Skip to main content
Erschienen in: Gut Pathogens 1/2019

Open Access 01.12.2019 | Short report

Hypervirulent clade 2, ribotype 019/sequence type 67 Clostridioides difficile strain from Japan

verfasst von: Ryoichi Saito, Yukino Usui, Alafate Ayibieke, Jun Nakajima, Isaac Prah, Kazunari Sonobe, Yoshibumi Aiso, Shiori Ito, Yasuhiro Itsui, Yoshiro Hadano, Yoko Nukui, Ryuji Koike, Shuji Tohda

Erschienen in: Gut Pathogens | Ausgabe 1/2019

Abstract

Background

Clostridioides difficile ribotype (RT) 019/sequence type (ST) 67 strains belong to a hypervirulent lineage closely related to RT027/ST1; however, limited data are available for hypervirulent clade 2 lineages in Japan. Herein, we report the draft genome of a C. difficile strain B18-123 belonging to clade 2, RT019/ST67 for the first time in Japan.

Results

The pathogenicity locus carried by B18-123 (19.6 kb) showed higher homology (97.29% nucleotide identity) with strain R20291 (RT027/ST1) than the reference strain 630 (RT012/ST54), and B18-123 harbored 8-nucleotide substitutions in tcdC. However, it did not contain an 18-base pair (bp) deletion or a single-bp deletion at position 117 in tcdC, which was identified in the previous strain R20291. A cytotoxicity assay revealed similar cytotoxicity levels between strains B18-123 and ATCC BAA-1870 (RT027/ST1). The B18-123 strain was found to be susceptible to metronidazole and vancomycin.

Conclusion

Our findings contribute to the further understanding of the characteristics of hypervirulent clade 2 including RT019/ST67 lineages.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
RT
ribotype
MLST
multilocus sequence typing
ST
sequence type
TcdA
toxin A
TcdB
toxin B
Cdt
binary toxin
PaLoc
pathogenicity locus
CdtLoc
Cdt locus
MIC
minimum inhibitory concentration

Introduction

Clostridioides difficile infection (CDI) is a well-known cause of healthcare-associated infections, ranging from mild diarrhea to severe pseudomembranous colitis and even death. It may be related to disruption of the indigenous intestinal microbiota by prolonged treatment with drugs such as antibiotics [1]. The incidence of CDI has increased over the past decade and clinical manifestations are typically triggered by toxin A (TcdA) and toxin B (TcdB) [2].
Epidemic hypervirulent strains with binary toxin (Cdt), North American pulsed-field type one (NAP1)/ribotype (RT) 027/sequence type (ST) 1 belonging to clade 2 and RT078/ST11 belonging to clade 5 have been identified as major lineages responsible for outbreaks in North America and Europe because of their high sporulation capacity and toxin production and resistance to fluoroquinolones [36]. However, these strains are rarely reported in Asia including in Japan, where other lineages such as RT014, RT017, and RT018 have been known to be predominant [7, 8]. RT019/ST67, which is closely related to NAP1/RT027/ST1 among clade 2 [9], produces high levels of toxins upon tcdC repression caused by an 18-base pair (bp) deletion and a single-bp deletion at position 117 in tcdC [10]. Although the previously reported RT019/ST67 strains [1012] have not shown any resistance to vancomycin and metronidazole, additional data are needed to understand the characteristics of the RT019/ST67 lineage to implement more effective treatments and methods of infection control.
In Japan, NAP1/RT027/ST1 strains are occasionally isolated from patients with fulminant colitis [13, 14]. However, limited data, including genome sequences, are available for hypervirulent clade 2 because of its extremely low prevalence among CDI incidences in Japan [15]. We reported the detection of a C. difficile strain B18-123 belonging to hypervirulent clade 2, RT019/ST67, in Japan isolated from a patient with multiple recurrent episodes of severe diarrhea.

Methods

Bacterial isolates and growth conditions

Clostridioides difficile strain B18-123 was obtained from a stool sample of a 81-year-old elderly male patient who underwent chemotherapy for hepatocellular carcinoma at Medical Hospital of Tokyo Medical and Dental University in 2018. Bacterial identification was performed using the C. DIFF QUIK CHEK COMPLETE test (Abbott Diagnostics Medical, Tokyo, Japan) and confirmed by 16S rRNA gene sequencing [16] for quality assurance of the following data. C. difficile strains American Type Culture Collection (ATCC) 700057 (RT038/ST48; TcdA, TcdB, Cdt) and ATCC BAA-1870 (NAP1/RT027/ST1; TcdA+, TcdB+, Cdt+) were also used in the study. The C. difficile strains were grown at 37 °C under anaerobic conditions using brain heart infusion (BHI; Oxoid, Basingstoke, UK) broth or agar plate supplemented with 5 g/L yeast extract and 0.1% (w/v) l-cysteine (BHIS).

