The complex methylome of the human gastric pathogen Helicobacter pylori

Juliane Krebes, Richard D Morgan, Boyke Bunk, Cathrin Spröer, Khai Luong, Raphael Parusel, Brian P Anton, Christoph König, Christine Josenhans, Jörg Overmann, Richard J Roberts, Jonas Korlach, Sebastian Suerbaum, Juliane Krebes, Richard D Morgan, Boyke Bunk, Cathrin Spröer, Khai Luong, Raphael Parusel, Brian P Anton, Christoph König, Christine Josenhans, Jörg Overmann, Richard J Roberts, Jonas Korlach, Sebastian Suerbaum

Abstract

The genome of Helicobacter pylori is remarkable for its large number of restriction-modification (R-M) systems, and strain-specific diversity in R-M systems has been suggested to limit natural transformation, the major driving force of genetic diversification in H. pylori. We have determined the comprehensive methylomes of two H. pylori strains at single base resolution, using Single Molecule Real-Time (SMRT®) sequencing. For strains 26695 and J99-R3, 17 and 22 methylated sequence motifs were identified, respectively. For most motifs, almost all sites occurring in the genome were detected as methylated. Twelve novel methylation patterns corresponding to nine recognition sequences were detected (26695, 3; J99-R3, 6). Functional inactivation, correction of frameshifts as well as cloning and expression of candidate methyltransferases (MTases) permitted not only the functional characterization of multiple, yet undescribed, MTases, but also revealed novel features of both Type I and Type II R-M systems, including frameshift-mediated changes of sequence specificity and the interaction of one MTase with two alternative specificity subunits resulting in different methylation patterns. The methylomes of these well-characterized H. pylori strains will provide a valuable resource for future studies investigating the role of H. pylori R-M systems in limiting transformation as well as in gene regulation and host interaction.

Figures

Figure 1.
Figure 1.
Circos plots displaying the distribution of methylated bases in the genomes of H. pylori 26695 (A) and J99-R3 (B). The outermost and innermost tracks represent the interpulse duration ratios. The colored tracks in between represent the location of methylation of the different motifs. Each motif is displayed by one track except for the asymmetric Type I recognition sites for which both motifs were combined. For m6A and m4C methylation, the cutoff for calling a site modified was Qmod >100 (kinetic score); for the TET-treated m5C methylation a kinetic score threshold >50 was used. The position and type of methylation for each motif is depicted in the legend. Novel motifs are highlighted in red. The genome tick marks display the genomic positions (kbp). The locations of the cag pathogenicity island (cagPAI) and PZ are indicated. Please note that the PZ of strain 26695 is split into two regions (21).
Figure 2.
Figure 2.
Graphical representation of the silenced JHP0414/JHP0415 Type I MTase of J99-R3 (A), the putative phase-variable Type III MTases (B), BcgI-like R-M systems (C) and Type ISP MTases (D). (A) The gene sequences of HsdM (JHP0415) and HsdS (JHP0414) are displayed as gray bars and the protein sequences as yellow bars. SMRT sequencing of the J99-R3 genome did not reveal methylation by the Type I MTase complex (dashed lines), while expression of the enzyme complex on a plasmid in E. coli 2796 and subsequent SMRT analyses demonstrate methylation of the recognition site AAAN6TGG. (B–D) The gene sequences (gray arrows) contain one or two putative or authentic frameshifts that would prevent translation of full-length proteins (colored bars). Each frameshift was repaired through site-directed mutagenesis (addition and deletion of an indicated number of nucleotides, +15 Nt—TAAGGTTAATATATG) and correction was verified by targeted Sanger sequencing. The activity of the MTase proteins was pretested with a methylation activity assay. If an MTase showed activity in the assay, the gDNA of the corresponding E. coli host strain ER2796 was subjected to SMRT sequencing and analyzed for methylation. ‘mut’ marks alleles with frameshift corrections, ‘trunc’ labels not fully translated alleles, ‘n.d.’ points out that no SMRT sequencing was performed and dashed lines indicate that no methylation could be detected through SMRT sequencing. Filled inverted triangles highlight the position of the frameshift mutations. JHP—ORFs of J99, HP—ORFs of 26695. Homologous systems are displayed in the same color.
Figure 3.
Figure 3.
