Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(32): e2401981121, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39078675

RESUMEN

Dihydrouridine (D), a prevalent and evolutionarily conserved base in the transcriptome, primarily resides in tRNAs and, to a lesser extent, in mRNAs. Notably, this modification is found at position 2449 in the Escherichia coli 23S rRNA, strategically positioned near the ribosome's peptidyl transferase site. Despite the prior identification, in E. coli genome, of three dihydrouridine synthases (DUS), a set of NADPH and FMN-dependent enzymes known for introducing D in tRNAs and mRNAs, characterization of the enzyme responsible for D2449 deposition has remained elusive. This study introduces a rapid method for detecting D in rRNA, involving reverse transcriptase-blockage at the rhodamine-labeled D2449 site, followed by PCR amplification (RhoRT-PCR). Through analysis of rRNA from diverse E. coli strains, harboring chromosomal or single-gene deletions, we pinpoint the yhiN gene as the ribosomal dihydrouridine synthase, now designated as RdsA. Biochemical characterizations uncovered RdsA as a unique class of flavoenzymes, dependent on FAD and NADH, with a complex structural topology. In vitro assays demonstrated that RdsA dihydrouridylates a short rRNA transcript mimicking the local structure of the peptidyl transferase site. This suggests an early introduction of this modification before ribosome assembly. Phylogenetic studies unveiled the widespread distribution of the yhiN gene in the bacterial kingdom, emphasizing the conservation of rRNA dihydrouridylation. In a broader context, these findings underscore nature's preference for utilizing reduced flavin in the reduction of uridines and their derivatives.


Asunto(s)
Escherichia coli , Escherichia coli/genética , Escherichia coli/metabolismo , ARN Ribosómico 23S/metabolismo , ARN Ribosómico 23S/genética , ARN Ribosómico 23S/química , Uridina/análogos & derivados , Uridina/metabolismo , Uridina/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/química , ARN Bacteriano/metabolismo , ARN Bacteriano/genética , ARN Bacteriano/química
2.
Front Cell Infect Microbiol ; 11: 803730, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35096652

RESUMEN

Campylobacter jejuni is a major cause of food poisoning worldwide, and remains the main infective agent in gastroenteritis and related intestinal disorders in Europe and the USA. As with all bacterial infections, the stages of adhesion to host tissue, survival in the host and eliciting disease all require the synthesis of proteinaceous virulence factors on the ribosomes of the pathogen. Here, we describe how C. jejuni virulence is attenuated by altering the methylation of its ribosomes to disrupt the composition of its proteome, and how this in turn provides a means of identifying factors that are essential for infection and pathogenesis. Specifically, inactivation of the C. jejuni Cj0588/TlyA methyltransferase prevents methylation of nucleotide C1920 in the 23S rRNA of its ribosomes and reduces the pathogen's ability to form biofilms, to attach, invade and survive in host cells, and to provoke the innate immune response. Mass spectrometric analyses of C. jejuni TlyA-minus strains revealed an array of subtle changes in the proteome composition. These included reduced amounts of the cytolethal distending toxin (CdtC) and the MlaEFD proteins connected with outer membrane vesicle (OMV) production. Inactivation of the cdtC and mlaEFD genes confirmed the importance of their encoded proteins in establishing infection. Collectively, the data identify a subset of genes required for the onset of human campylobacteriosis, and serve as a proof of principle for use of this approach in detecting proteins involved in bacterial pathogenesis.


Asunto(s)
Infecciones por Campylobacter , Campylobacter jejuni , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/metabolismo , Humanos , Metilación , Ribosomas/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
3.
Nucleic Acids Res ; 48(12): 6906-6918, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32459340

