Antimicrobial resistance patterns and genetic elements associated with the antibiotic resistance of Helicobacter pylori strains from Shanghai

Yixin Liu, Su Wang, Feng Yang, Wenjing Chi, Li Ding, Tao Liu, Feng Zhu, Danian Ji, Jun Zhou, Yi Fang, Jinghao Zhang, Ping Xiang, Yanmei Zhang, Hu Zhao, Yixin Liu, Su Wang, Feng Yang, Wenjing Chi, Li Ding, Tao Liu, Feng Zhu, Danian Ji, Jun Zhou, Yi Fang, Jinghao Zhang, Ping Xiang, Yanmei Zhang, Hu Zhao

Abstract

Background: Shanghai, in east China, has one of the world's highest burdens of Helicobacter pylori infection. While multidrug regimens can effectively eradicate H. pylori, the increasing prevalence of antibiotic resistance (AR) in H. pylori has been recognized by the WHO as 'high priority' for urgent need of new therapies. Moreover, the genetic characteristics of H. pylori AR in Shanghai is under-reported. The purpose of this study was to determine the resistance prevalence, re-substantiate resistance-conferring mutations, and investigate novel genetic elements associated with H. pylori AR.

Results: We performed whole genome sequencing and antimicrobial susceptibility testing of 112 H. pylori strains isolated from gastric biopsy specimens from Shanghai patients with different gastric diseases. No strains were resistant to amoxicillin. Levofloxacin, metronidazole and clarithromycin resistance was observed in 39 (34.8%), 73 (65.2%) and 18 (16.1%) strains, respectively. There was no association between gastroscopy diagnosis and resistance phenotypes. We reported the presence or absence of several subsystem protein coding genes including hopE, hofF, spaB, cagY and pflA, and a combination of CRISPRs, which were potentially correlated with resistance phenotypes. The H. pylori strains were also annotated for 80 genome-wide AR genes (ARGs). A genome-wide ARG analysis was performed for the three antibiotics by correlating the phenotypes with the genetic variants, which identified the well-known intrinsic mutations conferring resistance to levofloxacin (N87T/I and/or D91G/Y mutations in gyrA), metronidazole (I38V mutation in fdxB), and clarithromycin (A2143G and/or A2142G mutations in 23S rRNA), and added 174 novel variations, including 23 non-synonymous SNPs and 48 frameshift Indels that were significantly enriched in either the antibiotic-resistant or antibiotic-susceptible bacterial populations. The variant-level linkage disequilibrium analysis highlighted variations in a protease Lon with strong co-occurring correlation with a series of resistance-associated variants.

Conclusion: Our study revealed multidrug antibiotic resistance in H. pylori strains from Shanghai, which was characterized by high metronidazole and moderate levofloxacin resistance, and identified specific genomic characteristics in relation to H. pylori AR. Continued surveillance of H. pylori AR in Shanghai is warranted in order to establish appropriate eradication treatment regimens for this population.