Functional characterization of JHP0785/JHP0786 and JHP0726 through insertion mutagenesis (A) and expression of the R-M genes in E. coli 2796 (B) confirmed that the Type I MTase JHP0786 interacts with the two S subunits JHP0785 or JHP0726 to achieve methylation of two different recognition sites. The methylation patterns of the J99-R3 parental and mutant genomes and the E. coli ER2796 host genomes were analyzed by SMRT sequencing. Motifs displayed in gray are not modified. Methylation of a recognition site is indicated by coloring and boldface type. The methylated nucleotide within a site is highlighted by the addition of a methyl group (CH3) to the sequence. ‘M’ is the abbreviation for methyltransferase, which acts as a homodimer. The two S subunits are composed of either one or two TRDs and interaction of TRDs is achieved by helical connector regions (H) adjacent to each TRD. (A) Expression of the three R-M components in J99-R3 and functional inactivation of either component (labeled by a black cross) resulted in different methylation patterns of the two distinct recognition sites. (B) Analysis of enzyme activity through plasmid-based expression of either JHP0786/JHP0785 (pRRS) alone or together with JHP0726 (pACYC184) in E. coli.
Figure 4.
Figure 4.
Graphical representation of the unique ability of the Type I MTase complex JHP1422/JHP1423 (HsdS/HsdM) to recognize and methylate two distinct recognition sites. (A) Schematic representation of the domain structure of the JHP1422 specificity subunit (HsdS) deduced from the J99 genome sequence (21). The proposed protein harbors three TRDs of which TRD1 is unique and TRD2 and 3 are duplicated as indicated by the coloring. Helical connector regions (H), known as conserved regions (CR), flank each TRD. (B) Inactivation of JHP1423 (HsdM) by insertion mutagenesis and subsequent SMRT analysis resulted in a lack of methylation of two different recognition sites as indicated by the gray color of the two sites. (C) Joint expression of JHP1423 (M) and several allele variants of JHP1422 led to different methylation patterns in the E. coli host genome. Cloning of JHP1422/JHP1423 yielded two naturally occurring allelic variants of JHP1422 (left panel: allele variant 1 and 2, JHP1423 sequence not shown) that differ in composition and protein length. In contrast to the proposed sequence, the H2 region following TRD2 is replaced by the H1 for allele variant 1. Allele variant 2 shows the same exchange but, in addition, has a second TRD2 flanked by the H2 region. Both variants have a different impact on methylation of the two recognition sites (right panel). Allele variants 3–6 were obtained by mutagenesis to reveal the specificity of the TRDs. Coloring and boldface type highlight methylation of the recognition sites, and the methyl group (CH3) indicates the modified nucleotide. Recognition sequences displayed in gray are not modified. The methylation patterns were analyzed by SMRT sequencing of the J99-R3jhp1423 mutant genome (B) and E. coli ER2796 genomes (C).
Figure 5.
Figure 5.
Graphical representation of the specificity switching phenomenon of the two homologous Type IIG MTases JHP1272 and HP1353-HP1354. (A) The gene sequences (gray arrows) contain two putative or authentic frameshifts that would prevent translation of full-length proteins (blue bars). Each frameshift was repaired through site-directed mutagenesis [addition or deletion of C nucleotide(s) is indicated] and correction was verified by targeted Sanger sequencing. Plasmid DNA (Hpy99XIV) or gDNA (remainder) of the E. coli host strains ER2796 was subjected to SMRT sequencing and analyzed for methylation. ‘mut’ marks alleles with frameshift corrections, ‘n.d.’ points out that no SMRT sequencing was performed and dashed lines indicate that no methylation could be detected. Filled inverted triangles highlight the position of the frameshift mutations. JHP—ORF of J99, HP—ORF of 26695. (B–D) Portions of the amino-acid multiple sequence alignments including secondary structure predictions (raw output of PROMALS) of the MTases JHP1272 and HP1353-HP1354. The numbers preceding each sequence display the position within the amino acid sequence. An asterisk at the end of a sequence indicates a stop codon. CTRD—C-terminal additional recognition domain, TRD—target recognition domain. (B) Secondary structure-guided alignment of JHP1272 and HP1353 CTRD and TRD with two MmeI family enzymes. The alignment shows the similarity of both MTases to the secondary structure of MmeI and MaqI in the MmeI region known to specify recognition of the −2 and −1 bases (relative to the methylated base within a recognition site). For these positions a changing specificity in the two active variants of JHP1272 and HP1353-HP1354 was observed. The red color text indicates predicted alpha helices and blue indicates predicted beta strands. The arrows indicate the four beta strands forming the structural motif in MmeI family enzymes that present −2 and −1 contact amino acids. The underlined PPPP region represents the first homopolymeric repeat of C nucleotides. (C) Putative recognition region for −1 and −2 base positions within the regular TRD and CTRD of the two MTases. Amino acid identities are depicted in bold black, similarities in bold green. (D) Alignment of HP1353-HP1354 and JHP1272 CTRD. Both regions show 87% amino acid identity (82 of 94), which most likely accounts for the identical specificity switch for −1 and −2 base positions observed for both protein variants containing the CTRD.