RESUMEN

The universal L-shaped tertiary structure of tRNAs is maintained with the help of nucleotide modifications within the D- and T-loops, and these modifications are most extensive within hyperthermophilic species. The obligate-commensal Nanoarchaeum equitans and its phylogenetically-distinct host Ignicoccus hospitalis grow physically coupled under identical hyperthermic conditions. We report here two fundamentally different routes by which these archaea modify the key conserved nucleotide U54 within their tRNA T-loops. In N. equitans, this nucleotide is methylated by the S-adenosylmethionine-dependent enzyme NEQ053 to form m5U54, and a recombinant version of this enzyme maintains specificity for U54 in Escherichia coli. In N. equitans, m5U54 is subsequently thiolated to form m5s2U54. In contrast, I. hospitalis isomerizes U54 to pseudouridine prior to methylating its N1-position and thiolating the O4-position of the nucleobase to form the previously uncharacterized nucleotide m1s4Ψ. The methyl and thiol groups in m1s4Ψ and m5s2U are presented within the T-loop in a spatially identical manner that stabilizes the 3'-endo-anti conformation of nucleotide-54, facilitating stacking onto adjacent nucleotides and reverse-Hoogsteen pairing with nucleotide m1A58. Thus, two distinct structurally-equivalent solutions have evolved independently and convergently to maintain the tertiary fold of tRNAs under extreme hyperthermic conditions.


Asunto(s)
Desulfurococcaceae/genética , Nanoarchaeota/genética , Conformación de Ácido Nucleico , ARN de Transferencia/ultraestructura , Archaea/genética , Archaea/ultraestructura , Escherichia coli/genética , Metilación , Filogenia , ARN de Transferencia/genética , ARNt Metiltransferasas/genética , ARNt Metiltransferasas/ultraestructura
4.
Biomolecules ; 10(4)2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290235

RESUMEN

The C5-methylation of uracil to form 5-methyluracil (m5U) is a ubiquitous base modification of nucleic acids. Four enzyme families have converged to catalyze this methylation using different chemical solutions. Here, we investigate the evolution of 5-methyluracil synthase families in Mollicutes, a class of bacteria that has undergone extensive genome erosion. Many mollicutes have lost some of the m5U methyltransferases present in their common ancestor. Cases of duplication and subsequent shift of function are also described. For example, most members of the Spiroplasma subgroup use the ancestral tetrahydrofolate-dependent TrmFO enzyme to catalyze the formation of m5U54 in tRNA, while a TrmFO paralog (termed RlmFO) is responsible for m5U1939 formation in 23S rRNA. RlmFO has replaced the S-adenosyl-L-methionine (SAM)-enzyme RlmD that adds the same modification in the ancestor and which is still present in mollicutes from the Hominis subgroup. Another paralog of this family, the TrmFO-like protein, has a yet unidentified function that differs from the TrmFO and RlmFO homologs. Despite having evolved towards minimal genomes, the mollicutes possess a repertoire of m5U-modifying enzymes that is highly dynamic and has undergone horizontal transfer.


Asunto(s)
Evolución Molecular , Ácidos Nucleicos/metabolismo , Tenericutes/metabolismo , Uracilo/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Dinitrocresoles/metabolismo , Ácido Fólico/metabolismo , Metilación , Metiltransferasas/metabolismo , Modelos Moleculares , ARN Ribosómico 23S/metabolismo , ARN de Transferencia/metabolismo , Tenericutes/genética
5.
Cell Microbiol ; 22(7): e13199, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32134554

RESUMEN

Campylobacter jejuni is a bacterial pathogen that is generally acquired as a zoonotic infection from poultry and animals. Adhesion of C. jejuni to human colorectal epithelial cells is weakened after loss of its cj0588 gene. The Cj0588 protein belongs to the type I group of TlyA (TlyAI ) enzymes, which 2'-O-methylate nucleotide C1920 in 23S rRNA. Slightly longer TlyAII versions of the methyltransferase are found in actinobacterial species including Mycobacterium tuberculosis, and methylate not only C1920 but also nucleotide C1409 in 16S rRNA. Loss of TlyA function attenuates virulence of both M. tuberculosis and C. jejuni. We show here that the traits impaired in C. jejuni null strains can be rescued by complementation not only with the original cj0588 (tlyA I ) but also with a mycobacterial tlyA II gene. There are, however, significant differences in the recombinant phenotypes. While cj0588 restores motility, biofilm formation, adhesion to and invasion of human epithelial cells and stimulation of IL-8 production in a C. jejuni null strain, several of these properties are further enhanced by the mycobacterial tlyA II gene, in some cases to twice the original wild-type level. These findings strongly suggest that subtle changes in rRNA modification patterns can affect protein synthesis in a manner that has serious consequences for bacterial pathogenicity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Campylobacter jejuni/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Biopelículas , Células CACO-2 , Campylobacter jejuni/genética , Capreomicina , Células Epiteliales , Regulación Bacteriana de la Expresión Génica , Genes de ARNr/genética , Humanos , Macrófagos , Metilación , Ratones , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/genética , Células RAW 264.7 , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Virulencia , Factores de Virulencia/genética
6.
J Antimicrob Chemother ; 75(2): 371-378, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31670815