Keywords: Antibiotic resistance; CRISPRs; Genetic characteristics; Helicobacter pylori; Outer membrane protein; Variation; Virulence.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Genome-wide gene family analysis and functional classification of unique genes of antibiotic resistant categories. Venn diagrams showing the number of common and unique genes in MTZ- (A), CLA- (B) and LEV- (C) resistant (yellow) and susceptible (green) categories. The number in the parentheses indicates the total number of genes identified in each category. Pie diagrams revealing the status of unique genes of MTZ-R (D), CLA-R (E) and LEV-R (F) categories in NCBI-NR, SwissProt and COG databases
Fig. 2
Fig. 2
Analysis of OMP family genes in resistant and susceptible categories. Violin plots showing the distribution of the OMP genes in resistant and susceptible categories. The average presence levels of the total 62 OMP genes (A), the major OMP family genes along with two subfamilies genes (B), Hof family genes (C), Hom family genes (D), iron-regulated OMPs genes (F) and efflux pump OMP genes (F) in resistant and susceptible categories were compared, irrespective of the number of alleles of the genes. Solid lines indicate median levels and dotted lines indicate quartiles levels. G Proportion of strains with each OMP family gene in resistant and susceptible categories. Only genes with greater than 90% coverage were labelled as present. The number of strains containing the gene were listed on the right. AG *P < 0.05, ***P < 0.001. Where there were no statistical analysis results labeled, there were no significant difference
Fig. 3
Fig. 3
Analysis of virulence-associated genes in resistant and susceptible categories. Violin plots showing the distribution of the virulence-associated genes in resistant and susceptible categories. Comparisons of the average presence levels of the total virulence-associated genes (A), cagPAI genes (B) and flagellar genes (C) between resistant and susceptible categories were conducted, irrespective of the number of alleles of the genes. Solid lines indicate median levels and dotted lines indicate quartiles levels. Where there were no statistical analysis results labeled, there were no significant difference. D Two virulence-associated genes with significant different frequencies in MTZ-R and MTZ-S categories. *P < 0.05
Fig. 4
Fig. 4
Analysis of efflux pump genes in resistant and susceptible categories. Proportion of strains with each efflux pump gene in resistant and susceptible categories. The number of strains containing the gene were listed on the right. *P < 0.05, ***P < 0.001. Where there were no statistical analysis results labeled, there were no significant difference
Fig. 5
Fig. 5
Distribution and constitution of CRISPRs in resistant and susceptible categories and a previously unknown CRISPRs combination present exclusively in seven MTZ mono-R strains. The proportions of strains with 0, 1, 2, 3 and 4 CRISPRs in resistant and susceptible categories of MTZ (A), CLA (B) and LEV (C), respectively. **P < 0.01, n.s. not significant. The proportions of CRISPRs with 1, 2, 3, 4 and 6 spacers in resistant and susceptible categories of MTZ (D), CLA (E) and LEV (F), respectively. *P < 0.05, n.s. not significant. G The structural analysis of a previously unknown CRISPRs combination presented exclusively in seven MTZ mono-R strains, including DRs (purple, yellow and red diamonds in each CRISPR) and spacers (grey, blue and black rectangles in each CRISPR). The base sequences are shown below the CRISPR array. The DRs and spacers in the colored characters correspond with the colors of the respective diamonds and rectangles. The DR sequence in box, which exclusively presented in nine MTZ-R strains, indicated that there was a significant difference of the DR frequencies between MTZ-R and MTZ-S categories. *P < 0.05
Fig. 6
Fig. 6
Phylogenetic analysis with respect to antibiotic resistance patterns and specific genetic loci profiles in H. pylori strains. Maximum likelihood phylogenetic tree based on whole-genome level single-copy genes of the 112 H. pylori strains, the resistance patterns, and the significantly different genomic loci observed above between susceptible and resistant categories for the corresponding antibiotic. The susceptible and resistant patterns are denoted by blue and red rectangles, respectively. The presence and absence of the specific loci are denoted by green and grey rectangles, respectively
Fig. 7
Fig. 7
