ksgA mutations confer resistance to kasugamycin in Neisseria gonorrhoeae

https://doi.org/10.1016/j.ijantimicag.2008.08.030Get rights and content

Abstract

The aminoglycoside antibiotic kasugamycin (KSG) inhibits translation initiation and thus the growth of many bacteria. In this study, we tested the susceptibilities to KSG of 22 low-passage clinical isolates and 2 laboratory strains of Neisseria gonorrhoeae. Although the range of KSG minimum inhibitory concentrations (MICs) was narrow (seven-fold), clinical isolates and laboratory strains fell into three distinct classes of KSG sensitivity, susceptible, somewhat sensitive and resistant, with MICs of 30, 60–100 and 200 μg/mL, respectively. Two genes have previously been shown to be involved in bacterial KSG resistance: rpsI, which encodes the 30S ribosomal subunit S9 protein; and ksgA, which encodes a predicted dimethyltransferase. Although sequencing of rpsI and ksgA from clinical isolates revealed polymorphisms, none correlated with the MICs of KSG. Ten spontaneous KSG-resistant (KSGR) mutants were isolated from laboratory strain FA1090 at a frequency of <4.4 × 10−6 resistant colony-forming units (CFU)/total CFU. All isolated KSGR variants had mutations in ksgA, whilst no mutations were observed in rpsI. ksgA mutations conferring KSG resistance included four point mutations, two in-frame and one out-of-frame deletions, one in-frame duplication and two frame-shift insertions. These data show a narrow range of susceptibilities for the clinical isolates and laboratory strains examined; moreover, the differences in MICs do not correlate with nucleotide polymorphisms in rpsI or ksgA. Additionally, spontaneous KSGR mutants arise by a variety of ksgA mutations.

Introduction

The aminoglycoside antibiotic kasugamycin (KSG), first isolated from Streptomyces kasugaensis, has been used to prevent rice blast disease caused by the fungus Pyricularia oryzae[1], [2], [3]. KSG inhibits the growth of a wide variety of microorganisms, with reported low toxicity against plants, humans and other animals [2], [4], [5]. However, as an aminoglycoside, some degree of nephrotoxicity and ototoxicity is expected. Several Gram-negative bacteria, including Pseudomonas spp. and Escherichia coli strains, as well as the Gram-positive Bacillus spp. are sensitive to KSG [6]. Although clinical use of KSG has been explored as treatment for Pseudomonas aeruginosa infection in the bladder [4], it is currently only used agriculturally. KSG inhibits translation initiation by blocking transfer RNA (tRNA) binding to the 30S ribosomal subunit; it can be bacteriostatic or bactericidal depending on the concentration used [7]. Recent structural analysis of KSG translation inhibition has provided a detailed mechanism for KSG binding and inhibition [8], [9]. KSG is thought to mimic messenger RNA (mRNA) codon nucleotides and to occupy the peptidyl (P) and exit (E) sites of the ribosome, causing distortion of the mRNA–tRNA codon–anticodon interaction and blocking translation initiation in susceptible organisms [8], [9].

Several KSG resistance mutations have been identified and characterised in E. coli[10], [11], [12], [13] and Bacillus subtilis[14]. The E. coli gene product of ksgA, an adenosine dimethyltransferase KsgA, is responsible for methylation of 16S ribosomal RNA (rRNA) adenosines at positions 1518 and 1519 [15], [16], [17]. Interestingly, this rRNA modification by KsgA appears to be conserved in all species of bacteria, archaea and eukarya studied to date [18], [19]. The exact biological function of this rRNA modification is unknown, and in many bacteria loss of KsgA-dependent methylation is not lethal [20], [21], [22]. Mutations that disrupt KsgA-dependent rRNA methylation are the most common mechanism of KSG resistance in bacteria [11]. Other mutations known to confer KSG resistance include mutation of the target nucleotides A1518 and A1519 in the 16S rRNA [12] and amino acid substitutions in the 30S protein subunit S9, the gene product of rpsI[10]. These mutations may stabilise the mRNA–tRNA interaction perturbed by KSG leading to KSG resistance. Interestingly, rpsI mutations can result in both resistance to and dependence on KSG [10], [23]. For example, one KSG-resistant (KSGR) rpsI mutant, E. coli strain MV101, required KSG to depress the rate of protein synthesis, otherwise the enhanced ribosomal activity caused by the rpsI mutation was lethal [10]. Both 16S rRNA and rpsI mutations occur less frequently than mutations in ksgA[11], [12].