Multilocus sequence typing, ribotyping, and toxin profiles

Multilocus sequence typing (MLST) and ribotyping were performed as previously described [16, 17]. tcdA, tcdB, cdtA, and cdtB were detected by multiplex PCR [18], followed by analysis of the length of tcdA as described [19]. A cytotoxicity assay was performed for B18-123 together with the ATCC BAA-1870 and ATCC 700057 strains as described previously [16].

Genome sequencing, de novo assembly, and annotation

Genomic DNA was extracted with a NucleoSpin Tissue kit (Takara Bio, Shiga, Japan) and subjected to quality control by agarose gel electrophoresis. The DNA obtained was quantified with a Synergy H1 (BioTek, Winooski, VT, USA) and QuantiFluor dsDNA System (Promega, Madison, WI, USA), and sequenced using a NextSeq platform (Illumina, Inc., San Diego, CA, USA) using a 2 × 151-bp paired-end approach. After quality control on raw reads was performed using Sickle (https://​github.​com/​najoshi/​sickle), the reads were assembled de novo using SPAdes version 3.10.1. Gene annotation was determined with Prokka version 1.11. The draft genome was subjected to in silico analyses, such as MLST using MLST version 2.0 [20] and detection of antibiotic resistance mechanisms using ResFinder version 3.1 [21], ARG-ANNOT [22], and CARD [23].

Comparative genomics

Phylogenetic analysis was performed with a pathogenicity locus (PaLoc) and tcdC sequences of 12 complete and draft genomes, including nine ST67 strains deposited in the GenBank, by the maximum likelihood method based on the Tamura-Nei model [24] using MEGA X software [25]. Sequence comparisons of PaLoc and Cdt locus (CdtLoc) were conducted using ClustalW (https://​clustalw.​ddbj.​nig.​ac.​jp/​) and Easyfig version 2.2.2 [26]. The presence of insertion sequences and tRNAs were identified on the PaLoc and its flanking region of B18-123 using ISfinder [27] and tRNAscan-SE [28], respectively. The presence of phage-related sequences was investigated in the whole genome of B18-123 using PHASTER [29].

Antimicrobial susceptibility testing

The minimum inhibitory concentrations (MICs) of six antibiotics (metronidazole, vancomycin, rifampin, erythromycin, clindamycin, and moxifloxacin) were determined using an Etest (bioMérieux, Marcy-l’Étoile, France) and the breakpoints were based on EUCAST epidemiological cut-off values (http://​www.​eucast.​org/​clinical_​breakpoints/​).

Statistical analysis

One-way analysis of variance was performed using GraphPad Prism version 7 software to determine the statistical significance of the differences for comparison. p < 0.05 was considered to indicate significance.

Results and discussion

Following draft genome sequencing of B18-123, a total of 4,284,537 reads were assembled into 98 contigs larger than 200 bp (largest scaffold = 526,102 bp; N50 = 277, 155 bp; L50 = 5; average GC = 28.39%) with an estimated genome size of 4,221,572 bp and average coverage of 50 ×. MLST analysis in silico along with conventional PCR assigned C. difficile strain B18-123 to ST67, which belongs to hypervirulent clade 2. B18-123 strain was classified into RT019 by ribotyping. Although in Japan, only two ST67 strains without ribotype data have been deposited in the GenBank in 2019 (last accessed on September 22, 2019), no report of CDI incidences with RT019/ST67 in Japan has been published so far in English literature. Therefore, our results indicate that a hypervirulent strain with RT019/ST67 responsible for severe diarrhea has been reported for the first time in Japan.
Strain B18-123 possesses tcdA and tcdB, and their presence was supported by in silico analysis. PaLoc carried by B18-123 was located between two conserved genes, cdu1 and cdd1 (Fig. 1a). BLAST results indicated that the PaLoc in B18-123 (19.6 kb) was homologous with the reference genome 630 (accession number AM180355; RT012/ST54; 99% query cover and 94.88% identity), R20291 (accession number FN545816; NAP1/RT027/ST1; 99% query cover and 97.29% identity), and M120 (accession number NC_017174; classified as clade 5, RT078/ST11; 99% query cover and 94.02% identity) strains. This indicates that the PaLoc carried by B18-123 is genetically closely related to that of R20291. These findings were supported by phylogenetic tree analysis of PaLoc carried by 630, R20291, M120, B18-123, and ST67 strains with complete PaLoc deposited in the GenBank (last accessed on September 22, 2019) (Fig. 1b).
In this study, no intact insertion sequences were found 10 kb upstream and downstream from PaLoc; however, truncated sequences (≤ 20 bp) of many insertion sequence families matched with our sequence. These results agree with a previous finding that C. difficile PaLoc does not fit the genetically accepted definition of pathogenicity islands containing direct repeats and insertion sequences [30]. However, the transfer of a chromosomal fragment containing the PaLoc proved to be caused by a chromosomally integrated mobile genetic element such as conjugative transposon [31, 32]. The detailed mechanism of this transfer has not yet been clarified.
Although no tRNA was identified on the scaffold containing PaLoc (location on scaffold of NODE013; 119,372 bp), 76 tRNAs representing all 20 amino acids were identified in the whole genome of B18-123. As the reference strain 630 and strain R20291 possess 87 tRNAs and 65 tRNAs, respectively, further investigations considering the entire cellular population of tRNAs may reveal the functional role of tRNAs against cellular physiological process, such as modulation of toxin production.
Additionally, although strain B18-123 harbored an 8-nucleotide substitution in tcdC, a negative regulator of tcdA and tcdB [33] compared to the reference genome 630 (Fig. 1c), only a single amino acid substitution of Asp7Glu (caused by a nucleotide T21G substitution), which was located outside of the three predicted domains [34], was found in 232 amino acids among both strains. Moreover, we did not detect an 18-bp deletion or a single-bp deletion at position 117, or a 39-bp deletion in tcdC as previously reported [3, 10, 11, 33, 35, 36]. On the other hand, we found that the tcdC sequences of ST67 strains (20100502, Lei034, F314, 5550, 51680, FD094, and FD109), except for strain LIBA5757 [10], were 100% identical to that of B18-123 (Fig. 1d). However, we did not find any information indicating that these strains were associated with severe CDI episodes. The amino acid sequence of B18-123 TcdC showed 95.83% identity with strain R20291 but showed very low identity with strain M120 because of the 39-bp deletion in tcdC. Collectively, our results suggest that TcdC carried by B18-123 causes different regulation of TcdA and TcdB production compared to previously identified hypervirulent lineages such as in the NAP1/RT027/ST1, RT019/ST67 (LIBA5757), and RT078/ST11 stains. These findings also indicate that it may be difficult to directly determine the TcdC characteristics of B18-123 by nucleic acid amplification tests available in clinical settings [37].
Although conflicting findings have been reported regarding the functional role of TcdC [3840], the regulation of TcdA and TcdB production in B18-123, which possesses an Asp7Glu substitution in TcdC, may differ from that in previously identified hypervirulent clade 2 strains. Therefore, we further investigated the relationship between tcdC mutation and toxin activity in a cytotoxicity assay [16]. Vero cell viability in B18-123 was similar to that in strain ATCC BAA-1870 (NAP1/RT027/ST1) with an 18-bp deletion and a single-bp deletion at position 117 in tcdC (P = 0.7859) (Fig. 2a). This result was related to the morphological changes shown by cell rounding of Vero cells observed under a light microscope (Fig. 2b). These findings suggest that TcdC carried by B18-123 allows the cell to increase toxin production, leading to high virulence. However, we could not directly quantify both TcdA and TcdB or clarify the mechanism of this protein in toxin regulation in B18-123 in this study.
As strain B18-123 also possesses cdtA and cdtB, CdtLoc (5.2 kb) carried by B18-123 was compared to those of the R20291 and M120 strains. The BLAST results indicated that CdtLoc in B18-123 showed homology with the R20291 (100% query cover and 99.90% identity) and M120 (100% query cover and 97.99% identity) strains. The binary toxin regulator gene cdtR (747 bp) in B18-123 was completely conserved with that in strain R20291 (100% query cover and 100% identity) and highly conserved with that in strain M120 (100% query cover and 97.46% identity), indicating that CdtR carried by B18-123 modulates binary toxin production similar to in NAP1/RT027/ST1 strains.
PHASTER identified three complete prophage sequences (scores > 90 and over; Table 1) and showed that strain B18-123 possessed the C. difficile bacteriophages phiMMP02, phiCDHM19 (belong to Myoviridae family), and phiCD111 (belong to Siphoviridae family). Although the B18-123 genome also contained one questionable and three incomplete prophage sequences, further studies are needed to understand the role of the phages in the evolution of RT019/ST67 lineages.
Table 1
Summary of phage-related regions identified by PHASTER in the B18-123 genome
Region
Length of prophage (kb)
Completeness
PHASTER score
No. of total proteins
Region position
Most common phage
GC content
(%)
Location on scaffolds
1
69.2 
Intact
150
93
294078–363300
PHAGE_Clostr_phiMMP02_NC_019421 (26)
28.18
NODE001
6
44.8 
Intact
140
66
1–44849
PHAGE_Clostr_phiCDHM19_NC_028996 (21)
28.25
NODE021
7
40.4 
Intact
125
48
48–40472
PHAGE_Clostr_phiCD111_NC_028908 (46)
30.88
NODE022
All contigs were merged into a single continuous sequence on PHASTER (http://​phaster.​ca/​). B18-123 genome also contained three incomplete and one questionable prophage sequences (scores < 90)
Strain B18-123 was susceptible to metronidazole (MIC = 0.19 mg/L), vancomycin (MIC = 0.75 mg/L), rifampin (MIC ≤ 0.002 mg/L), and moxifloxacin (MIC = 1.0 mg/L). Additionally, B18-123 showed low-level MICs to erythromycin (1.5 mg/L) and clindamycin (0.75 mg/L). We then performed in silico analysis to investigate the presence of antibiotic resistance genes using ResFinder version 3.1 [21], ARG-ANNOT [22], and CARD [23]. We detected only a C656T nucleotide substitution within the 23S rRNA copy on the genome with CARD. However, as a previous study suggested, the substitution has been not associated with conferring resistance to macrolides and lincosamides in C. difficile [41]. Our findings obtained from the resistome analysis seem to be sound and coincide with phenotypic results of antibiotic MICs. As evidenced by our MIC results with vancomycin on B18-123, treatment with the same was very effective for treating the CDI in this patient. Additionally, no amino acid substitutions in GyrA or GyrB were identified between B18-123 and fluoroquinolone-susceptible C. difficile strain 630 [42]. These results agree with those of a previous study of RT019/ST67 strain [10].