References

    1. Suerbaum S, Michetti P. Helicobacter pylori infection. N. Engl. J. Med. 2002;347:1175–1186.
    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;299:1582–1585.
    1. Suerbaum S, Josenhans C. Helicobacter pylori evolution and phenotypic diversification in a changing host. Nat. Rev. Microbiol. 2007;5:441–452.
    1. Linz B, Balloux F, Moodley Y, Manica A, Liu H, Roumagnac P, Falush D, Stamer C, Prugnolle F, van der Merwe SW, et al. An African origin for the intimate association between humans and Helicobacter pylori. Nature. 2007;445:915–918.
    1. Bjorkholm B, Sjolund M, Falk PG, Berg OG, Engstrand L, Andersson DI. Mutation frequency and biological cost of antibiotic resistance in Helicobacter pylori. Proc. Natl Acad. Sci. USA. 2001;98:14607–14612.
    1. Suerbaum S, Maynard Smith J, Bapumia K, Morelli G, Smith NH, Kunstmann E, Dyrek I, Achtman M. Free recombination within Helicobacter pylori. Proc. Natl Acad. Sci. USA. 1998;95:12619–12624.
    1. Schwarz S, Morelli G, Kusecek B, Manica A, Balloux F, Owen RJ, Graham DY, van der MS, Achtman M, Suerbaum S. Horizontal versus familial transmission of Helicobacter pylori. PLoS Pathog. 2008;4:e1000180.
    1. Kennemann L, Didelot X, Aebischer T, Kuhn S, Drescher B, Droege M, Reinhardt R, Correa P, Meyer TF, Josenhans C, et al. Helicobacter pylori genome evolution during human infection. Proc. Natl Acad. Sci. USA. 2011;108:5033–5038.
    1. Falush D, Kraft C, Taylor NS, Correa P, Fox JG, Achtman M, Suerbaum S. Recombination and mutation during long-term gastric colonization by Helicobacter pylori: estimates of clock rates, recombination size, and minimal age. Proc. Natl Acad. Sci. USA. 2001;98:15056–15061.
    1. Ando T, Xu Q, Torres M, Kusugami K, Israel DA, Blaser MJ. Restriction-modification system differences in Helicobacter pylori are a barrier to interstrain plasmid transfer. Mol. Microbiol. 2000;37:1052–1065.
    1. Aras RA, Small AJ, Ando T, Blaser MJ. Helicobacter pylori interstrain restriction-modification diversity prevents genome subversion by chromosomal DNA from competing strains. Nucleic Acids Res. 2002;30:5391–5397.
    1. Humbert O, Dorer MS, Salama NR. Characterization of Helicobacter pylori factors that control transformation frequency and integration length during inter-strain DNA recombination. Mol. Microbiol. 2011;79:387–401.
    1. Roberts RJ, Vincze T, Posfai J, Macelis D. REBASE–a database for DNA restriction and modification: enzymes, genes and genomes. Nucleic Acids Res. 2010;38:D234–D236.
    1. Luria SE, Human ML. A nonhereditary, host-induced variation of bacterial viruses. J. Bacteriol. 1952;64:557–569.
    1. Bertani G, Weigle JJ. Host controlled variation in bacterial viruses. J. Bacteriol. 1953;65:113–121.
    1. Wilson GG. Organization of restriction-modification systems. Nucleic Acids Res. 1991;19:2539–2566.
    1. Roberts RJ, Belfort M, Bestor T, Bhagwat AS, Bickle TA, Bitinaite J, Blumenthal RM, Degtyarev SK, Dryden DT, Dybvig K, et al. A nomenclature for restriction enzymes, DNA methyltransferases, homing endonucleases and their genes. Nucleic Acids Res. 2003;31:1805–1812.
    1. Raleigh EA, Wilson G. Escherichia coli K-12 restricts DNA containing 5-methylcytosine. Proc. Natl Acad. Sci. USA. 1986;83:9070–9074.