RESUMEN

OBJECTIVES: To determine the mechanism of induction of erm(47) and its atypical expression in the Gram-positive opportunistic pathogen Helcococcus kunzii, where it confers resistance to a subset of clinically important macrolide, lincosamide and streptogramin B (MLSB) antibiotics. METHODS: The resistant H. kunzii clinical isolate UCN99 was challenged with subinhibitory concentrations of a wide range of ribosome-targeting drugs. The methylation status of the H. kunzii ribosomal RNA at the MLSB binding site was then determined using an MS approach and was correlated with any increase in resistance to the drugs. RESULTS: The H. kunzii erm(47) gene encodes a monomethyltransferase. Expression is induced by subinhibitory concentrations of the macrolide erythromycin, as is common for many erm genes, and surprisingly also by 16-membered macrolide, lincosamide, streptogramin, ketolide, chloramphenicol and linezolid antibiotics, all of which target the 50S ribosomal subunit. No induction was detected with spectinomycin, which targets the 30S subunit. CONCLUSIONS: The structure of the erm(47) leader sequence functions as a hair trigger for the induction mechanism that expresses resistance. Consequently, translation of the erm(47) mRNA is tripped by MLSB compounds and also by drugs that target the 50S ribosomal subunit outside the MLSB site. Expression of erm(47) thus extends previous assumptions about how erm genes can be induced.


Asunto(s)
Firmicutes , Lincosamidas , Macrólidos , Metiltransferasas , Estreptogramina B , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Firmicutes/efectos de los fármacos , Firmicutes/enzimología , Lincosamidas/farmacología , Macrólidos/farmacología , Metiltransferasas/genética , Ribosomas , Estreptogramina B/farmacología
7.
Front Microbiol ; 9: 1329, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29997583

RESUMEN

Strains of the Pasteurellaceae bacteria Pasteurella multocida and Mannheimia haemolytica are major etiological agents of bovine respiratory disease (BRD). Treatment of BRD with antimicrobials is becoming more challenging due to the increasing occurrence of resistance in infecting strains. In Pasteurellaceae strains exhibiting resistance to multiple antimicrobials including aminoglycosides, beta-lactams, macrolides and sulfonamides, the resistance determinants are often chromosomally encoded within integrative and conjugative elements (ICEs). To gain a more comprehensive picture of ICE structures, we sequenced the genomes of six strains of P. multocida and four strains of M. haemolytica; all strains were independent isolates and eight of them were multiple-resistant. ICE sequences varied in size from 49 to 79 kb, and were comprised of an array of conserved genes within a core region and varieties of resistance genes within accessory regions. These latter regions mainly account for the variation in the overall ICE sizes. From the sequence data, we developed a multiplex PCR assay targeting four conserved core genes required for integration and maintenance of ICE structures. Application of this assay on 75 isolates of P. multocida and M. haemolytica reveals how the presence and structures of ICEs are related to their antibiotic resistance phenotypes. The assay is also applicable to other members of the Pasteurellaceae family including Histophilus somni and indicates how clustering and dissemination of the resistance genes came about.

8.
Genome Announc ; 6(18)2018 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-29724850

RESUMEN

The chemically mutagenized Escherichia coli strain AS19 was isolated on the basis of its enhanced sensitivity to different antibiotics, in particular to actinomycin. The strain was later modified to study rRNA modifications that confer antibiotic resistance. Here, we present the genome sequence of the variant E. coli AS19-RrmA.