Conclusion

In conclusion, we reported for the first time that C. difficile strain B18-123 in Japan belongs to hypervirulent clade 2, RT019/ST67. Our findings enhance our understanding of the characteristics of hypervirulent clade 2 strains and further emphasize the need for continuous monitoring of C. difficile isolates to assess the spread of clade 2 strains in Japan.

Acknowledgements

The manuscript was edited by Editage, a language editing company.
As this study did not involve human participants or animals, informed consent and ethical approval were not required in this study.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat McDonald LC, Owings M, Jernigan DB. Clostridium difficile infection in patients discharged from US short-stay hospitals, 1996-2003. Emerg Infect Dis. 2006;12(3):409–15.PubMedPubMedCentralCrossRef McDonald LC, Owings M, Jernigan DB. Clostridium difficile infection in patients discharged from US short-stay hospitals, 1996-2003. Emerg Infect Dis. 2006;12(3):409–15.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Aktories K, Schwan C, Jank T. Clostridium difficile toxin biology. Annu Rev Microbiol. 2017;71:281–307.PubMedCrossRef Aktories K, Schwan C, Jank T. Clostridium difficile toxin biology. Annu Rev Microbiol. 2017;71:281–307.PubMedCrossRef
3.
Zurück zum Zitat Warny M, Pepin J, Fang A, Killgore G, Thompson A, Brazier J, et al. Toxin production by an emerging strain of Clostridium difficile associated with outbreaks of severe disease in North America and Europe. Lancet. 2005;366(9491):1079–84.PubMedCrossRef Warny M, Pepin J, Fang A, Killgore G, Thompson A, Brazier J, et al. Toxin production by an emerging strain of Clostridium difficile associated with outbreaks of severe disease in North America and Europe. Lancet. 2005;366(9491):1079–84.PubMedCrossRef
4.
Zurück zum Zitat Goorhuis A, Bakker D, Corver J, Debast SB, Harmanus C, Notermans DW, et al. Emergence of Clostridium difficile infection due to a new hypervirulent strain, polymerase chain reaction ribotype 078. Clin Infect Dis. 2008;47(9):1162–70.PubMedCrossRef Goorhuis A, Bakker D, Corver J, Debast SB, Harmanus C, Notermans DW, et al. Emergence of Clostridium difficile infection due to a new hypervirulent strain, polymerase chain reaction ribotype 078. Clin Infect Dis. 2008;47(9):1162–70.PubMedCrossRef
5.
Zurück zum Zitat Merrigan M, Venugopal A, Mallozzi M, Roxas B, Viswanathan VK, Johnson S, et al. Human hypervirulent Clostridium difficile strains exhibit increased sporulation as well as robust toxin production. J Bacteriol. 2010;192(19):4904–11.PubMedPubMedCentralCrossRef Merrigan M, Venugopal A, Mallozzi M, Roxas B, Viswanathan VK, Johnson S, et al. Human hypervirulent Clostridium difficile strains exhibit increased sporulation as well as robust toxin production. J Bacteriol. 2010;192(19):4904–11.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Drudy D, Quinn T, O’Mahony R, Kyne L, O’Gaora P, Fanning S. High-level resistance to moxifloxacin and gatifloxacin associated with a novel mutation in gyrB in toxin-A-negative, toxin-B-positive Clostridium difficile. J Antimicrob Chemother. 2006;58(6):1264–7.PubMedCrossRef Drudy D, Quinn T, O’Mahony R, Kyne L, O’Gaora P, Fanning S. High-level resistance to moxifloxacin and gatifloxacin associated with a novel mutation in gyrB in toxin-A-negative, toxin-B-positive Clostridium difficile. J Antimicrob Chemother. 2006;58(6):1264–7.PubMedCrossRef
7.
Zurück zum Zitat Jia HB, Du PC, Yang H, Zhang YY, Wang J, Zhang W, et al. Nosocomial transmission of Clostridium difficile ribotype 027 in a Chinese hospital, 2012–2014, traced by whole genome sequencing. BMC Genomics. 2016;17:405.PubMedPubMedCentralCrossRef Jia HB, Du PC, Yang H, Zhang YY, Wang J, Zhang W, et al. Nosocomial transmission of Clostridium difficile ribotype 027 in a Chinese hospital, 2012–2014, traced by whole genome sequencing. BMC Genomics. 2016;17:405.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Griffiths D, Fawley W, Kachrimanidou M, Bowden R, Crook DW, Fung R, et al. Multilocus sequence typing of Clostridium difficile. J Clin Microbiol. 2010;48(3):770–8.PubMedCrossRef Griffiths D, Fawley W, Kachrimanidou M, Bowden R, Crook DW, Fung R, et al. Multilocus sequence typing of Clostridium difficile. J Clin Microbiol. 2010;48(3):770–8.PubMedCrossRef
10.
Zurück zum Zitat Quesada-Gomez C, Lopez-Urena D, Chumbler N, Kroh HK, Castro-Pena C, Rodriguez C, et al. Analysis of TcdB proteins within the hypervirulent clade 2 reveals an impact of RhoA glucosylation on Clostridium difficile proinflammatory activities. Infect Immun. 2016;84(3):856–65.PubMedPubMedCentralCrossRef Quesada-Gomez C, Lopez-Urena D, Chumbler N, Kroh HK, Castro-Pena C, Rodriguez C, et al. Analysis of TcdB proteins within the hypervirulent clade 2 reveals an impact of RhoA glucosylation on Clostridium difficile proinflammatory activities. Infect Immun. 2016;84(3):856–65.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Pires RN, Monteiro AA, Saldanha GZ, Falci DR, Caurio CFB, Sukiennik TCT, et al. Hypervirulent Clostridium difficile strain has arrived in Brazil. Infect Control Hosp Epidemiol. 2018;39(3):371–3.PubMedCrossRef Pires RN, Monteiro AA, Saldanha GZ, Falci DR, Caurio CFB, Sukiennik TCT, et al. Hypervirulent Clostridium difficile strain has arrived in Brazil. Infect Control Hosp Epidemiol. 2018;39(3):371–3.PubMedCrossRef
12.
Zurück zum Zitat Cao H, Wong SC, Yam WC, Liu MC, Chow KH, Wu AK, et al. Genomic investigation of a sequence type 67 Clostridium difficile causing community-acquired fulminant colitis in Hong Kong. Int J Med Microbiol. 2019;309(5):270–3.PubMedCrossRef Cao H, Wong SC, Yam WC, Liu MC, Chow KH, Wu AK, et al. Genomic investigation of a sequence type 67 Clostridium difficile causing community-acquired fulminant colitis in Hong Kong. Int J Med Microbiol. 2019;309(5):270–3.PubMedCrossRef
13.
Zurück zum Zitat Nakamura I, Yamaguchi T, Tsukimori A, Sato A, Fukushima S, Mizuno Y, et al. Fulminant colitis from Clostridium difficile infection, the epidemic strain ribotype 027, in Japan. J Infect Chemother. 2014;20(5–6):380–3.PubMedCrossRef Nakamura I, Yamaguchi T, Tsukimori A, Sato A, Fukushima S, Mizuno Y, et al. Fulminant colitis from Clostridium difficile infection, the epidemic strain ribotype 027, in Japan. J Infect Chemother. 2014;20(5–6):380–3.PubMedCrossRef
14.
Zurück zum Zitat Nishimura S, Kou T, Kato H, Watanabe M, Uno S, Senoh M, et al. Fulminant pseudomembranous colitis caused by Clostridium difficile PCR ribotype 027 in a healthy young woman in Japan. J Infect Chemother. 2014;20(11):729–31.PubMedCrossRef Nishimura S, Kou T, Kato H, Watanabe M, Uno S, Senoh M, et al. Fulminant pseudomembranous colitis caused by Clostridium difficile PCR ribotype 027 in a healthy young woman in Japan. J Infect Chemother. 2014;20(11):729–31.PubMedCrossRef
15.
Zurück zum Zitat Kato H, Senoh M, Honda H, Fukuda T, Tagashira Y, Horiuchi H, et al. Clostridioides (Clostridium) difficile infection burden in Japan: a multicenter prospective study. Anaerobe. 2019;12(19):30046. Kato H, Senoh M, Honda H, Fukuda T, Tagashira Y, Horiuchi H, et al. Clostridioides (Clostridium) difficile infection burden in Japan: a multicenter prospective study. Anaerobe. 2019;12(19):30046.
16.
Zurück zum Zitat Kuwata Y, Tanimoto S, Sawabe E, Shima M, Takahashi Y, Ushizawa H, et al. Molecular epidemiology and antimicrobial susceptibility of Clostridium difficile isolated from a University teaching hospital in Japan. Eur J Clin Microbiol Infect Dis. 2015;34(4):763–72.PubMedCrossRef Kuwata Y, Tanimoto S, Sawabe E, Shima M, Takahashi Y, Ushizawa H, et al. Molecular epidemiology and antimicrobial susceptibility of Clostridium difficile isolated from a University teaching hospital in Japan. Eur J Clin Microbiol Infect Dis. 2015;34(4):763–72.PubMedCrossRef