Neisseria gonorrhoeae is an obligate human pathogen and is the only causative agent of the sexually transmitted infection gonorrhoea. In the USA, gonorrhoea is the second most frequently reported communicable disease, with 339 593 reported cases in 2005 and as many as 700 000 total cases yearly [24]. Many infected individuals may be asymptomatic [25]; however, serious symptomatic infections can occur both in men and women. Notably, N. gonorrhoeae can cause pelvic inflammatory disease (PID) in women [26], epididymitis in men and arthritis both in men and women [27]. Additionally, the pharynx, rectum and conjunctiva are sites often infected by N. gonorrhoeae, thus antibiotics must be effective at controlling the bacteria at multiple mucosal sites. Although antibiotic treatment has historically been effective in control of the disease, the increased prevalence of antibiotic-resistant N. gonorrhoeae has severely limited available treatment options [28]. Accordingly, the US Centers for Disease Control and Prevention (CDC) has classified N. gonorrhoeae as a ‘superbug’, and only cephalosporin drugs are recommended for treatment of gonorrhoea in the USA [29]. With ever-increasing resistance to antibiotics and with disease incidence on the rise in many countries, novel therapies are needed to control the spread of disease. Here we examined the susceptibilities of 22 clinical isolates and 2 laboratory strains of N. gonorrhoeae to KSG and isolated 10 spontaneous KSGR mutants.

Section snippets

Bacterial strains and growth conditions

KSG susceptibility was examined for a previously assembled panel of characterised, low-passage clinical isolates of N. gonorrhoeae from three US laboratories (Table 1) [30]. These included seven isolates from PID patients collected by the CDC, seven isolates from disseminated gonococcal infections (DGIs) described by O’Brien et al. [27], four endometrial isolates provided by the laboratory of Peter A. Rice and four local infection (i.e. urethritis or cervicitis) isolates from the Bell Flower

Neisseria gonorrhoeae strains vary in susceptibility to the antibiotic KSG

Since KSG is active against various Gram-negative bacteria [6], [7] and has never been examined in N. gonorrhoeae, we determined the MICs of KSG for 2 laboratory strains and 22 low-passage clinical isolates of N. gonorrhoeae (Table 1). These clinical isolates are from diverse disease courses including PID, DGI, endometrial involvement and local infection (urethritis or cervicitis) (Table 1). The MIC for the laboratory strain FA1090 (RM11.2) was 100 μg/mL, whilst the laboratory strain MS11 was

Discussion

Here we investigated the activity of the aminoglycoside antibiotic KSG against numerous clinical isolates and laboratory strains of N. gonorrhoeae. Although a previous report showed that pathogenic E. coli, P. aeruginosa, Klebsiella pneumoniae and Serratia spp. can have KSG resistance at levels between 100 and 400 μg/mL [6], most clinical isolates of N. gonorrhoeae investigated in this study were sensitive to lower levels (30–100 μg/mL) of KSG. Although there are no established MIC

Acknowledgments

The authors thank Allen Helm and Alison K. Criss for critical reading and editing of the manuscript.

Funding: This work was supported by National Institutes of Health (NIH) grants R01 AI055977, R01 AI044239 and R37 AI033493 from the US National Institutes of Health to HSS.

Competing interests: None declared.

Ethical approval: Not required.

References (48)

  • H. Umezawa et al.

    Kasugamycin, a new antibiotic

    Antimicrob Agents Chemother

    (1965)
  • T. Takeuchi et al.

    Pharmacology of kasugamycin and the effect on Pseudomonas infection

    J Antibiot (Tokyo)

    (1965)
  • T. Tamamura et al.

    Comparative studies on in vitro activities of kasugamycin and clinically-used aminoglycoside antibiotics

    Jpn J Antibiot

    (1999)
  • A.A. Levitan

    In vitro antibacterial activity of kasugamycin

    Appl Microbiol

    (1967)
  • F. Schluenzen et al.

    The antibiotic kasugamycin mimics mRNA nucleotides to destabilize tRNA binding and inhibit canonical translation initiation

    Nat Struct Mol Biol

    (2006)
  • B.S. Schuwirth et al.