9.
Nucleic Acids Res ; 45(4): 2007-2015, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28204608

RESUMEN

In all free-living organisms a late-stage checkpoint in the biogenesis of the small ribosomal subunit involves rRNA modification by an RsmA/Dim1 methyltransferase. The hyperthermophilic archaeon Nanoarchaeum equitans, whose existence is confined to the surface of a second archaeon, Ignicoccus hospitalis, lacks an RsmA/Dim1 homolog. We demonstrate here that the I. hospitalis host possesses the homolog Igni_1059, which dimethylates the N6-positions of two invariant adenosines within helix 45 of 16S rRNA in a manner identical to other RsmA/Dim1 enzymes. However, Igni_1059 is not transferred from I. hospitalis to N. equitans across their fused cell membrane structures and the corresponding nucleotides in N. equitans 16S rRNA remain unmethylated. An alternative mechanism for ribosomal subunit maturation in N. equitans is suggested by sRNA interactions that span the redundant RsmA/Dim1 site to introduce 2΄-O-ribose methylations within helices 44 and 45 of the rRNA.


Asunto(s)
Adenosina/metabolismo , Metiltransferasas/metabolismo , Nanoarchaeota/genética , ARN Ribosómico 16S/metabolismo , Desulfurococcaceae/enzimología , Desulfurococcaceae/genética , Escherichia coli/genética , Metilación , Metiltransferasas/genética , Nanoarchaeota/enzimología , ARN Ribosómico 16S/química , Subunidades Ribosómicas Pequeñas de Archaea/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-29404277

RESUMEN

Numerous bacterial pathogens express an ortholog of the enzyme TlyA, which is an rRNA 2'-O-methyltransferase associated with resistance to cyclic peptide antibiotics such as capreomycin. Several other virulence traits have also been attributed to TlyA, and these appear to be unrelated to its methyltransferase activity. The bacterial pathogen Campylobacter jejuni possesses the TlyA homolog Cj0588, which has been shown to contribute to virulence. Here, we investigate the mechanism of Cj0588 action and demonstrate that it is a type I homolog of TlyA that 2'-O-methylates 23S rRNA nucleotide C1920. This same specific function is retained by Cj0588 both in vitro and also when expressed in Escherichia coli. Deletion of the cj0588 gene in C. jejuni or substitution with alanine of K80, D162, or K188 in the catalytic center of the enzyme cause complete loss of 2'-O-methylation activity. Cofactor interactions remain unchanged and binding affinity to the ribosomal substrate is only slightly reduced, indicating that the inactivated proteins are folded correctly. The substitution mutations thus dissociate the 2'-O-methylation function of Cj0588/TlyA from any other putative roles that the protein might play. C. jejuni strains expressing catalytically inactive versions of Cj0588 have the same phenotype as cj0588-null mutants, and show altered tolerance to capreomycin due to perturbed ribosomal subunit association, reduced motility and impaired ability to form biofilms. These functions are reestablished when methyltransferase activity is restored and we conclude that the contribution of Cj0588 to virulence in C. jejuni is a consequence of the enzyme's ability to methylate its rRNA.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Campylobacter jejuni/enzimología , Campylobacter jejuni/fisiología , Locomoción , ARN Ribosómico 23S/metabolismo , ARNt Metiltransferasas/metabolismo , Sustitución de Aminoácidos , Campylobacter jejuni/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Eliminación de Gen , Expresión Génica , Metilación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Virulencia/metabolismo , ARNt Metiltransferasas/genética
11.
Hum Mol Genet ; 24(25): 7286-94, 2015 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-26464487

RESUMEN

Mitochondrial dysfunction is a well-established cause of sensorineural deafness, but the pathophysiological events are poorly understood. Non-syndromic deafness and predisposition to aminoglycoside-induced deafness can be caused by specific mutations in the 12S rRNA gene of mtDNA and are thus maternally inherited traits. The pathophysiology induced by mtDNA mutations has traditionally been attributed to deficient oxidative phosphorylation, which causes energy crisis with functional impairment of multiple cellular processes. In contrast, it was recently reported that signaling induced by 'hypermethylation' of two conserved adenosines of 12S rRNA in the mitoribosome is of key pathophysiological importance in sensorineural deafness. In support for this concept, it was reported that overexpression of the essential mitochondrial methyltransferase TFB1M in the mouse was sufficient to induce mitoribosomal hypermethylation and deafness. At variance with this model, we show here that 12S rRNA is near fully methylated in vivo in the mouse and thus cannot be further methylated to any significant extent. Furthermore, bacterial artificial chromosome transgenic mice overexpressing TFB1M have no increase of 12S rRNA methylation levels and hear normally. We thus conclude that therapies directed against mitoribosomal methylation are unlikely to be beneficial to patients with sensorineural hearing loss or other types of mitochondrial disease.