Synoptic representation of the genetic variations in the genome-wide resistance genes and linkage disequilibrium analysis. A The densities of total SNPs and Indels identified in the 112 H. pylori strains against H. pylori 26695 genome are overviewed. The numbers next to the rulers represent the number of SNPs and Indels per unit length of H. pylori 26695 genome. B The proportions of the strains with the nsSNPs and fsIndels present in the H. pylori genome-wide resistant genes in the resistant and susceptible categories of MTZ, CLA and LEV were analyzed, respectively. Only nsSNPs and fsIndels with significantly higher (red cells) or lower (blue cells) frequencies in the resistant categories of MTZ, CLA and LEV were displayed. *P < 0.05, **P < 0.01, ***P < 0.001. Conversely, grey cells indicate the variations not significantly associated with resistance or susceptibility to the antibiotics. Loci, genes, and variations are reported in rows and antibiotics in columns. The nature of the variations is color-coded on the left. The right panel represents the extent of linkage disequilibrium between variations
Fig. 8
Fig. 8
Localization of the nsSNPs and fsIndels in the four proteins with the largest number of resistance-associated variations within the H. pylori genome-wide resistant genes. The available crystal structure of each protein was used to indicate the sites with amino acid changes. A Lon, B BabB, C XerD, and D TrpS. The color scale represents the corresponding antibiotics of the resistance or susceptibility association
Fig. 9
Fig. 9
Cartoon model showing the putative genetic characteristics likely related to H. pylori resistance- or susceptibility- to MTZ, CLA and LEV at locus- and variant-levels. The intrinsic resistance or resistance-associated compensation related genetic features involves the variations in the genome-wide resistant gene-encoding proteins including antibiotic targets, translation-associated factors, transporters, OMPs, proteins probably related to biofilm formation, and other sporadic factors including lon protease with variations in linkage disequilibrium with many others, as well as the presence or absence of a porin, and an efflux pump transporter. In the underlying acquired resistance related genetic elements, the presence or absence of a virulence-associated factor belonging to ComB T4SS and a certain CRISPRs, as well as variations in several recombination required proteins, were demonstrated. Different colors of the genetic element names denote they are in relation to resistance or susceptibility of different antibiotics. Red (MTZ), gold (CLA), pink (LEV), orange (MTZ and CLA), blue (MTZ and LEV), brown (CLA and LEV)

References

    1. Blaser MJ, Perez-Perez GI, Kleanthous H, Cover TL, Peek RM, Chyou PH, Stemmermann GN, Nomura A. Infection with Helicobacter pylori strains possessing cagA is associated with an increased risk of developing adenocarcinoma of the stomach. Cancer Res. 1995;55(10):2111–2115.
    1. Hatakeyama M. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe. 2014;15(3):306–316.
    1. Kuipers EJ, Perez-Perez GI, Meuwissen SG, Blaser MJ. Helicobacter pylori and atrophic gastritis: importance of the cagA status. J Natl Cancer Inst. 1995;87(23):1777–1780.
    1. Chey WD, Leontiadis GI, Howden CW, Moss SF. ACG Clinical guideline: treatment of Helicobacter pylori infection. Am J Gastroenterol. 2017;112(2):212–239.
    1. Nezami BG, Jani M, Alouani D, Rhoads DD, Sadri N. Helicobacter pylori mutations detected by next-generation sequencing in formalin-fixed, paraffin-embedded gastric biopsy specimens are associated with treatment failure. J Clin Microbiol. 2019 doi: 10.1128/JCM.01834-18.
    1. Savoldi A, Carrara E, Graham DY, Conti M, Tacconelli E. Prevalence of antibiotic resistance in Helicobacter pylori: a systematic review and meta-analysis in world health organization regions. Gastroenterology. 2018;155(5):1372–1382 e1317.
    1. Liu WZ, Xie Y, Lu H, Cheng H, Zeng ZR, Zhou LY, Chen Y, Wang JB, Du YQ, Lu NH, et al. Fifth Chinese national consensus report on the management of Helicobacter pylori infection. Helicobacter. 2018;23(2):e12475.
    1. Chen D, Cunningham SA, Cole NC, Kohner PC, Mandrekar JN, Patel R. Phenotypic and molecular antimicrobial susceptibility of Helicobacter pylori. Antimicrob Agents Chemother. 2017 doi: 10.1128/AAC.02530-16.
    1. Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, Monnet DL, Pulcini C, Kahlmeter G, Kluytmans J, Carmeli Y, et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis. 2018;18(3):318–327.
    1. Alba C, Blanco A, Alarcon T. Antibiotic resistance in Helicobacter pylori. Curr Opin Infect Dis. 2017;30(5):489–497.
    1. Gong Y, Yuan Y. Resistance mechanisms of Helicobacter pylori and its dual target precise therapy. Crit Rev Microbiol. 2018;44(3):371–392.
    1. Smiley R, Bailey J, Sethuraman M, Posecion N, Showkat Ali M. Comparative proteomics analysis of sarcosine insoluble outer membrane proteins from clarithromycin resistant and sensitive strains of Helicobacter pylori. J Microbiol. 2013;51(5):612–618.