    1. Tomb JF, White O, Kerlavage AR, Clayton RA, Sutton GG, Fleischmann RD, Ketchum KA, Klenk HP, Gill S, Dougherty BA, et al. The complete genome sequence of the gastric pathogen Helicobacter pylori. Nature. 1997;388:539–547.
    1. Nobusato A, Uchiyama I, Kobayashi I. Diversity of restriction-modification gene homologues in Helicobacter pylori. Gene. 2000;259:89–98.
    1. Alm RA, Ling LS, Moir DT, King BL, Brown ED, Doig PC, Smith DR, Noonan B, Guild BC, deJonge BL, et al. Genomic-sequence comparison of two unrelated isolates of the human gastric pathogen Helicobacter pylori. Nature. 1999;397:176–180.
    1. Kong H, Lin LF, Porter N, Stickel S, Byrd D, Posfai J, Roberts RJ. Functional analysis of putative restriction-modification system genes in the Helicobacter pylori J99 genome. Nucleic Acids Res. 2000;28:3216–3223.
    1. Lin LF, Posfai J, Roberts RJ, Kong H. Comparative genomics of the restriction-modification systems in Helicobacter pylori. Proc. Natl Acad. Sci. USA. 2001;98:2740–2745.
    1. Vitkute J, Stankevicius K, Tamulaitiene G, Maneliene Z, Timinskas A, Berg DE, Janulaitis A. Specificities of eleven different DNA methyltransferases of Helicobacter pylori strain 26695. J. Bacteriol. 2001;183:443–450.
    1. Banerjee A, Rao DN. Functional analysis of an acid adaptive DNA adenine methyltransferase from Helicobacter pylori 26695. PLoS One. 2011;6:e16810.
    1. Flusberg BA, Webster DR, Lee JH, Travers KJ, Olivares EC, Clark TA, Korlach J, Turner SW. Direct detection of DNA methylation during single-molecule, real-time sequencing. Nat. Methods. 2010;7:461–465.
    1. Clark TA, Murray IA, Morgan RD, Kislyuk AO, Spittle KE, Boitano M, Fomenkov A, Roberts RJ, Korlach J. Characterization of DNA methyltransferase specificities using single-molecule, real-time DNA sequencing. Nucleic Acids Res. 2012;40:e29.
    1. Murray IA, Clark TA, Morgan RD, Boitano M, Anton BP, Luong K, Fomenkov A, Turner SW, Korlach J, Roberts RJ. The methylomes of six bacteria. Nucleic Acids Res. 2012;40:11450–11462.
    1. Fang G, Munera D, Friedman DI, Mandlik A, Chao MC, Banerjee O, Feng Z, Losic B, Mahajan MC, Jabado OJ, et al. Genome-wide mapping of methylated adenine residues in pathogenic Escherichia coli using single-molecule real-time sequencing. Nat. Biotechnol. 2012;30:1232–1239.
    1. Lluch-Senar M, Luong K, Llorens-Rico V, Delgado J, Fang G, Spittle K, Clark TA, Schadt E, Turner SW, Korlach J, et al. Comprehensive methylome characterization of Mycoplasma genitalium and Mycoplasma pneumoniae at single-base resolution. PLoS Genet. 2013;9:e1003191.
    1. Kulick S, Moccia C, Didelot X, Falush D, Kraft C, Suerbaum S. Mosaic DNA imports with interspersions of recipient sequence after natural transformation of Helicobacter pylori. PLoS One. 2008;3:e3797.
    1. Moccia C, Krebes J, Kulick S, Didelot X, Kraft C, Bahlawane C, Suerbaum S. The nucleotide excision repair (NER) system of Helicobacter pylori: role in mutation prevention and chromosomal import patterns after natural transformation. BMC Microbiol. 2012;12:67.
    1. Sambrook J, Russell DG. 2004 Molecular Cloning: A Laboratory ManuaL. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY.
    1. He YF, Li BZ, Li Z, Liu P, Wang Y, Tang Q, Ding J, Jia Y, Chen Z, Li L, et al. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science. 2011;333:1303–1307.
    1. Ferrero RL, Cussac V, Courcoux P, Labigne A. Construction of isogenic urease-negative mutants of Helicobacter pylori by allelic exchange. J. Bacteriol. 1992;174:4212–4217.
    1. Haas R, Meyer TF, van Putten JP. Aflagellated mutants of Helicobacter pylori generated by genetic transformation of naturally competent strains using transposon shuttle mutagenesis. Mol. Microbiol. 1993;8:753–760.