Asunto(s)
ADN Mitocondrial/genética , Audición/genética , Ribosomas Mitocondriales/metabolismo , Factores de Transcripción/genética , Animales , Sordera/genética , Femenino , Pérdida Auditiva Sensorineural/inducido químicamente , Pérdida Auditiva Sensorineural/genética , Masculino , Metilación , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mutación Puntual/genética , ARN Ribosómico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Proc Natl Acad Sci U S A ; 112(42): 12956-61, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438831

RESUMEN

Ketolides are promising new antimicrobials effective against a broad range of Gram-positive pathogens, in part because of the low propensity of these drugs to trigger the expression of resistance genes. A natural ketolide pikromycin and a related compound methymycin are produced by Streptomyces venezuelae strain ATCC 15439. The producer avoids the inhibitory effects of its own antibiotics by expressing two paralogous rRNA methylase genes pikR1 and pikR2 with seemingly redundant functions. We show here that the PikR1 and PikR2 enzymes mono- and dimethylate, respectively, the N6 amino group in 23S rRNA nucleotide A2058. PikR1 monomethylase is constitutively expressed; it confers low resistance at low fitness cost and is required for ketolide-induced activation of pikR2 to attain high-level resistance. The regulatory mechanism controlling pikR2 expression has been evolutionary optimized for preferential activation by ketolide antibiotics. The resistance genes and the induction mechanism remain fully functional when transferred to heterologous bacterial hosts. The anticipated wide use of ketolide antibiotics could promote horizontal transfer of these highly efficient resistance genes to pathogens. Taken together, these findings emphasized the need for surveillance of pikR1/pikR2-based bacterial resistance and the preemptive development of drugs that can remain effective against the ketolide-specific resistance mechanism.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Cetólidos/farmacología , Metiltransferasas/genética , ARN Ribosómico 23S/genética
13.
J Antimicrob Chemother ; 70(2): 420-3, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25261417

RESUMEN

OBJECTIVES: To determine how resistance to macrolides is conferred in field isolates of Pasteurella multocida and Mannheimia haemolytica that lack previously identified resistance determinants for rRNA methylation, efflux and macrolide-modifying enzymes. METHODS: Isolates of P. multocida and M. haemolytica identified as being highly resistant (MICs >64 mg/L) to the macrolides erythromycin, gamithromycin, tilmicosin, tildipirosin and tulathromycin were screened by multiplex PCR for the previously identified resistance genes erm(42), msr(E) and mph(E). Strains lacking these determinants were analysed by genome sequencing and primer extension on the rRNAs. RESULTS: Macrolide resistance in one M. haemolytica isolate was conferred by the 23S rRNA mutation A2058G; resistance in three P. multocida isolates were caused by mutations at the neighbouring nucleotide A2059G. In each strain, all six copies of the rrn operons encoded the respective mutations. There were no mutations in the ribosomal protein genes rplD or rplV, and no other macrolide resistance mechanism was evident. CONCLUSIONS: High-level macrolide resistance can arise from 23S rRNA mutations in P. multocida and M. haemolytica despite their multiple copies of rrn. Selective pressures from exposure to different macrolide or lincosamide drugs presumably resulted in consolidation of either the A2058G or the A2059G mutation.


Asunto(s)
Farmacorresistencia Bacteriana , Genes de ARNr , Macrólidos/farmacología , Mannheimia haemolytica/efectos de los fármacos , Mannheimia haemolytica/genética , Mutación , Pasteurella multocida/efectos de los fármacos , Pasteurella multocida/genética , Antibacterianos/farmacología , Genoma Bacteriano , Humanos , Mannheimia haemolytica/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Operón , Pasteurella multocida/aislamiento & purificación , ARN Ribosómico 23S/genética
14.
Nucleic Acids Res ; 42(12): 8073-82, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24939895