    1. Co EM, Schiller NL. Resistance mechanisms in an in vitro-selected amoxicillin-resistant strain of Helicobacter pylori. Antimicrob Agents Chemother. 2006;50(12):4174–4176.
    1. Cai Y, Wang C, Chen Z, Xu Z, Li H, Li W, Sun Y. Transporters HP0939, HP0497, and HP0471 participate in intrinsic multidrug resistance and biofilm formation in Helicobacter pylori by enhancing drug efflux. Helicobacter. 2020;25(4):e12715.
    1. Iwamoto A, Tanahashi T, Okada R, Yoshida Y, Kikuchi K, Keida Y, Murakami Y, Yang L, Yamamoto K, Nishiumi S, et al. Whole-genome sequencing of clarithromycin resistant Helicobacter pylori characterizes unidentified variants of multidrug resistant efflux pump genes. Gut Pathog. 2014;6:27.
    1. Krzyżek P, Grande R, Migdał P, Paluch E, Gościniak G. Biofilm formation as a complex result of virulence and adaptive responses of Helicobacter pylori. Pathogens. 2020 doi: 10.3390/pathogens9121062.
    1. Miftahussurur M, Syam AF, Nusi IA, Makmun D, Waskito LA, Zein LH, Akil F, Uwan WB, Simanjuntak D, Wibawa ID, et al. Surveillance of Helicobacter pylori antibiotic susceptibility in indonesia: different resistance types among regions and with novel genetic mutations. PLoS ONE. 2016;11(12):e0166199.
    1. Miftahussurur M, Shrestha PK, Subsomwong P, Sharma RP, Yamaoka Y. Emerging Helicobacter pylori levofloxacin resistance and novel genetic mutation in Nepal. BMC Microbiol. 2016;16(1):256.
    1. Hussein NR, Tunjel I, Majed HS, Yousif ST, Aswad SI, Assafi MS. Duodenal ulcer promoting gene 1 (dupA1) is associated with A2147G clarithromycin-resistance mutation but not interleukin-8 secretion from gastric mucosa in Iraqi patients. New Microbes New Infect. 2015;6:5–10.
    1. Fasciana T, Cala C, Bonura C, Di Carlo E, Matranga D, Scarpulla G, Manganaro M, Camilleri S, Giammanco A. Resistance to clarithromycin and genotypes in Helicobacter pylori strains isolated in Sicily. J Med Microbiol. 2015;64(11):1408–1414.
    1. Li HY, Kao CY, Lin WH, Zheng PX, Yan JJ, Wang MC, Teng CH, Tseng CC, Wu JJ. Characterization of CRISPR-Cas systems in clinical Klebsiella pneumoniae isolates uncovers its potential association with antibiotic susceptibility. Front Microbiol. 2018;9:1595.
    1. Mangas EL, Rubio A, Alvarez-Marin R, Labrador-Herrera G, Pachon J, Pachon-Ibanez ME, Divina F, Perez-Pulido AJ. Pangenome of Acinetobacter baumannii uncovers two groups of genomes, one of them with genes involved in CRISPR/Cas defence systems associated with the absence of plasmids and exclusive genes for biofilm formation. Microb Genom. 2019 doi: 10.1099/mgen.0.000309.
    1. Touchon M, Charpentier S, Pognard D, Picard B, Arlet G, Rocha EP, Denamur E, Branger C. Antibiotic resistance plasmids spread among natural isolates of Escherichia coli in spite of CRISPR elements. Microbiology. 2012;158(12):2997–3004.
    1. Touchon M, Cury J, Yoon EJ, Krizova L, Cerqueira GC, Murphy C, Feldgarden M, Wortman J, Clermont D, Lambert T, et al. The genomic diversification of the whole Acinetobacter genus: origins, mechanisms, and consequences. Genome Biol Evol. 2014;6(10):2866–2882.
    1. Shehreen S, Chyou TY, Fineran PC, Brown CM. Genome-wide correlation analysis suggests different roles of CRISPR-Cas systems in the acquisition of antibiotic resistance genes in diverse species. Philos Trans R Soc Lond B Biol Sci. 2019;374(1772):20180384.