    1. Kulick S, Moccia C, Kraft C, Suerbaum S. The Helicobacter pylori mutY homologue HP0142 is an antimutator gene that prevents specific C to A transversions. Arch. Microbiol. 2008;189:263–270.
    1. Labigne-Roussel A, Courcoux P, Tompkins L. Gene disruption and replacement as a feasible approach for mutagenesis of Campylobacter jejuni. J. Bacteriol. 1988;170:1704–1708.
    1. Schmitz A, Josenhans C, Suerbaum S. Cloning and characterization of the Helicobacter pylori flbA gene, which codes for a membrane protein involved in coordinated expression of flagellar genes. J. Bacteriol. 1997;179:987–997.
    1. Morgan RD, Luyten YA. Rational engineering of type II restriction endonuclease DNA binding and cleavage specificity. Nucleic Acids Res. 2009;37:5222–5233.
    1. Pradhan S, Bacolla A, Wells RD, Roberts RJ. Recombinant human DNA (cytosine-5) methyltransferase. I. Expression, purification, and comparison of de novo and maintenance methylation. J. Biol. Chem. 1999;274:33002–33010.
    1. Morgan RD, Dwinell EA, Bhatia TK, Lang EM, Luyten YA. The MmeI family: type II restriction-modification enzymes that employ single-strand modification for host protection. Nucleic Acids Res. 2009;37:5208–5221.
    1. Humbert O, Salama NR. The Helicobacter pylori HpyAXII restriction-modification system limits exogenous DNA uptake by targeting GTAC sites but shows asymmetric conservation of the DNA methyltransferase and restriction endonuclease components. Nucleic Acids Res. 2008;36:6893–6906.
    1. Smith RM, Diffin FM, Savery NJ, Josephsen J, Szczelkun MD. DNA cleavage and methylation specificity of the single polypeptide restriction-modification enzyme LlaGI. Nucleic Acids Res. 2009;37:7206–7218.
    1. Ito S, Shen L, Dai Q, Wu SC, Collins LB, Swenberg JA, He C, Zhang Y. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science. 2011;333:1300–1303.
    1. Clark TA, Lu X, Luong K, Dai Q, Boitano M, Turner SW, He C, Korlach J. Enhanced 5-methylcytosine detection in single-molecule, real-time sequencing via Tet1 oxidation. BMC Biol. 2013;11:4.
    1. Censini S, Lange C, Xiang Z, Crabtree JE, Ghiara P, Borodovsky M, Rappuoli R, Covacci A. cag, a pathogenicity island of Helicobacter pylori, encodes type I-specific and disease-associated virulence factors. Proc. Natl Acad. Sci. USA. 1996;93:14648–14653.
    1. Gressmann H, Linz B, Ghai R, Pleissner KP, Schlapbach R, Yamaoka Y, Kraft C, Suerbaum S, Meyer TF, Achtman M. Gain and loss of multiple genes during the evolution of Helicobacter pylori. PLoS Genet. 2005;1:e43.
    1. Dryden DTF, Murray NE, Rao DN. Nucleoside triphosphate-dependent restriction enzymes. Nucleic Acids Res. 2001;29:3728–3741.
    1. Furuta Y, Kawai M, Uchiyama I, Kobayashi I. Domain movement within a gene: a novel evolutionary mechanism for protein diversification. PLoS One. 2011;6:e18819.
    1. Abadjieva A, Patel J, Webb M, Zinkevich V, Firman K. A deletion mutant of the type IC restriction endonuclease EcoR1241 expressing a novel DNA specificity. Nucleic Acids Res. 1993;21:4435–4443.
    1. MacWilliams MP, Bickle TA. Generation of new DNA binding specificity by truncation of the type IC EcoDXXI hsdS gene. EMBO J. 1996;15:4775–4783.
    1. Janulaitis A, Vaisvila R, Timinskas A, Klimasauskas S, Butkus V. Cloning and sequence analysis of the genes coding for Eco57I type IV restriction-modification enzymes. Nucleic Acids Res. 1992;20:6051–6056.
    1. Morgan RD, Bhatia TK, Lovasco L, Davis TB. MmeI: a minimal Type II restriction-modification system that only modifies one DNA strand for host protection. Nucleic Acids Res. 2008;36:6558–6570.
    1. Saunders NJ, Peden JF, Hood DW, Moxon ER. Simple sequence repeats in the Helicobacter pylori genome. Mol. Microbiol. 1998;27:1091–1098.
    1. Peck B, Ortkamp M, Diehl KD, Hundt E, Knapp B. Conservation, localization and expression of HopZ, a protein involved in adhesion of Helicobacter pylori. Nucleic Acids Res. 1999;27:3325–3333.
    1. Ilver D, Arnqvist A, Ogren J, Frick IM, Kersulyte D, Incecik ET, Berg DE, Covacci A, Engstrand L, Boren T. Helicobacter pylori adhesin binding fucosylated histo-blood group antigens revealed by retagging. Science. 1998;279:373–377.
    1. Solnick JV, Hansen LM, Salama NR, Boonjakuakul JK, Syvanen M. Modification of Helicobacter pylori outer membrane protein expression during experimental infection of rhesus macaques. Proc. Natl Acad. Sci. USA. 2004;101:2106–2111.
    1. Mahdavi J, Sonden B, Hurtig M, Olfat FO, Forsberg L, Roche N, Angstrom J, Larsson T, Teneberg S, Karlsson KA, et al. Helicobacter pylori SabA adhesin in persistent infection and chronic inflammation. Science. 2002;297:573–578.
    1. Goodwin AC, Weinberger DM, Ford CB, Nelson JC, Snider JD, Hall JD, Paules CI, Peek RM, Jr, Forsyth MH. Expression of the Helicobacter pylori adhesin SabA is controlled via phase variation and the ArsRS signal transduction system. Microbiology. 2008;154:2231–2240.
    1. Josenhans C, Eaton KA, Thevenot T, Suerbaum S. Switching of flagellar motility in Helicobacter pylori by reversible length variation of a short homopolymeric sequence repeat in fliP, a gene encoding a basal body protein. Infect. Immun. 2000;68:4598–4603.
    1. de Vries N, Duinsbergen D, Kuipers EJ, Pot RG, Wiesenekker P, Penn CW, van Vliet AH, Vandenbroucke-Grauls CM, Kusters JG. Transcriptional phase variation of a type III restriction-modification system in Helicobacter pylori. J. Bacteriol. 2002;184:6615–6623.
    1. Srikhanta YN, Gorrell RJ, Steen JA, Gawthorne JA, Kwok T, Grimmond SM, Robins-Browne RM, Jennings MP. Phasevarion mediated epigenetic gene regulation in Helicobacter pylori. PLoS One. 2011;6:e27569.
    1. Blaser MJ, Kirschner D. The equilibria that allow bacterial persistence in human hosts. Nature. 2007;449:843–849.
    1. Srikhanta YN, Maguire TL, Stacey KJ, Grimmond SM, Jennings MP. The phasevarion: a genetic system controlling coordinated, random switching of expression of multiple genes. Proc. Natl Acad. Sci. USA. 2005;102:5547–5551.
    1. Srikhanta YN, Dowideit SJ, Edwards JL, Falsetta ML, Wu HJ, Harrison OB, Fox KL, Seib KL, Maguire TL, Wang AH, et al. Phasevarions mediate random switching of gene expression in pathogenic Neisseria. PLoS Pathog. 2009;5:e1000400.
    1. Srikhanta YN, Fox KL, Jennings MP. The phasevarion: phase variation of type III DNA methyltransferases controls coordinated switching in multiple genes. Nat. Rev. Microbiol. 2010;8:196–206.
    1. Hanahan D. Studies on transformation of Escherichia coli with plasmids. J. Mol. Biol. 1983;166:557–580.
    1. Sibley MH, Raleigh EA. Cassette-like variation of restriction enzyme genes in Escherichia coli C and relatives. Nucleic Acids Res. 2004;32:522–534.
    1. Casadaban MJ, Cohen SN. Analysis of gene control signals by DNA fusion and cloning in Escherichia coli. J. Mol. Biol. 1980;138:179–207.
    1. Chang AC, Cohen SN. Construction and characterization of amplifiable multicopy DNA cloning vehicles derived from the P15A cryptic miniplasmid. J. Bacteriol. 1978;134:1141–1156.
    1. Rose RE. The nucleotide sequence of pACYC184. Nucleic Acids Res. 1988;16:355.
    1. Skoglund CM, Smith HO, Chandrasegaran S. Construction of an efficient overproducer clone of HinfI restriction endonuclease using the polymerase chain reaction. Gene. 1990;88:1–5.
    1. Yanisch-Perron C, Vieira J, Messing J. Improved M13 phage cloning vectors and host strains: nucleotide sequences of the M13mp18 and pUC19 vectors. Gene. 1985;33:103–119.

Source: PubMed

3
Abonnieren