RESUMEN

Efficient protein synthesis in all organisms requires the post-transcriptional methylation of specific ribosomal ribonucleic acid (rRNA) and transfer RNA (tRNA) nucleotides. The methylation reactions are almost invariably catalyzed by enzymes that use S-adenosylmethionine (AdoMet) as the methyl group donor. One noteworthy exception is seen in some bacteria, where the conserved tRNA methylation at m5U54 is added by the enzyme TrmFO using flavin adenine dinucleotide together with N5,N10-methylenetetrahydrofolate as the one-carbon donor. The minimalist bacterium Mycoplasma capricolum possesses two homologs of trmFO, but surprisingly lacks the m5U54 tRNA modification. We created single and dual deletions of the trmFO homologs using a novel synthetic biology approach. Subsequent analysis of the M. capricolum RNAs by mass spectrometry shows that the TrmFO homolog encoded by Mcap0476 specifically modifies m5U1939 in 23S rRNA, a conserved methylation catalyzed by AdoMet-dependent enzymes in all other characterized bacteria. The Mcap0476 methyltransferase (renamed RlmFO) represents the first folate-dependent flavoprotein seen to modify ribosomal RNA.


Asunto(s)
Proteínas Bacterianas/metabolismo , Flavoproteínas/metabolismo , Metiltransferasas/metabolismo , Mycoplasma capricolum/enzimología , ARN Ribosómico 23S/metabolismo , Proteínas Bacterianas/genética , Biocatálisis , Flavoproteínas/genética , Metilación , Metiltransferasas/genética , Mycoplasma capricolum/genética , ARN Ribosómico 23S/química , ARN de Transferencia/metabolismo , Uridina/metabolismo
15.
RNA Biol ; 10(8): 1324-32, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23948732

RESUMEN

Aminoglycoside antibiotics remain the drugs of choice for treatment of Pseudomonas aeruginosa infections, particularly for respiratory complications in cystic-fibrosis patients. Previous studies on other bacteria have shown that aminoglycosides have their primary target within the decoding region of 16S rRNA helix 44 with a secondary target in 23S rRNA helix 69. Here, we have mapped P. aeruginosa rRNAs using MALDI mass spectrometry and reverse transcriptase primer extension to identify nucleotide modifications that could influence aminoglycoside interactions. Helices 44 and 45 contain indigenous (housekeeping) modifications at m (4)Cm1402, m (3)U1498, m (2)G1516, m (6) 2A1518, and m (6) 2A1519; helix 69 is modified at m (3)Ψ1915, with m (5)U1939 and m (5)C1962 modification in adjacent sequences. All modifications were close to stoichiometric, with the exception of m (3)Ψ1915, where about 80% of rRNA molecules were methylated. The modification status of a virulent clinical strain expressing the acquired methyltransferase RmtD was altered in two important respects: RmtD stoichiometrically modified m (7)G1405 conferring high resistance to the aminoglycoside tobramycin and, in doing so, impeded one of the methylation reactions at C1402. Mapping the nucleotide methylations in P. aeruginosa rRNAs is an essential step toward understanding the architecture of the aminoglycoside binding sites and the rational design of improved drugs against this bacterial pathogen.


Asunto(s)
Aminoglicósidos/química , Pseudomonas aeruginosa/genética , ARN Ribosómico 16S/química , ARN Ribosómico 16S/metabolismo , ARN Ribosómico 23S/química , ARN Ribosómico 23S/metabolismo , Aminoglicósidos/genética , Aminoglicósidos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Dicroismo Circular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Metilación , Metiltransferasas/química , Metiltransferasas/genética , Metiltransferasas/metabolismo , Modelos Moleculares , Conformación de Ácido Nucleico , Estructura Terciaria de Proteína , Pseudomonas aeruginosa/metabolismo , ARN Bacteriano/química , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Ribosómico 16S/genética , ARN Ribosómico 23S/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
16.
J Antimicrob Chemother ; 68(7): 1543-50, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23580560