17.
Zurück zum Zitat Kato H, Ito Y, Akahane T, Izumida S, Yokoyama T, Kaji C, et al. Typing of Clostridium difficile isolates endemic in Japan by sequencing of slpA and its application to direct typing. J Med Microbiol. 2010;59(5):556–62.PubMedCrossRef Kato H, Ito Y, Akahane T, Izumida S, Yokoyama T, Kaji C, et al. Typing of Clostridium difficile isolates endemic in Japan by sequencing of slpA and its application to direct typing. J Med Microbiol. 2010;59(5):556–62.PubMedCrossRef
18.
Zurück zum Zitat Persson S, Torpdahl M, Olsen KEP. New multiplex PCR method for the detection of Clostridium difficile toxin A (tcdA) and toxin B (tcdB) and the binary toxin (cdtA/cdtB) genes applied to a Danish strain collection. Clin Microbiol Infect. 2008;14(11):1057–64.PubMedCrossRef Persson S, Torpdahl M, Olsen KEP. New multiplex PCR method for the detection of Clostridium difficile toxin A (tcdA) and toxin B (tcdB) and the binary toxin (cdtA/cdtB) genes applied to a Danish strain collection. Clin Microbiol Infect. 2008;14(11):1057–64.PubMedCrossRef
19.
Zurück zum Zitat Kato H, Kato N, Watanabe K, Iwai N, Nakamura H, Yamamoto T, et al. Identification of toxin A-negative, toxin B-positive Clostridium difficile by PCR. J Clin Microbiol. 1998;36(8):2178–82.PubMedPubMedCentral Kato H, Kato N, Watanabe K, Iwai N, Nakamura H, Yamamoto T, et al. Identification of toxin A-negative, toxin B-positive Clostridium difficile by PCR. J Clin Microbiol. 1998;36(8):2178–82.PubMedPubMedCentral
20.
Zurück zum Zitat Larsen MV, Cosentino S, Rasmussen S, Friis C, Hasman H, Marvig RL, et al. Multilocus sequence typing of total-genome-sequenced bacteria. J Clin Microbiol. 2012;50(4):1355–61.PubMedPubMedCentralCrossRef Larsen MV, Cosentino S, Rasmussen S, Friis C, Hasman H, Marvig RL, et al. Multilocus sequence typing of total-genome-sequenced bacteria. J Clin Microbiol. 2012;50(4):1355–61.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Zankari E, Hasman H, Cosentino S, Vestergaard M, Rasmussen S, Lund O, et al. Identification of acquired antimicrobial resistance genes. J Antimicrob Chemother. 2012;67(11):2640–4.PubMedPubMedCentralCrossRef Zankari E, Hasman H, Cosentino S, Vestergaard M, Rasmussen S, Lund O, et al. Identification of acquired antimicrobial resistance genes. J Antimicrob Chemother. 2012;67(11):2640–4.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat 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;58(1):212–20.PubMedPubMedCentralCrossRef 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;58(1):212–20.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Jia BF, Raphenya AR, Alcock B, Waglechner N, Guo PY, Tsang KK, et al. CARD 2017: expansion and model-centric curation of the comprehensive antibiotic resistance database. Nucleic Acids Res. 2017;45(D1):D566–73.PubMedCrossRef Jia BF, Raphenya AR, Alcock B, Waglechner N, Guo PY, Tsang KK, et al. CARD 2017: expansion and model-centric curation of the comprehensive antibiotic resistance database. Nucleic Acids Res. 2017;45(D1):D566–73.PubMedCrossRef
24.
Zurück zum Zitat Tamura K, Nei M. Estimation of the number of nucleotide substitutions in the control region of mitochondrial-DNA in humans and chimpanzees. Mol Biol Evol. 1993;10(3):512–26.PubMed Tamura K, Nei M. Estimation of the number of nucleotide substitutions in the control region of mitochondrial-DNA in humans and chimpanzees. Mol Biol Evol. 1993;10(3):512–26.PubMed
25.
Zurück zum Zitat Kumar S, Stecher G, Li M, Knyaz C, Tamura K. MEGA X: molecular evolutionary genetics analysis across computing platforms. Mol Biol Evol. 2018;35(6):1547–9.PubMedPubMedCentralCrossRef Kumar S, Stecher G, Li M, Knyaz C, Tamura K. MEGA X: molecular evolutionary genetics analysis across computing platforms. Mol Biol Evol. 2018;35(6):1547–9.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Siguier P, Perochon J, Lestrade L, Mahillon J, Chandler M. ISfinder: the reference centre for bacterial insertion sequences. Nucleic Acids Res. 2006;34:D32–6.PubMedCrossRef Siguier P, Perochon J, Lestrade L, Mahillon J, Chandler M. ISfinder: the reference centre for bacterial insertion sequences. Nucleic Acids Res. 2006;34:D32–6.PubMedCrossRef
28.
Zurück zum Zitat Lowe TM, Chan PP. tRNAscan-SE on-line: integrating search and context for analysis of transfer RNA genes. Nucleic Acids Res. 2016;44(W1):W54–7.PubMedPubMedCentralCrossRef Lowe TM, Chan PP. tRNAscan-SE on-line: integrating search and context for analysis of transfer RNA genes. Nucleic Acids Res. 2016;44(W1):W54–7.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Arndt D, Grant JR, Marcu A, Sajed T, Pon A, Liang YJ, et al. PHASTER: a better, faster version of the PHAST phage search tool. Nucleic Acids Res. 2016;44(W1):W16–21.PubMedPubMedCentralCrossRef Arndt D, Grant JR, Marcu A, Sajed T, Pon A, Liang YJ, et al. PHASTER: a better, faster version of the PHAST phage search tool. Nucleic Acids Res. 2016;44(W1):W16–21.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Rupnik M, Dupuy B, Fairweather NF, Gerding DN, Johnson S, Just I, et al. Revised nomenclature of Clostridium difficile toxins and associated genes. J Med Microbiol. 2005;54(2):113–7.PubMedCrossRef Rupnik M, Dupuy B, Fairweather NF, Gerding DN, Johnson S, Just I, et al. Revised nomenclature of Clostridium difficile toxins and associated genes. J Med Microbiol. 2005;54(2):113–7.PubMedCrossRef
31.
Zurück zum Zitat Mullany P, Allan E, Roberts AP. Mobile genetic elements in Clostridium difficile and their role in genome function. Res Microbiol. 2015;166(4):361–7.PubMedPubMedCentralCrossRef Mullany P, Allan E, Roberts AP. Mobile genetic elements in Clostridium difficile and their role in genome function. Res Microbiol. 2015;166(4):361–7.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Brouwer MSM, Roberts AP, Hussain H, Williams RJ, Allan E, Mullany P. Horizontal gene transfer converts non-toxigenic Clostridium difficile strains into toxin producers. Nature Commun. 2013;4:2601.CrossRef Brouwer MSM, Roberts AP, Hussain H, Williams RJ, Allan E, Mullany P. Horizontal gene transfer converts non-toxigenic Clostridium difficile strains into toxin producers. Nature Commun. 2013;4:2601.CrossRef
33.
Zurück zum Zitat Matamouros S, England P, Dupuy B. Clostridium difficile toxin expression is inhibited by the novel regulator TcdC. Mol Microbiol. 2007;64(5):1274–88.PubMedCrossRef Matamouros S, England P, Dupuy B. Clostridium difficile toxin expression is inhibited by the novel regulator TcdC. Mol Microbiol. 2007;64(5):1274–88.PubMedCrossRef
34.
Zurück zum Zitat van Leeuwen HC, Bakker D, Steindel P, Kuijper EJ, Corver J. Clostridium difficile TcdC protein binds four-stranded G-quadruplex structures. Nucleic Acids Res. 2013;41(4):2382–93.PubMedPubMedCentralCrossRef van Leeuwen HC, Bakker D, Steindel P, Kuijper EJ, Corver J. Clostridium difficile TcdC protein binds four-stranded G-quadruplex structures. Nucleic Acids Res. 2013;41(4):2382–93.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Dupuy B, Govind R, Antunes A, Matamouros S. Clostridium difficile toxin synthesis is negatively regulated by TcdC. J Med Microbiol. 2008;57(6):685–9.PubMedCrossRef Dupuy B, Govind R, Antunes A, Matamouros S. Clostridium difficile toxin synthesis is negatively regulated by TcdC. J Med Microbiol. 2008;57(6):685–9.PubMedCrossRef
36.
Zurück zum Zitat He M, Sebaihia M, Lawley TD, Stabler RA, Dawson LF, Martin MJ, et al. Evolutionary dynamics of Clostridium difficile over short and long time scales. Proc Natl Acad Sci USA. 2010;107(16):7527–32.PubMedCrossRefPubMedCentral He M, Sebaihia M, Lawley TD, Stabler RA, Dawson LF, Martin MJ, et al. Evolutionary dynamics of Clostridium difficile over short and long time scales. Proc Natl Acad Sci USA. 2010;107(16):7527–32.PubMedCrossRefPubMedCentral
37.
Zurück zum Zitat Crobach MJT, Planche T, Eckert C, Barbut F, Terveer EM, Dekkers OM, et al. European society of clinical microbiology and infectious diseases: update of the diagnostic guidance document for Clostridium difficile infection. Clin Microbiol Infect. 2016;22:S63–81.PubMedCrossRef Crobach MJT, Planche T, Eckert C, Barbut F, Terveer EM, Dekkers OM, et al. European society of clinical microbiology and infectious diseases: update of the diagnostic guidance document for Clostridium difficile infection. Clin Microbiol Infect. 2016;22:S63–81.PubMedCrossRef
38.
Zurück zum Zitat Cartman ST, Kelly ML, Heeg D, Heap JT, Minton NP. Precise manipulation of the Clostridium difficile chromosome reveals a lack of association between the tcdC genotype and toxin production. Appl Environ Microbiol. 2012;78(13):4683–90.PubMedPubMedCentralCrossRef Cartman ST, Kelly ML, Heeg D, Heap JT, Minton NP. Precise manipulation of the Clostridium difficile chromosome reveals a lack of association between the tcdC genotype and toxin production. Appl Environ Microbiol. 2012;78(13):4683–90.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Carter GP, Douce GR, Govind R, Howarth PM, Mackin KE, Spencer J, et al. The anti-sigma factor TcdC modulates hypervirulence in an epidemic BI/NAP1/027 clinical isolate of Clostridium difficile. Plos Pathogens. 2011;7(10):e1002317.PubMedPubMedCentralCrossRef Carter GP, Douce GR, Govind R, Howarth PM, Mackin KE, Spencer J, et al. The anti-sigma factor TcdC modulates hypervirulence in an epidemic BI/NAP1/027 clinical isolate of Clostridium difficile. Plos Pathogens. 2011;7(10):e1002317.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Murray R, Boyd D, Levett PN, Mulvey MR, Alfa MJ. Truncation in the tcdC region of the Clostridium difficile pathLoc of clinical isolates does not predict increased biological activity of toxin B or toxin A. BMC Infect Dis. 2009;9:103.PubMedPubMedCentralCrossRef Murray R, Boyd D, Levett PN, Mulvey MR, Alfa MJ. Truncation in the tcdC region of the Clostridium difficile pathLoc of clinical isolates does not predict increased biological activity of toxin B or toxin A. BMC Infect Dis. 2009;9:103.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Wang BY, Lv Z, Zhang PP, Su JR. Molecular epidemiology and antimicrobial susceptibility of human Clostridium difficile isolates from a single institution in Northern. Medicine. 2018;97(25):e11219.PubMedPubMedCentralCrossRef Wang BY, Lv Z, Zhang PP, Su JR. Molecular epidemiology and antimicrobial susceptibility of human Clostridium difficile isolates from a single institution in Northern. Medicine. 2018;97(25):e11219.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Wasels F, Kuehne SA, Cartman ST, Spigaglia P, Barbanti F, Minton NP, et al. Fluoroquinolone resistance does not impose a cost on the fitness of Clostridium difficile in vitro. Antimicrob Agents Chemother. 2015;59(3):1794–6.PubMedPubMedCentralCrossRef Wasels F, Kuehne SA, Cartman ST, Spigaglia P, Barbanti F, Minton NP, et al. Fluoroquinolone resistance does not impose a cost on the fitness of Clostridium difficile in vitro. Antimicrob Agents Chemother. 2015;59(3):1794–6.PubMedPubMedCentralCrossRef
Metadaten
Titel
Hypervirulent clade 2, ribotype 019/sequence type 67 Clostridioides difficile strain from Japan
verfasst von
Ryoichi Saito
Yukino Usui
Alafate Ayibieke
Jun Nakajima
Isaac Prah
Kazunari Sonobe
Yoshibumi Aiso
Shiori Ito
Yasuhiro Itsui
Yoshiro Hadano
Yoko Nukui
Ryuji Koike
Shuji Tohda
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Gut Pathogens / Ausgabe 1/2019
Elektronische ISSN: 1757-4749
DOI
https://doi.org/10.1186/s13099-019-0336-3

Weitere Artikel der Ausgabe 1/2019

Gut Pathogens 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.