage (Table 2). The number of resistance genes detected per sample ranged from 1–14 genes (Fig 1), and included 36 non-redundant genes (Fig 2). AMR genes were detected in at least one sample from each of the 23 selected infants (Fig 3). The same types and number of AMR genes were detected from all bacterial contigs combined compared to those detected from non-streptococcal and streptococcal contigs separately. The most common resistance genes detected were those conferring resistance to beta-lactams (52%, 172/329), macrolide-lincosamide-streptogramin antibiotics (MLS) (17%, 56/329), and tetracyclines (12%, 38/329) (Table 2 and Fig 2). A high number of AMR genes conferring resistance to MLS (n = 38), tetracyclines (n = 25), aminoglycosides (n = 17), fluoroquinolones (n = 4), and trimethoprim (n = 3) were detected at a cut-off of 90% identity over 50% gene coverage (Table 2). Different patterns of carriage of AMR genes were observed, with only one infant having a stable carriage of mefA, msrD and tetM over a long period (Fig 3).

thumbnail
Fig 1. Distribution of antimicrobial-resistance (AMR) genes within 196 longitudinal NP samples selected from 23 infants.

https://doi.org/10.1371/journal.pone.0231887.g001

thumbnail
Fig 2. Frequency of antimicrobial-resistance (AMR) genes within 196 longitudinal NP samples selected from 23 infants.

MLS-Macrolide-lincosamide-streptogramin. (*) AMR genes detected from streptococcal contigs. (**) AMR gene detected from streptococcal contigs in 23 out of 24 samples.

https://doi.org/10.1371/journal.pone.0231887.g002

thumbnail
Fig 3. Antibiotic-resistance genes detected from 196 longitudinal NP samples selected from 23 infants (assigned 1 to 23).

(A) Streptococcal resistome. (B) Non-streptococcal resistome. Blue line indicates samples collected before antibiotic administration, for lower respiratory tract infection. Red line indicates samples collected after antibiotic administration, for lower respiratory tract infection. MLS- Macrolide-lincosamide-streptogramin, AMR- Antimicrobial resistance.

https://doi.org/10.1371/journal.pone.0231887.g003

thumbnail
Table 2. Detection of AMR genes conferring resistance to various classes of antibiotics from 196 NP samples using different stringency criteria.

https://doi.org/10.1371/journal.pone.0231887.t002

Streptococcal resistome

Shotgun sequencing detected streptococcal reads in all 174 samples that were culture positive for S. pneumoniae. Seventy AMR genes (four non-redundant genes) were detected from streptococcal contigs; the average depth of coverage was 103X (range 1 – 411X). MLS and tetracycline resistance genes were the only genes detected from streptococcal contigs. The most commonly detected gene was tetM (n = 23), followed by msrD (n = 22), mefA (n = 21), and ermB (n = 4). MsrD, ermB, and mefA genes were only identified from streptococcal contigs. The combination of msrD, mefA and tetM genes was detected in 10 samples from 3 infants and all were identified on the same contig in 9 out of 10 samples.

Non-streptococcal resistome

A total of 259 AMR genes were detected from non-streptococcal contigs with an average coverage depth of 10X (range, 1 – 862X). Nine types of AMR genes were detected from non-streptococcal contigs (Fig 3), with beta-lactam resistance genes (66%, 172/259) being the most commonly detected genes (Table 2 and Fig 2). The most commonly detected beta-lactam resistance gene was blaOXA-60, followed by blaOXA-22, blaBRO-1, blaTEM, blaBRO-2, cfxA, and blaZ (Fig 2). The fluoroquinolone resistance gene norA was detected in six samples from four infants (Fig 3).

Association between antibiotic use and the NP resistome

No significant difference was observed between the presence of AMR genes in samples collected before and after the treatment of LRTI (Fig 3). A large proportion of AMR genes (69% 227/329) were detected in samples from a subset of eight infants (Fig 3).

Discussion

This proof-of-concept study investigated the composition of the NP antibiotic resistome in an intensively sampled South African birth cohort. 329 AMR genes were detected across 64% of the selected NP samples using targeted enrichment culture and shotgun metagenomic sequencing. We detected the same types and number of AMR genes from all contigs combined compared to those detected from non-streptococcal and streptococcal contigs separately, suggesting that our resistome analysis was reliable. The average depth of coverage for the resistance genes from streptococcal contigs (103X) was higher than that from non- streptococcal contigs (10X). This observation is likely due to the short streptococcal enrichment culture step using Todd Hewitt broth (without antibiotics) [24].

We detected resistance genes using a lower stringency criteria of ≥ 90% identity over 25% coverage of the reference gene, which has been shown to be more reliable, than more stringent criteria, in detecting AMR genes [27]. Yang et al., reported a high accuracy (99%) for detecting AMR genes using these less stringent parameters in metagenomic analysis [28]. In the current study, at least 25% gene coverage was used [26], and this cut-off was higher than the suggested coverage of ≥ 25 amino acids [28]. Only 30% of the AMR genes were detected using ≥ 90% identity over 80% coverage of the reference gene [26]. The more stringent parameters detected mainly MLS, tetracycline, and aminoglycoside resistance genes which are frequently carried by Streptococcus species, presumably due to the higher depth of coverage as a result of the enrichment step [29,30].

We observed differences in the types and numbers of AMR genes identified from streptococcal and non-streptococcal contigs. With the exception of one sample in which tetM was detected; tetM, msrD, ermB, and mefA genes were only detected from streptococcal contigs (Fig 3). The msrD, ermB, and mefA genes are most frequently detected among streptococcal isolates [31]. Pneumococci which are resistant to MLS antibiotics are commonly also resistant to tetracycline due to the insertion of an MLS gene into the conjugative transposons of the Tn916 family, which typically carry the tetM gene [32,33]. Although transposons were not evaluated in the current study, msrD and tetM genes were commonly identified on the same contig (9/10 samples) suggesting they could be carried on the same transposon [33].

The predominant AMR genes detected from non-streptococcal contigs were beta-lactamase genes, specifically blaOXA-60, blaOXA-22, blaBRO-1, and blaTEM. All blaTEM gene variants detected in the current study encode narrow spectrum beta-lactamase enzymes, and these have previously been detected in the Enterobacteriaceae, H. influenzae, and Neisseria gonorrhoea [34]. The blaBRO-1 gene was more commonly detected than blaBRO-2 (Fig 2), both are typically found among Moraxella catarrhalis isolates, with blaBRO-1 more prevalent than blaBRO-2 in M. catarrhalis [35].

blaOXA-60 and blaOXA-22 genes, encoding the chromosomal and inducible class D beta-lactamases have only been described in Ralstonia pickettii or R. mannitolilytica [3640]. Ralstonia sp. are Gram-negative, non-fermentative bacilli, commonly isolated from the respiratory tract and their carriage among infants in this study warrants further investigation [41]. OXA-22 is an oxacillinase with the ability to hydrolyse narrow-spectrum beta-lactams [40]. Unlike OXA-22, the hydrolysis spectrum of OXA-60, although narrow, includes carbapenems. Whilst R. pickettii infrequently causes infections, the potential for transfer of this gene to other NP bacteria should be studied [36,39].

The norA gene, which encodes a fluoroquinolone efflux transporter protein, has been described mainly in Staphylococcus aureus and can render resistance to both fluoroquinolones and other classes of antibiotics with dissimilar structures [42,43].

Beta-lactamase genes were the most commonly detected resistance genes in the current study. Amoxicillin, a beta-lactam antibiotic, was the most commonly prescribed antibiotic for both acute and ambulatory care in children in this study, which could explain the high number and types of beta-lactamase genes detected [44].

There were several limitations to the current study. Firstly, the enrichment culture for streptococci altered the composition of the NP resistome and the prevalence of the different AMR genes detected may therefore differ from that found in directly tested samples. Secondly, the purposively selected sample set is unlikely to be broadly representative of infants in this study. Thirdly, the reference database used for the resistome analysis is not comprehensive, and excludes chromosomal mutations associated with resistance. Penicillin resistance associated with pbp gene mutations, such as pbp1a and pbp2x, and trimethoprim sulphamethoxazole resistance, associated with folA I100L substitutions and folP insertions, would not be detected using this database, and further work will be done to characterise these associations.

This study demonstrates that WMGS can provide a broad snapshot of the NP resistome. Recent work has highlighted that the nasopharynx is a conducive environment for the exchange of AMR genes between related Streptococcus species responsible for respiratory tract infections in children [45]. WMGS has the potential to provide a comprehensive assessment of all resistance elements present in this niche.

Acknowledgments

We thank Anne von Gottberg, Linda de Gouveia and the staff of the Centre for Respiratory Diseases and Meningitis (CRDM), National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service for training, sharing of standard operating procedures, supplying control isolates and performing Quellung serotyping. We thank the clinical research staff involved in the Drakenstein Child Lung Health Study for collection of samples and the children and parents for participating in the study. We further wish to thank Charmaine Barthus, Widaad Zemanay, Whitney Barnett, and the Drakenstein Child Lung Health Study team for their help and technical assistance.

References

  1. 1. Carlet J, Jarlier V, Harbarth S, Voss A, Goossens H, Pittet D. Ready for a world without antibiotics? The Pensières Antibiotic Resistance Call to Action. Antimicrob Resist Infect Control. 2012 Jan 14;1(1):11. pmid:22958833
  2. 2. Brook I, Gober AE. Antimicrobial resistance in the nasopharyngeal flora of children with acute maxillary sinusitis and maxillary sinusitis recurring after amoxicillin therapy. J Antimicrob Chemother. 2004 Feb;53(2):399–402. pmid:14729759
  3. 3. Faden H, Stanievich J, Brodsky L, Bernstein J, Ogra PL. Changes in nasopharyngeal flora during otitis media of childhood. Pediatr Infect Dis J. 1990 Sep;9(9):623–6. pmid:2122410
  4. 4. Wolf B, Rey LC, Moreira LB, Milatovic D, Fleer A, Verhoef J, et al. Carriage of gram-negative bacilli in young Brazilian children with community-acquired pneumonia. Int J Infect Dis. 2001;5(3):155–9. pmid:11724673
  5. 5. García-Rodríguez JA, Fresnadillo Martínez MJ. Dynamics of nasopharyngeal colonization by potential respiratory pathogens. J Antimicrob Chemother. 2002 Dec;50 Suppl S:59–73.
  6. 6. Lima ABM, de Oliveira Leão LSN, Oliveira LS da C, Pimenta FC. Nasopharyngeal Gram-Negative bacilli colonization in brazilian children attending day-care centers. Braz J Microbiol. 2010 Jan;41(1):24–7. pmid:24031458
  7. 7. Leiberman A, Dagan R, Leibovitz E, Yagupsky P, Fliss DM. The bacteriology of the nasopharynx in childhood. Int J Pediatr Otorhinolaryngol. 1999 Oct 5;49 Suppl 1:S151–3.
  8. 8. Berman S. Epidemiology of acute respiratory infections in children of developing countries. Rev Infect Dis. 1991 Jan;13 Suppl 6:S454–62.
  9. 9. Sazawal S, Black RE. Meta-analysis of intervention trials on case-management of pneumonia in community settings. Lancet (London, England). 1992 Aug 29;340(8818):528–33.
  10. 10. De Lencastre H, Tomasz A. From ecological reservoir to disease: the nasopharynx, day-care centres and drug-resistant clones of Streptococcus pneumoniae. J Antimicrob Chemother. 2002 Dec;50 Suppl S:75–81. pmid:12556436
  11. 11. Aminov RI, Mackie RI. Evolution and ecology of antibiotic resistance genes. FEMS Microbiol Lett. 2007 Jun;271(2):147–61. pmid:17490428
  12. 12. Salyers AA, Gupta A, Wang Y. Human intestinal bacteria as reservoirs for antibiotic resistance genes. Trends Microbiol. 2004 Sep;12(9):412–6. pmid:15337162
  13. 13. Li G, Liang Z, Wang X, Yang Y, Shao Z, Li M, et al. Addiction of Hypertransformable Pneumococcal Isolates to Natural Transformation for In Vivo Fitness and Virulence. Camilli A, editor. Infect Immun. 2016 Jun;84(6):1887–901. pmid:27068094
  14. 14. Lingling Wang ZY. Antibiotic Resistant Bacteria—A Continuous Challenge in the New Millennium. Pana M, editor. InTech; 2012. 369–484 p.
  15. 15. Jalali S, Kohli S, Latka C, Bhatia S, Vellarikal SK, Sivasubbu S, et al. Screening currency notes for microbial pathogens and antibiotic resistance genes using a shotgun metagenomic approach. PLoS One. 2015 Jan;10(6):e0128711. pmid:26035208
  16. 16. van Schaik W. The human gut resistome. Philos Trans R Soc B Biol Sci. 2015 Apr 27;370(1670):20140087–20140087.
  17. 17. Buelow E, Gonzalez TB, Versluis D, Oostdijk EAN, Ogilvie LA, van Mourik MSM, et al. Effects of selective digestive decontamination (SDD) on the gut resistome. J Antimicrob Chemother. 2014 Aug;69(8):2215–23. pmid:24710024
  18. 18. Forslund K, Sunagawa S, Coelho LP, Bork P. Metagenomic insights into the human gut resistome and the forces that shape it. Bioessays. 2014 Mar;36(3):316–29. pmid:24474281
  19. 19. Hu Y, Yang X, Lu N, Zhu B. The abundance of antibiotic resistance genes in human guts has correlation to the consumption of antibiotics in animal. Gut Microbes. 2014 Jan;5(2):245–9. pmid:24637798
  20. 20. Pérez-Cobas AE, Artacho A, Knecht H, Ferrús ML, Friedrichs A, Ott SJ, et al. Differential effects of antibiotic therapy on the structure and function of human gut microbiota. PLoS One. 2013 Jan;8(11):e80201. pmid:24282523
  21. 21. Manenzhe RI, Moodley C, Abdulgader SM, Robberts FJL, Zar HJ, Nicol MP, et al. Nasopharyngeal Carriage of Antimicrobial-Resistant Pneumococci in an Intensively Sampled South African Birth Cohort. Front Microbiol. 2019 Mar 27;10:610. pmid:30972052
  22. 22. Zar HJ, Barnett W, Myer L, Stein DJ, Nicol MP. Investigating the early-life determinants of illness in Africa: the Drakenstein Child Health Study. Thorax. 2015 Jun; 70:592–4. pmid:25228292
  23. 23. Dube FS, Ramjith J, Gardner-Lubbe S, Nduru P, Robberts FJL, Wolter N, et al. Longitudinal characterization of nasopharyngeal colonization with Streptococcus pneumoniae in a South African birth cohort post 13-valent pneumococcal conjugate vaccine implementation. Sci Rep. 2018 Dec 21;8(1):12497. pmid:30131607
  24. 24. Wright MS, McCorrison J, Gomez AM, Beck E, Harkins D, Shankar J, et al. Strain Level Streptococcus Colonization Patterns during the First Year of Life. Front Microbiol. 2017;8:1661. pmid:28932211
  25. 25. Nurk S, Meleshko D, Korobeynikov A, Pevzner PA. metaSPAdes: a new versatile metagenomic assembler. Genome Res. 2017 May;27(5):824–34. pmid:28298430
  26. 26. Gupta SK, Padmanabhan BR, Diene SM, Lopez-Rojas R, Kempf M, Landraud L, et al. ARG-ANNOT, a New Bioinformatic Tool To Discover Antibiotic Resistance Genes in Bacterial Genomes. Antimicrob Agents Chemother. 2014 Jan;58(1):212–20. pmid:24145532
  27. 27. Kristiansson E, Fick J, Janzon A, Grabic R, Rutgersson C, Weijdegård B, et al. Pyrosequencing of Antibiotic-Contaminated River Sediments Reveals High Levels of Resistance and Gene Transfer Elements. Rodriguez-Valera F, editor. PLoS One. 2011 Feb 16;6(2):e17038. pmid:21359229
  28. 28. Yang Y, Li B, Ju F, Zhang T. Exploring Variation of Antibiotic Resistance Genes in Activated Sludge over a Four-Year Period through a Metagenomic Approach. Environ Sci Technol. 2013 Sep 17;47(18):10197–205. pmid:23919449
  29. 29. Reinert RR. The antimicrobial resistance profile of Streptococcus pneumoniae. Clin Microbiol Infect. 2009 Apr 1;15:7–11. pmid:19366363
  30. 30. Poyart C, Jardy L, Quesne G, Berche P, Trieu-Cuot P. Genetic basis of antibiotic resistance in Streptococcus agalactiae strains isolated in a French hospital. Antimicrob Agents Chemother. 2003 Feb 1;47(2):794–7. pmid:12543695
  31. 31. Xu X, Cai L, Xiao M, Kong F, Oftadeh S, Zhou F, et al. Distribution of serotypes, genotypes, and resistance determinants among macrolide-resistant Streptococcus pneumoniae isolates. Antimicrob Agents Chemother. 2010 Mar 1;54(3):1152–9. pmid:20065057
  32. 32. McDougal LK, Tenover FC, Lee LN, Rasheed JK, Patterson JE, Jorgensen JH, et al. Detection of Tn917-like sequences within a Tn916-like conjugative transposon (Tn3872) in erythromycin-resistant isolates of Streptococcus pneumoniae. Antimicrob Agents Chemother. 1998 Sep 1;42(9):2312–8. pmid:9736555
  33. 33. Clewell DB, Flannagan SE, Jaworski DD, Clewell DB. Unconstrained bacterial promiscuity: the Tn916–Tn1545 family of conjugative transposons. Trends Microbiol. 1995 Jun 1;3(6):229–36. pmid:7648031
  34. 34. Paterson DL, Bonomo RA. Extended-spectrum beta-lactamases: a clinical update. Clin Microbiol Rev. 2005 Oct 1;18(4):657–86. pmid:16223952
  35. 35. Khan MA, Northwood JB, Levy F, Verhaegh SJC, Farrell DJ, Van Belkum A, et al. bro β-lactamase and antibiotic resistances in a global cross-sectional study of Moraxella catarrhalis from children and adults. J Antimicrob Chemother. 2010 Jan 1;65(1):91–7. pmid:19889789
  36. 36. Suzuki M, Nishio H, Asagoe K, Kida K, Suzuki S, Matsui M, et al. Genome Sequence of a Carbapenem-Resistant Strain of Ralstonia mannitolilytica. Genome Announc. 2015 Jun 25;3(3):e00405–15. pmid:25953190
  37. 37. Girlich D, Kolb A, Naas T, Nordmann P. Characterization of regulatory element Rp3 of regulation of Î2-lactamases from Ralstonia pickettii. FEMS Microbiol Lett. 2009 Dec;301(1):50–6. pmid:19817868
  38. 38. Girlich D, Naas T, Nordmann P. Regulation of class D beta-lactamase gene expression in Ralstonia pickettii. Microbiology. 2006 Sep 1;152(9):2661–72.
  39. 39. Girlich D, Naas T, Nordmann P. OXA-60, a Chromosomal, Inducible, and Imipenem-Hydrolyzing Class D -Lactamase from Ralstonia pickettii. Antimicrob Agents Chemother. 2004 Nov 1;48(11):4217–25. pmid:15504844
  40. 40. Nordmann P, Poirel L, Kubina M, Casetta A, Naas T. Biochemical-genetic characterization and distribution of OXA-22, a chromosomal and inducible class D beta-lactamase from Ralstonia (Pseudomonas) pickettii. Antimicrob Agents Chemother. 2000 Aug;44(8):2201–4. pmid:10898703
  41. 41. Daxboeck F, Stadler M, Assadian O, Marko E, Hirschl AM, Koller W. Characterization of clinically isolated Ralstonia mannitolilytica strains using random amplification of polymorphic DNA (RAPD) typing and antimicrobial sensitivity, and comparison of the classification efficacy of phenotypic and genotypic assays. J Med Microbiol. 2005 Jan 1;54(1):55–61.
  42. 42. Neyfakh AA, Borsch CM, Kaatz GW. Fluoroquinolone resistance protein NorA of Staphylococcus aureus is a multidrug efflux transporter. Antimicrob Agents Chemother. 1993 Jan 1;37(1):128–9. pmid:8431010
  43. 43. Schmitz F-J, Köhrer K, Scheuring S, Verhoef J, Fluit A, Heinz H-P, et al. The stability of grlA, grlB, gyrA, gyrB and norA mutations and MIC values of five fluoroquinolones in three different clonal populations of methicillin-resistant Staphylococcus aureus. Clin Microbiol Infect. 1999 May;5(5):287–90. pmid:11856269
  44. 44. Levy SB. Factors impacting on the problem of antibiotic resistance. J Antimicrob Chemother. 2002 Jan 1;49(1):25–30. pmid:11751763
  45. 45. Tantivitayakul P, Lapirattanakul J, Vichayanrat T, Muadchiengka T. Antibiotic Resistance Patterns and Related Mobile Genetic Elements of Pneumococci and β-Hemolytic Streptococci in Thai Healthy Children. Indian J Microbiol. 2016 Dec;56(4):417–25. pmid:27784937