RESUMEN

OBJECTIVES: 16S rRNA methyltransferases are an emerging mechanism conferring high-level resistance to clinically relevant aminoglycosides and have been associated with important mechanisms such as NDM-1. We sought genes encoding these enzymes in isolates highly resistant (MIC >200 mg/L) to gentamicin and amikacin from an Indian hospital and we additionally screened for the novel RmtF enzyme in 132 UK isolates containing NDM. METHODS: All highly aminoglycoside-resistant isolates were screened for armA and rmtA-E by PCR, with cloning experiments performed for isolates negative for these genes. Matrix-assisted laser desorption/ionization (MALDI) mass spectrometry was used to determine the methylation target of the novel RmtF methyltransferase. RmtF-bearing strains were characterized further, including susceptibility testing, PFGE, electroporation, PCR-based replicon typing and multilocus sequence typing of rmtF-bearing plasmids. RESULTS: High-level aminoglycoside resistance was detected in 140/1000 (14%) consecutive isolates of Enterobacteriaceae from India. ArmA, RmtB and RmtC were identified among 46%, 20% and 27% of these isolates, respectively. The novel rmtF gene was detected in 34 aminoglycoside-resistant isolates (overall prevalence 3.4%), most (59%) of which also possessed a bla(NDM) gene; rmtF was detected in 6 NDM producers from the UK. It was found on different plasmid backbones. Four and two isolates showed resistance to tigecycline and colistin, respectively. CONCLUSIONS: RmtF was often found in association with NDM in members of the Enterobacteriaceae and on diverse plasmids. It is of clinical concern that the RmtF- and NDM-positive strains identified here show additional resistance to tigecycline and colistin, current drugs of last resort for the treatment of serious bacterial infections.


Asunto(s)
Aminoglicósidos/farmacología , Proteínas Bacterianas/genética , Carbapenémicos/farmacología , Farmacorresistencia Bacteriana Múltiple , Enterobacteriaceae/efectos de los fármacos , Enterobacteriaceae/genética , Metiltransferasas/genética , beta-Lactamasas/genética , Amicacina/farmacología , ADN Bacteriano/química , ADN Bacteriano/genética , Enterobacteriaceae/enzimología , Enterobacteriaceae/aislamiento & purificación , Infecciones por Enterobacteriaceae/microbiología , Gentamicinas/farmacología , Humanos , India , Datos de Secuencia Molecular , Plásmidos , Análisis de Secuencia de ADN , Reino Unido
17.
Antimicrob Agents Chemother ; 56(11): 6033-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22926570

RESUMEN

Tildipirosin is a 16-membered-ring macrolide developed to treat bacterial pathogens, including Mannheimia haemolytica and Pasteurella multocida, that cause respiratory tract infections in cattle and swine. Here we evaluated the efficacy of tildipirosin at inhibiting protein synthesis on the ribosome (50% inhibitory concentration [IC(50)], 0.23 ± 0.01 µM) and compared it with the established veterinary macrolides tylosin, tilmicosin, and tulathromycin. Mutation and methylation at key rRNA nucleotides revealed differences in the interactions of these macrolides within their common ribosomal binding site.


Asunto(s)
Antibacterianos/química , Mannheimia haemolytica/efectos de los fármacos , Pasteurella multocida/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Ribosomas/efectos de los fármacos , Tilosina/análogos & derivados , Drogas Veterinarias/química , Antibacterianos/farmacología , Sitios de Unión , Disacáridos/química , Disacáridos/farmacología , Escherichia coli/química , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/farmacología , Mannheimia haemolytica/química , Mannheimia haemolytica/genética , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mutación , Nucleótidos/genética , Pasteurella multocida/química , Pasteurella multocida/genética , ARN Ribosómico/química , ARN Ribosómico/genética , Ribosomas/química , Fracciones Subcelulares/química , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Transcripción Genética/efectos de los fármacos , Tilosina/química , Tilosina/farmacología , Drogas Veterinarias/farmacología
18.
Mol Microbiol ; 85(6): 1194-203, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22779429