    1. Corbinais C, Mathieu A, Kortulewski T, Radicella JP, Marsin S. Following transforming DNA in Helicobacter pylori from uptake to expression. Mol Microbiol. 2016;101(6):1039–1053.
    1. Jaillard M, van Belkum A, Cady KC, Creely D, Shortridge D, Blanc B, Barbu EM, Dunne WM, Jr, Zambardi G, Enright M, et al. Correlation between phenotypic antibiotic susceptibility and the resistome in Pseudomonas aeruginosa. Int J Antimicrob Agents. 2017;50(2):210–218.
    1. Clinical and Laboratory Standards Institute. Methods for antimicrobial dilution and disk susceptibility testing of infrequently isolated or fastidious bacteria (M45). 3rd ed. 2016.
    1. Jin Y, Deng J, Liang J, Shan C, Tong M. Efficient bacteria capture and inactivation by cetyltrimethylammonium bromide modified magnetic nanoparticles. Colloids Surf B Biointerfaces. 2015;136:659–665.
    1. Schubert M, Lindgreen S, Orlando L. AdapterRemoval v2: rapid adapter trimming, identification, and read merging. BMC Res Notes. 2016;9:88.
    1. Luo R, Liu B, Xie Y, Li Z, Huang W, Yuan J, He G, Chen Y, Pan Q, Liu Y, et al. SOAPdenovo2: an empirically improved memory-efficient short-read de novo assembler. Gigascience. 2012;1(1):18.
    1. Coil D, Jospin G, Darling AE. A5-miseq: an updated pipeline to assemble microbial genomes from Illumina MiSeq data. Bioinformatics. 2015;31(4):587–589.
    1. Delcher AL, Harmon D, Kasif S, White O, Salzberg SL. Improved microbial gene identification with GLIMMER. Nucleic Acids Res. 1999;27(23):4636–4641.
    1. Lowe TM, Eddy SR. tRNAscan-SE: a program for improved detection of transfer RNA genes in genomic sequence. Nucleic Acids Res. 1997;25(5):955–964.
    1. Lagesen K, Hallin P, Rodland EA, Staerfeldt HH, Rognes T, Ussery DW. RNAmmer: consistent and rapid annotation of ribosomal RNA genes. Nucleic Acids Res. 2007;35(9):3100–3108.
    1. Bland C, Ramsey TL, Sabree F, Lowe M, Brown K, Kyrpides NC, Hugenholtz P. CRISPR recognition tool (CRT): a tool for automatic detection of clustered regularly interspaced palindromic repeats. BMC Bioinformatics. 2007;8:209.
    1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol. 1990;215(3):403–410.
    1. Blake JD, Cohen FE. Pairwise sequence alignment below the twilight zone. J Mol Biol. 2001;307(2):721–735.
    1. Powell S, Forslund K, Szklarczyk D, Trachana K, Roth A, Huerta-Cepas J, Gabaldon T, Rattei T, Creevey C, Kuhn M, et al. eggNOG v4.0: nested orthology inference across 3686 organisms. Nucleic Acids Res. 2014;42(Database issue):D231–239.
    1. Fischer S, Brunk BP, Chen F, Gao X, Harb OS, Iodice JB, Shanmugam D, Roos DS, Stoeckert CJ. Using OrthoMCL to assign proteins to OrthoMCL-DB groups or to cluster proteomes into new ortholog groups. Vol 12. In: Goodsell DS, editor. Current protocols in bioinformatics. Hoboken: Wiley; 2011. pp. 11–19.
    1. Chen L, Xiong Z, Sun L, Yang J, Jin Q. VFDB 2012 update: toward the genetic diversity and molecular evolution of bacterial virulence factors. Nucleic Acids Res. 2012;40(Database issue):D641–645.
    1. Bortolaia V, Kaas RS, Ruppe E, Roberts MC, Schwarz S, Cattoir V, Philippon A, Allesoe RL, Rebelo AR, Florensa AF, et al. ResFinder 4.0 for predictions of phenotypes from genotypes. J Antimicrob Chemother. 2020;75(12):3491–3500.
    1. McArthur AG, Waglechner N, Nizam F, Yan A, Azad MA, Baylay AJ, Bhullar K, Canova MJ, De Pascale G, Ejim L, et al. The comprehensive antibiotic resistance database. Antimicrob Agents Chemother. 2013;57(7):3348–3357.
    1. Naas T, Oueslati S, Bonnin RA, Dabos ML, Zavala A, Dortet L, Retailleau P, Iorga BI. Beta-lactamase database (BLDB)—structure and function. J Enzyme Inhib Med Chem. 2017;32(1):917–919.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics. 2009;25(14):1754–1760.
    1. Zhu P, He L, Li Y, Huang W, Xi F, Lin L, Zhi Q, Zhang W, Tang YT, Geng C, et al. Correction: OTG-snpcaller: an optimized pipeline based on TMAP and GATK for SNP calling from ion torrent data. PLoS ONE. 2015;10(9):e0138824.
    1. Ye K, Schulz MH, Long Q, Apweiler R, Ning Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics. 2009;25(21):2865–2871.
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, Maller J, Sklar P, de Bakker PI, Daly MJ, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81(3):559–575.
    1. Waterhouse A, Bertoni M, Bienert S, Studer G, Tauriello G, Gumienny R, Heer FT, de Beer TAP, Rempfer C, Bordoli L, et al. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 2018;46(W1):W296–W303.
    1. Mistry J, Chuguransky S, Williams L, Qureshi M, Salazar GA, Sonnhammer ELL, Tosatto SCE, Paladin L, Raj S, Richardson LJ, et al. Pfam: the protein families database in 2021. Nucleic Acids Res. 2021;49(D1):D412–D419.
    1. Fischbach L, Evans EL. Meta-analysis: the effect of antibiotic resistance status on the efficacy of triple and quadruple first-line therapies for Helicobacter pylori. Aliment Pharmacol Ther. 2007;26(3):343–357.
    1. Malfertheiner P, Megraud F, O'Morain CA, Atherton J, Axon AT, Bazzoli F, Gensini GF, Gisbert JP, Graham DY, Rokkas T, et al. Management of Helicobacter pylori infection—the Maastricht IV/Florence consensus report. Gut. 2012;61(5):646–664.
    1. Megraud F. H pylori antibiotic resistance: prevalence, importance, and advances in testing. Gut. 2004;53(9):1374–1384.
    1. Badr MT, Omar M, Hacker G. Comprehensive integration of genome-wide association and gene expression studies reveals novel gene signatures and potential therapeutic targets for Helicobacter pylori-induced gastric disease. Front Immunol. 2021;12:624117.
    1. Gunaletchumy SP, Seevasant I, Tan MH, Croft LJ, Mitchell HM, Goh KL, Loke MF, Vadivelu J. Helicobacter pylori genetic diversity and gastro-duodenal diseases in Malaysia. Sci Rep. 2014;4:7431.
    1. Falush D, Wirth T, Linz B, Pritchard JK, Stephens M, Kidd M, Blaser MJ, Graham DY, Vacher S, Perez-Perez GI, et al. Traces of human migrations in Helicobacter pylori populations. Science. 2003;299(5612):1582–1585.
    1. Masi M, Winterhalter M, Pages JM. Outer membrane porins. Subcell Biochem. 2019;92:79–123.
    1. Beceiro A, Tomas M, Bou G. Antimicrobial resistance and virulence: a successful or deleterious association in the bacterial world? Clin Microbiol Rev. 2013;26(2):185–230.
    1. Bekhit A, Fukamachi T, Saito H, Kobayashi H. The role of OmpC and OmpF in acidic resistance in Escherichia coli. Biol Pharm Bull. 2011;34(3):330–334.
    1. Liu YF, Yan JJ, Lei HY, Teng CH, Wang MC, Tseng CC, Wu JJ. Loss of outer membrane protein C in Escherichia coli contributes to both antibiotic resistance and escaping antibody-dependent bactericidal activity. Infect Immun. 2012;80(5):1815–1822.
    1. Catel-Ferreira M, Nehme R, Molle V, Aranda J, Bouffartigues E, Chevalier S, Bou G, Jouenne T, De E. Deciphering the function of the outer membrane protein OprD homologue of Acinetobacter baumannii. Antimicrob Agents Chemother. 2012;56(7):3826–3832.
    1. del Mar TM, Beceiro A, Perez A, Velasco D, Moure R, Villanueva R, Martinez-Beltran J, Bou G. Cloning and functional analysis of the gene encoding the 33- to 36-kilodalton outer membrane protein associated with carbapenem resistance in Acinetobacter baumannii. Antimicrob Agents Chemother. 2005;49(12):5172–5175.
    1. Limansky AS, Mussi MA, Viale AM. Loss of a 29-kilodalton outer membrane protein in Acinetobacter baumannii is associated with imipenem resistance. J Clin Microbiol. 2002;40(12):4776–4778.
    1. Sugawara E, Nikaido H. OmpA is the principal nonspecific slow porin of Acinetobacter baumannii. J Bacteriol. 2012;194(15):4089–4096.
    1. Bina J, Bains M, Hancock RE. Functional expression in Escherichia coli and membrane topology of porin HopE, a member of a large family of conserved proteins in Helicobacter pylori. J Bacteriol. 2000;182(9):2370–2375.
    1. Doig P, Exner MM, Hancock RE, Trust TJ. Isolation and characterization of a conserved porin protein from Helicobacter pylori. J Bacteriol. 1995;177(19):5447–5452.
    1. Boehnke KF, Valdivieso M, Bussalleu A, Sexton R, Thompson KC, Osorio S, Novoa Reyes I, Crowley JJ, Baker LH, Xi C. Antibiotic resistance among Helicobacter pylori clinical isolates in Lima. Peru Infect Drug Resist. 2017;10:85–90.
    1. Brennan DE, Dowd C, O'Morain C, McNamara D, Smith SM. Can bacterial virulence factors predict antibiotic resistant Helicobacter pylori infection? World J Gastroenterol. 2018;24(9):971–981.
    1. Haddadi MH, Negahdari B, Asadolahi R, Bazargani A. Helicobacter pylori antibiotic resistance and correlation with cagA motifs and homB gene. Postgrad Med. 2020;132(6):512–520.
    1. Barrozo RM, Hansen LM, Lam AM, Skoog EC, Martin ME, Cai LP, Lin Y, Latoscha A, Suerbaum S, Canfield DR, et al. CagY is an immune-sensitive regulator of the Helicobacter pylori type IV secretion system. Gastroenterology. 2016;151(6):1164–1175 e1163.
    1. Damke PP, Di Guilmi AM, Varela PF, Velours C, Marsin S, Veaute X, Machouri M, Gunjal GV, Rao DN, Charbonnier JB, et al. Identification of the periplasmic DNA receptor for natural transformation of Helicobacter pylori. Nat Commun. 2019;10(1):5357.
    1. Hirata K, Suzuki H, Nishizawa T, Tsugawa H, Muraoka H, Saito Y, Matsuzaki J, Hibi T. Contribution of efflux pumps to clarithromycin resistance in Helicobacter pylori. J Gastroenterol Hepatol. 2010;25(Suppl 1):S75–S79.
    1. Mehrabadi JF, Sirous M, Daryani NE, Eshraghi S, Akbari B, Shirazi MH. Assessing the role of the RND efflux pump in metronidazole resistance of Helicobacter pylori by RT-PCR assay. J Infect Dev Ctries. 2011;5(2):88–93.
    1. Price VJ, McBride SW, Hullahalli K, Chatterjee A, Duerkop BA, Palmer KL. Enterococcus faecalis CRISPR-Cas is a robust barrier to conjugative antibiotic resistance dissemination in the murine intestine. mSphere. 2019 doi: 10.1128/mSphere.00464-19.
    1. Lerminiaux NA, Cameron ADS. Horizontal transfer of antibiotic resistance genes in clinical environments. Can J Microbiol. 2019;65(1):34–44.
    1. Moore ME, Lam A, Bhatnagar S, Solnick JV. Environmental determinants of transformation efficiency in Helicobacter pylori. J Bacteriol. 2014;196(2):337–344.
    1. Levine SM, Lin EA, Emara W, Kang J, DiBenedetto M, Ando T, Falush D, Blaser MJ. Plastic cells and populations: DNA substrate characteristics in Helicobacter pylori transformation define a flexible but conservative system for genomic variation. FASEB J. 2007;21(13):3458–3467.
    1. Aydin S, Personne Y, Newire E, Laverick R, Russell O, Roberts AP, Enne VI. Presence of Type I-F CRISPR/Cas systems is associated with antimicrobial susceptibility in Escherichia coli. J Antimicrob Chemother. 2017;72(8):2213–2218.
    1. Palmer KL, Gilmore MS. Multidrug-resistant enterococci lack CRISPR-cas. mBio. 2010 doi: 10.1128/mBio.00227-10.
    1. Kraft C, Suerbaum S. Mutation and recombination in Helicobacter pylori: mechanisms and role in generating strain diversity. Int J Med Microbiol. 2005;295(5):299–305.
    1. Tseng YS, Wu DC, Chang CY, Kuo CH, Yang YC, Jan CM, Su YC, Kuo FC, Chang LL. Amoxicillin resistance with beta-lactamase production in Helicobacter pylori. Eur J Clin Invest. 2009;39(9):807–812.
    1. Hanafi A, Lee WC, Loke MF, Teh X, Shaari A, Dinarvand M, Lehours P, Megraud F, Leow AH, Vadivelu J, et al. Molecular and proteomic analysis of levofloxacin and metronidazole resistant Helicobacter pylori. Front Microbiol. 2016;7:2015.
    1. Hu Y, Zhang M, Lu B, Dai J. Helicobacter pylori and antibiotic resistance, a continuing and intractable problem. Helicobacter. 2016;21(5):349–363.
    1. Andrianova AG, Kudzhaev AM, Abrikosova VA, Gustchina AE, Smirnov IV, Rotanova TV. Involvement of the N domain residues E34, K35, and R38 in the functionally active structure of Escherichia coli Lon protease. Acta Naturae. 2020;12(4):86–97.
    1. Tzeng SR, Tseng YC, Lin CC, Hsu CY, Huang SJ, Kuo YT, Chang CI. Molecular insights into substrate recognition and discrimination by the N-terminal domain of Lon AAA+ protease. Elife. 2021 doi: 10.7554/eLife.64056.
    1. Pang SS, Nguyen STS, Perry AJ, Day CJ, Panjikar S, Tiralongo J, Whisstock JC, Kwok T. The three-dimensional structure of the extracellular adhesion domain of the sialic acid-binding adhesin SabA from Helicobacter pylori. J Biol Chem. 2014;289(10):6332–6340.
    1. Hathroubi S, Zerebinski J, Ottemann KM. Helicobacter pylori biofilm involves a multigene stress-biased response, including a structural role for flagella. mBio. 2018 doi: 10.1128/mBio.01973-18.
    1. Krzyzek P, Grande R, Migdal P, Paluch E, Gosciniak G. Biofilm formation as a complex result of virulence and adaptive responses of Helicobacter pylori. Pathogens. 2020 doi: 10.3390/pathogens9121062.
    1. Wong EH, Ng CG, Chua EG, Tay AC, Peters F, Marshall BJ, Ho B, Goh KL, Vadivelu J, Loke MF. Comparative genomics revealed multiple helicobacter pylori genes associated with biofilm formation in vitro. PLoS ONE. 2016;11(11):e0166835.
    1. Servetas SL, Doster RS, Kim A, Windham IH, Cha JH, Gaddy JA, Merrell DS. ArsRS-dependent regulation of homB contributes to helicobacter pylori biofilm formation. Front Microbiol. 2018;9:1497.
    1. Yonezawa H, Osaki T, Hojo F, Kamiya S. Effect of Helicobacter pylori biofilm formation on susceptibility to amoxicillin, metronidazole and clarithromycin. Microb Pathog. 2019;132:100–108.

Source: PubMed

3
S'abonner