    Structural analysis of kasugamycin inhibition of translation

    Nat Struct Mol Biol

    (2006)
  • E.R. Dabbs

    Escherichia coli kasugamycin dependence arising from mutation at the rpsI locus

    J Bacteriol

    (1983)
  • P.F. Sparling et al.

    Two genetic loci for resistance to kasugamycin in Escherichia coli

    J Bacteriol

    (1973)
  • R.A. Zimmermann et al.

    Alteration of ribosomal protein S4 by mutation linked to kasugamycin-resistance in Escherichia coli

    Proc Natl Acad Sci U S A

    (1973)
  • A. Tominaga et al.

    Kasugamycin-resistant mutants of Bacillus subtilis

    J Bacteriol

    (1978)
  • O.S. Andresson et al.

    Some properties of the ribosomal RNA methyltransferase encoded by ksgA and the polarity of ksgA transcription

    Mol Gen Genet

    (1980)
  • K. Inoue et al.

    Dissection of 16S rRNA methyltransferase (KsgA) function in Escherichia coli

    J Bacteriol

    (2007)
  • C.P. Van Buul et al.

    Kasugamycin resistant mutants of Bacillus stearothermophilus lacking the enzyme for the methylation of two adjacent adenosines in 16S ribosomal RNA

    Mol Gen Genet

    (1983)
  • K. Igarashi et al.

    Relationship between methylation of adenine near the 3′ end of 16-S ribosomal RNA and the activity of 30-S ribosomal subunits

    Eur J Biochem

    (1981)
  • Cited by (19)

    • Bacterial protein synthesis

      2023, Molecular Medical Microbiology, Third Edition
    • Gene interaction network to unravel the role of gut bacterial species in cardiovascular diseases: E. coli O157:H7 host-bacterial interaction study

      2021, Computers in Biology and Medicine
      Citation Excerpt :

      It is also reported that the ksgA gene has a novel activity of DNA glycosylase which prevent mutations in E. coli [28]. The mutations in the ksgA gene are responsible for the Kasugamycin resistance in Neisseria gonorrhoeae and they have a role in cell-envelop fitness in Salmonella enteritidis [29]. The gene rpsT codes for 30S ribosomal subunit protein S20, a key component of the 30S subunit of ribosome was involved in translation initiation and association of 30S and 50S subunits.

    • Targeted mutagenesis of Mycoplasma gallisepticum using its endogenous CRISPR/Cas system

      2020, Veterinary Microbiology
      Citation Excerpt :

      The ksgA gene encodes a ribosomal RNA adenine dimethylase family protein that methylates the 16S rRNA. Loss of KsgA methyltransferase activity modifies the ribosomal binding site for the aminoglycoside antimicrobial kasugamycin, resulting in resistance to it (Duffin and Seifert, 2009). The orthologues of the ksgA gene can be found in some mycoplasma genomes, but the role of the product of these orthologues in resistance to kasugamycin has not been confirmed experimentally (Li et al., 2017).

    • Dimethyl adenosine transferase (KsgA) contributes to cell-envelope fitness in Salmonella Enteritidis

      2018, Microbiological Research
      Citation Excerpt :

      The two adenosine targets and the DMAT activity of KsgA are universally conserved in bacteria (Harris et al., 2003; O’Farrell et al., 2006, 2008; Inoue et al., 2007; Binet and Maurelli, 2009). Lack of DMAT activity due to KsgA-deficiency invariably results in resistance to the aminoglycoside antibiotic kasugamycin in Gram-positive and Gram-negative bacteria (Helser et al., 1972; Inoue et al., 2007; Duffin and Seifert, 2009; Ochi et al., 2009; O’Farrell and Rife, 2012; Chiok et al., 2013). Thus, resistance to kasugamycin is considered as a hallmark of KsgA-deficiency.

    • Characterization of 16S rRNA Processing with Pre-30S Subunit Assembly Intermediates from E. coli

      2018, Journal of Molecular Biology
      Citation Excerpt :

      Crystal structures of the 70S ribosome and the individual subunits have shed light on many important aspects of the mechanism of translation, yet they provide limited information on the mechanism of ribosome assembly [4–6]. Understanding the process of ribosomal subunit biogenesis is highly significant, as these events have been linked to changes in virulence and drug resistance in various pathogens [7–10]. In addition, defects in ribosome assembly in humans can cause predispositions to some cancers and various disease states known as ribosomopathies (i.e., Diamond–Blackfan anemia) [11].

    View all citing articles on Scopus
    View full text