RESUMEN

The binding site of the cyclic peptide antibiotics capreomycin and viomycin is located on the ribosomal subunit interface close to nucleotides C1409 in 16S rRNA and C1920 in 23S rRNA. In Mycobacterium tuberculosis, the 2'-hydroxyls of both nucleotides are methylated by the enzyme TlyA. Loss of these methylations through inactivation of TlyA confers resistance to capreomycin and viomycin. We report here that TlyA orthologues occur in diverse bacteria and fall into two distinct groups. One group, now termed TlyA(I) , has shorter N- and C-termini and methylates only C1920; the second group (now TlyA(II) ) includes the mycobacterial enzyme, and these longer orthologues methylate at both C1409 and C1920. Ribosomal subunits are the preferred substrates for both groups of orthologues. Amino acid substitutions at the N-terminus of TlyA(II) reduce its ability to methylate these substrates. Growing pairs of recombinant TlyA(II) Escherichia coli strains in competition shows that even subtle changes in the level of rRNA methylation lead to significant differences in susceptibility to sub-inhibitory concentrations of capreomycin. The findings reveal that 2'-O-methyls at both C1409 and C1920 play a role in facilitating the inhibitory effects of capreomycin and viomycin on the bacterial ribosome.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/enzimología , Proteínas Bacterianas/metabolismo , Capreomicina/farmacología , ARN Ribosómico/metabolismo , ARNt Metiltransferasas/metabolismo , Proteínas Bacterianas/genética , Metilación , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación de Ácido Nucleico , Subunidades Ribosómicas/metabolismo , Viomicina/farmacología , ARNt Metiltransferasas/genética
19.
ACS Chem Biol ; 7(8): 1351-5, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22563863

RESUMEN

The veterinary antibiotic tildipirosin (20,23-dipiperidinyl-mycaminosyl-tylonolide, Zuprevo) was developed recently to treat bovine and swine respiratory tract infections caused by bacterial pathogens such as Pasteurella multocida. Tildipirosin is a derivative of the naturally occurring compound tylosin. Here, we define drug-target interactions by combining chemical footprinting with structure modeling and show that tildipirosin, tylosin, and an earlier tylosin derivative, tilmicosin (20-dimethylpiperidinyl-mycaminosyl-tylonolide, Micotil), bind to the same macrolide site within the large subunit of P. multocida and Escherichia coli ribosomes. The drugs nevertheless differ in how they occupy this site. Interactions of the two piperidine components, which are unique to tildipirosin, distinguish this drug from tylosin and tilmicosin. The 23-piperidine of tildipirosin contacts ribosomal residues on the tunnel wall while its 20-piperidine is oriented into the tunnel lumen and is positioned to interfere with the growing nascent peptide.


Asunto(s)
Macrólidos/química , Ribosomas/química , Tilosina/análogos & derivados , Antibacterianos/química , Sitios de Unión , Escherichia coli/metabolismo , Modelos Químicos , Modelos Moleculares , Pasteurella multocida/metabolismo , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , ARN Ribosómico/metabolismo , Tilosina/química
20.
Antimicrob Agents Chemother ; 56(7): 3664-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22564832

RESUMEN

The bacterial pathogens Mannheimia haemolytica and Pasteurella multocida are major etiological agents in respiratory tract infections of cattle. Although these infections can generally be successfully treated with veterinary macrolide antibiotics, a few recent isolates have shown resistance to these drugs. Macrolide resistance in members of the family Pasteurellaceae is conferred by combinations of at least three genes: erm(42), which encodes a monomethyltransferase and confers a type I MLS(B) (macrolide, lincosamide, and streptogramin B) phenotype; msr(E), which encodes a macrolide efflux pump; and mph(E), which encodes a macrolide-inactivating phosphotransferase. Here, we describe a multiplex PCR assay that detects the presence of erm(42), msr(E), and mph(E) and differentiates between these genes. In addition, the assay distinguishes P. multocida from M. haemolytica by amplifying distinctive fragments of the 23S rRNA (rrl) genes. One rrl fragment acts as a general indicator of gammaproteobacterial species and confirms whether the PCR assay has functioned as intended on strains that are negative for erm(42), msr(E), and mph(E). The multiplex system has been tested on more than 40 selected isolates of P. multocida and M. haemolytica and correlated with MICs for the veterinary macrolides tulathromycin and tilmicosin, and the newer compounds gamithromycin and tildipirosin. The multiplex PCR system gives a rapid and robustly accurate determination of macrolide resistance genotypes and bacterial genus, matching results from microbiological methods and whole-genome sequencing.


Asunto(s)
Antibacterianos/farmacología , Macrólidos/farmacología , Mannheimia haemolytica/efectos de los fármacos , Reacción en Cadena de la Polimerasa Multiplex/métodos , Pasteurella multocida/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Lincosamidas/farmacología , Mannheimia haemolytica/genética , Pasteurella multocida/genética , Estreptogramina B/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA