Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Iran J Microbiol ; 3(2): 58-67, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22347584

ABSTRACT

BACKGROUND AND OBJECTIVES: Streptococcus pneumoniae, a major human pathogen, is closely related to the commensal species S. mitis and S. oralis. S. pneumoniae surface proteins are implicated in virulence and host interaction of this species, but many of them have recently been detected in S. mitis B6 in silico. We tested for the presence of such genes usinga set of eight S. mitis and eleven S. oralis strains from different geographic locations. MATERIALS AND METHODS: An oligonucleotide microarray was designed based on the genomes of S. pneumoniae R6 and TIGR4 as well as S. mitis B6 to include 63 cell surface proteins. The S. pneumoniae genes encoding neuraminidases, hyaluronidase and pneumolysin were also included. In addition to comparative genomic hybridization experiments, homologues were identified in silico in the genome of S. oralis Uo5. RESULTS AND CONCLUSIONS: The results document that many S. pneumoniae related surface proteins are ubiquitously present among the Mitis group of streptococci. All 19 samples hybridized with the pavA probe representing a gene important for adherence and invasion of S. pneumoniae. Only eight genes were not recognized in any strain, including the S. pneumoniae PcpC gene as the only virulence gene of the S. pneumoniae core genome.The fact that only 12 out of 26 genes present in the S. oralis Uo5 genome could be detected by microarray analysis confirms the sequence variation of surface components.

2.
FEMS Microbiol Lett ; 205(1): 99-104, 2001 Nov 27.
Article in English | MEDLINE | ID: mdl-11728722

ABSTRACT

The surface-associated subtilisin-like serine protease PrtA was identified by screening a genomic expression library from Streptococcus pneumoniae using a convalescent-phase serum. In Western blot analysis two forms of PrtA were detected in whole cell lysate and a truncated form only in culture supernatant suggesting that PrtA is produced as a precursor protein, translocated to the cell surface, truncated, and released into the surroundings. A 5' fragment of the gene was found highly conserved among 78 pneumococcal isolates of clinical relevance. Immunogenicity of PrtA, limited genetic variation, and the involvement in pneumococcal virulence demonstrated in in vivo experiments might identify PrtA as a promising candidate for a protein based vaccine.


Subject(s)
Cell Wall/enzymology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/pathogenicity , Amino Acid Sequence , Animals , Conserved Sequence , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pneumococcal Infections/mortality , Sequence Analysis, DNA , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Virulence
3.
J Clin Microbiol ; 39(7): 2565-71, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11427569

ABSTRACT

The emergence of disease caused by penicillin-resistant and multidrug-resistant pneumococci has become a global concern, necessitating the identification of the epidemiological spread of such strains. The Pneumococcal Molecular Epidemiology Network was established in 1997 under the auspices of the International Union of Microbiological Societies with the aim of characterizing, standardizing, naming, and classifying antimicrobial agent-resistant pneumococcal clones. Here we describe the nomenclature for 16 pneumococcal clones that have contributed to the increase in antimicrobial resistance worldwide. Guidelines for the recognition of these clones using molecular typing procedures (pulsed-field gel electrophoresis, BOX-PCR, and multilocus sequence typing) are presented, as are the penicillin-binding profiles and macrolide resistance determinants for the 16 clones. This network can serve as a prototype for the collaboration of scientists in identifying clones of important human pathogens and as a model for the development of other networks.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Erythromycin/pharmacology , Hexosyltransferases , Peptidyl Transferases , Pneumococcal Infections/epidemiology , Streptococcus pneumoniae , Terminology as Topic , Bacterial Typing Techniques , Carrier Proteins/genetics , DNA, Bacterial/analysis , Drug Resistance, Microbial/genetics , Humans , Microbial Sensitivity Tests , Molecular Epidemiology , Muramoylpentapeptide Carboxypeptidase/genetics , Penicillin-Binding Proteins , Pneumococcal Infections/microbiology , Polymerase Chain Reaction/methods , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics
4.
Infect Immun ; 69(4): 2477-86, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254610

ABSTRACT

Streptococcus pneumoniae remains a major causative agent of serious human diseases. The worldwide increase of antibiotic resistant strains revealed the importance of horizontal gene transfer in this pathogen, a scenario that results in the modulation of the species-specific gene pool. We investigated genomic variation in 20 S. pneumoniae isolates representing major antibiotic-resistant clones and 10 different capsular serotypes. Variation was scored as decreased hybridization signals visualized on a high-density oligonucleotide array representing 1,968 genes of the type 4 reference strain KNR.7/87. Up to 10% of the genes appeared altered between individual isolates and the reference strain; variability within clones was below 2.1%. Ten gene clusters covering 160 kb account for half of the variable genes. Most of them are associated with transposases and are assumed to be part of a flexible gene pool within the bacterial population; other variable loci include mosaic genes encoding antibiotic resistance determinants and gene clusters related to bacteriocin production. Genomic comparison between S. pneumoniae and commensal Streptococcus mitis and Streptococcus oralis strains indicates distinct antigenic profiles and suggests a smooth transition between these species, supporting the validity of the microarray system as an epidemiological and diagnostic tool.


Subject(s)
Chromosomes, Bacterial , Genome, Bacterial , Streptococcus pneumoniae/genetics , Alleles , Base Sequence , Drug Resistance, Microbial , Genetic Variation , Humans , Molecular Sequence Data , Mosaicism , Streptococcus pneumoniae/drug effects , Trimethoprim/pharmacology
5.
Infect Immun ; 69(2): 845-52, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159977

ABSTRACT

In pneumococcal meningitis it is assumed that bacteria cross the blood-brain barrier (BBB), which consists mainly of cerebral endothelial cells. The effect of Streptococcus pneumoniae on the BBB was investigated with an in vitro BBB model using a human brain microvascular endothelial cell line (HBMEC) and primary cultures of bovine brain microvascular endothelial cells (BBMEC). Within a few hours of incubation with pneumococci, rounding and detachment of the HBMEC were observed, and the transendothelial electrical resistance of the BBMEC monolayer decreased markedly. An S. pneumoniae mutant deficient in pneumolysin did not affect the integrity of the endothelial cell monolayer. Neither cell wall fragments nor isolated pneumococcal cell walls induced changes of endothelial cell morphology. However, purified pneumolysin caused endothelial cell damage comparable to that caused by the viable pneumococci. The cell detachment was dependent on de novo protein synthesis and required the activities of caspase and tyrosine kinases. The results show that pneumolysin is an important component for damaging the BBB and may contribute to the entry of pneumococci into the cerebral compartment and to the development of brain edema in pneumococcal meningitis.


Subject(s)
Brain/blood supply , Endothelium, Vascular/drug effects , Streptococcus pneumoniae/pathogenicity , Streptolysins/toxicity , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Blood-Brain Barrier , Caspases/physiology , Cattle , Cell Wall/physiology , Endothelium, Vascular/pathology , Hot Temperature , Microcirculation/drug effects , Phosphorylation , Protein Biosynthesis , Rats
6.
Methods Mol Med ; 48: 265-71, 2001.
Article in English | MEDLINE | ID: mdl-21374426

ABSTRACT

Penicillin-resistance in Streptococcus pneumoniae is mediated by altered penicillin-target enzymes, the penicillin-binding proteins or PBPs. PBPs interact withß-lactam antibiotics by forming an active penicilloyl-PBP complex via an active site serine. This complex is enzymatically inactive, and stable enough so that it can be visualized by incubating cells, cell lysates or membrane fractions with radioactive ß-lactam, followed by SDS-polyacrylamidegel electrophoresis (PAGE) and fluorography. The increasing frequency of ß-lactam resistant isolates necessitates techniques for describing such strains. PBP profile analysis allows the detection of the variation in six proteins simultaneously and thus each PBP profile is basically a fingerprint of the strain, allowing to assign hundreds of isolates into distinct clonal groups (1, 2).

7.
FEMS Microbiol Lett ; 190(2): 231-6, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11034284

ABSTRACT

The peptide SpiP of Streptococcus pneumoniae regulates the induction of a complex signal transduction system spiR1spiR2spiH. Distinct alleles of spiP and the receptor histidine protein kinase gene spiH were recognized in different pneumococcal clones. The spi system in strain KNR7/87 is adjacent to a bacteriocin gene cluster encoding putative double glycine-type bacteriocins, immunity proteins, and translocator proteins. A direct repeat element upstream of the spiR1 promoter and another three potential transcription start sites within the bacteriocin cluster indicate that SpiP functions as an inducing peptide for bacteriocin synthesis in S. pneumoniae.


Subject(s)
Alleles , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Genetic Variation , Signal Transduction , Streptococcus pneumoniae/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacteriocins/biosynthesis , Bacteriocins/genetics , Base Sequence , Genes, Bacterial , Molecular Sequence Data , Multigene Family , Protein Sorting Signals/genetics , Streptococcus pneumoniae/metabolism
8.
J Bacteriol ; 182(20): 5919-21, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11004197

ABSTRACT

The beta-galactosidase gene of Streptococcus pneumoniae, bgaA, encodes a putative 2,235-amino-acid protein with the two amino acid motifs characteristic of the glycosyl hydrolase family of proteins. In addition, an N-terminal signal sequence and a C-terminal LPXTG motif typical of surface-associated proteins of gram-positive bacteria are present. Trypsin treatment of cells resulted in solubilization of the enzyme, documenting that it is associated with the cell envelope. In order to obtain defined mutants suitable for lacZ reporter experiments, the bgaA gene was disrupted, resulting in a complete absence of endogenous beta-galactosidase activity. The results are consistent with beta-galactosidase being a surface protein that seems not to be involved in lactose metabolism but that may play a role during pathogenesis.


Subject(s)
Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism , Amino Acid Sequence , Cell Membrane/enzymology , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Sorting Signals/chemistry , Protein Sorting Signals/metabolism , Reading Frames , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Trypsin , beta-Galactosidase/chemistry
9.
Res Microbiol ; 151(6): 453-6, 2000.
Article in English | MEDLINE | ID: mdl-10961458

ABSTRACT

The presence of highly divergent mosaic blocks in penicillin binding protein genes responsible for penicillin resistance in Streptococcus pneumoniae implies that transformation is an important tool for the evolution of this pathogen. Genetic competence depends on production of the competence signaling peptide CSP, the processed product of comC, which is curiously part of a mosaic gene arrangement itself. Expression of comC is part of a complex regulatory network involving at least two receptor kinase/transcriptional regulator pairs: ComD/E, which is responsible for induction, and CiaH/R, which inhibits expression of the comCDE operon.


Subject(s)
Genes, Bacterial , Hexosyltransferases , Mosaicism , Multienzyme Complexes , Peptidyl Transferases , Streptococcus pneumoniae/genetics , Transformation, Bacterial , Bacterial Proteins/genetics , Carrier Proteins/genetics , Genetic Variation , Histidine Kinase , Muramoylpentapeptide Carboxypeptidase/genetics , Penicillin-Binding Proteins , Protein Kinases/genetics
10.
FEMS Microbiol Lett ; 188(1): 81-5, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10867238

ABSTRACT

Penicillin resistance in pneumococci is mediated by modified penicillin-binding proteins (PBPs) that have decreased affinity to beta-lactams. In high-level penicillin-resistant transformants of the laboratory strain Streptococcus pneumoniae R6 containing various combinations of low-affinity PBPs, disruption of the fib locus results in a collapse of PBP-mediated resistance. In addition, crosslinked muropeptides are highly reduced. The fib operon consists of two genes, fibA and fibB, homologous to Staphylococcus aureus femA/B which are also required for expression of methicillin resistance in this organism. FibA and FibB belong to a family of proteins of Gram-positive bacteria involved in the formation of interpeptide bridges, thus representing interesting new targets for antimicrobial compounds for this group of pathogens.


Subject(s)
Bacterial Proteins/genetics , Carrier Proteins/metabolism , Hexosyltransferases , Muramoylpentapeptide Carboxypeptidase/metabolism , Operon , Peptidoglycan/metabolism , Peptidyl Transferases , Streptococcus pneumoniae/genetics , beta-Lactam Resistance/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chromatography, High Pressure Liquid , Microbial Sensitivity Tests , Molecular Sequence Data , Mutagenesis, Insertional , Penicillin-Binding Proteins , Peptidoglycan/chemistry , Sequence Analysis, DNA , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/metabolism , beta-Lactams/pharmacology
11.
Infect Immun ; 68(6): 3740-3, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10816539

ABSTRACT

A genomic expression library of Streptococcus pneumoniae was screened with a convalescent-phase serum for immunoreactive proteins. Six known and 17 unknown pneumococcal proteins were detected. Five of the known proteins were surface-located virulence factors, and eight of the unknown proteins were putative membrane proteins.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/isolation & purification , Bacterial Proteins/isolation & purification , Membrane Proteins/isolation & purification , Pneumococcal Infections/immunology , Bacterial Proteins/immunology , Convalescence , Membrane Proteins/immunology , Virulence
12.
Mol Microbiol ; 33(4): 673-8, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10447877

ABSTRACT

The beta-lactams are by far the most widely used and efficacious of all antibiotics. Over the past few decades, however, widespread resistance has evolved among most common pathogens. Streptococcus pneumoniae has become a paradigm for understanding the evolution of resistance mechanisms, the simplest of which, by far, is the production of beta-lactamases. As these enzymes are frequently plasmid encoded, resistance can readily be transmitted between bacteria. Despite the fact that pneumococci are naturally transformable organisms, no beta-lactamase-producing strain has yet been described. A much more complex resistance mechanism has evolved in S. pneumoniae that is mediated by a sophisticated restructuring of the targets of the beta-lactams, the penicillin-binding proteins (PBPs); however, this may not be the whole story. Recently, a third level of resistance mechanisms has been identified in laboratory mutants, wherein non-PBP genes are mutated and resistance development is accompanied by deficiency in genetic transformation. Two such non-PBP genes have been described: a putative glycosyltransferase, CpoA, and a histidine protein kinase, CiaH. We propose that these non-PBP genes are involved in the biosynthesis of cell wall components at a step prior to the biosynthetic functions of PBPs, and that the mutations selected during beta-lactam treatment counteract the effects caused by the inhibition of penicillin-binding proteins.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Carrier Proteins/metabolism , Hexosyltransferases , Muramoylpentapeptide Carboxypeptidase/metabolism , Peptidyl Transferases , Streptococcus pneumoniae/drug effects , beta-Lactams/pharmacology , Glycosyltransferases/genetics , Histidine Kinase , Models, Molecular , Mutation , Penicillin-Binding Proteins , Protein Kinases/genetics , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , beta-Lactam Resistance
14.
J Biol Chem ; 274(27): 19175-80, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10383423

ABSTRACT

Penicillin-binding protein 2x (PBP2x) isolated from clinical beta-lactam-resistant strains of Streptococcus pneumoniae (R-PBP2x) have a reduced affinity for beta-lactam antibiotics. Their transpeptidase domain carries numerous substitutions compared with homologous sequences from beta-lactam-sensitive streptococci (S-PBP2x). Comparison of R-PBP2x sequences suggested that the mutation Gln552 --> Glu is important for resistance development. Mutants selected in the laboratory with cephalosporins frequently contain a mutation Thr550 --> Ala. The high resolution structure of a complex between S-PBP2x* and cefuroxime revealed that Gln552 and Thr550, which belong to strand beta3, are in direct contact with the cephalosporin. We have studied the effect of alterations at positions 552 and 550 in soluble S-PBP2x (S-PBP2x*) expressed in Escherichia coli. Mutation Q552E lowered the acylation efficiency for both penicillin G and cefotaxime when compared with S-PBP2x*. We propose that the introduction of a negative charge in strand beta3 conflicts with the negative charge of the beta-lactam. Mutation T550A lowered the acylation efficiency of the protein for cefotaxime but not for penicillin G. The in vitro data presented here are in agreement with the distinct resistance profiles mediated by these mutations in vivo and underline their role as powerful resistance determinants.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Mutation , Penicillin-Binding Proteins , Streptococcus pneumoniae/genetics , Acylation , Binding Sites , Cefuroxime/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Penicillin G/metabolism , Penicillin Resistance/genetics , Protein Structure, Secondary , Streptococcus pneumoniae/metabolism
15.
J Bacteriol ; 181(12): 3852-6, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10368166

ABSTRACT

One group of penicillin target enzymes, the class A high-molecular-weight penicillin-binding proteins (PBPs), are bimodular enzymes. In addition to a central penicillin-binding-transpeptidase domain, they contain an N-terminal putative glycosyltransferase domain. Mutations in the genes for each of the three Streptococcus pneumoniae class A PBPs, PBP1a, PBP1b, and PBP2a, were isolated by insertion duplication mutagenesis within the glycosyltransferase domain, documenting that their function is not essential for cellular growth in the laboratory. PBP1b PBP2a and PBP1a PBP1b double mutants could also be isolated, and both showed defects in positioning of the septum. Attempts to obtain a PBP2a PBP1a double mutant failed. All mutants with a disrupted pbp2a gene showed higher sensitivity to moenomycin, an antibiotic known to inhibit PBP-associated glycosyltransferase activity, indicating that PBP2a is the primary target for glycosyltransferase inhibitors in S. pneumoniae.


Subject(s)
Bacterial Proteins , Carrier Proteins/metabolism , Escherichia coli Proteins , Hexosyltransferases/metabolism , Multienzyme Complexes/metabolism , Muramoylpentapeptide Carboxypeptidase/metabolism , Peptide Synthases , Peptidoglycan Glycosyltransferase , Peptidyl Transferases/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Escherichia coli/drug effects , Hexosyltransferases/chemistry , Hexosyltransferases/genetics , Microbial Sensitivity Tests , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Muramoylpentapeptide Carboxypeptidase/chemistry , Muramoylpentapeptide Carboxypeptidase/genetics , Mutagenesis, Insertional , Penicillin-Binding Proteins , Penicillins/pharmacology , Peptidyl Transferases/chemistry , Peptidyl Transferases/genetics , Polymerase Chain Reaction , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Streptococcus pneumoniae/drug effects
17.
Proc Natl Acad Sci U S A ; 95(23): 13403-6, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9811812

ABSTRACT

Streptococcus pneumoniae is the main causal agent of pathologies that are increasingly resistant to antibiotic treatment. Clinical resistance of S. pneumoniae to beta-lactam antibiotics is linked to multiple mutations of high molecular mass penicillin-binding proteins (H-PBPs), essential enzymes involved in the final steps of bacterial cell wall synthesis. H-PBPs from resistant bacteria have a reduced affinity for beta-lactam and a decreased hydrolytic activity on substrate analogues. In S. pneumoniae, the gene coding for one of these H-PBPs, PBP2x, is located in the cell division cluster (DCW). We present here structural evidence linking multiple beta-lactam resistance to amino acid substitutions in PBP2x within a buried cavity near the catalytic site that contains a structural water molecule. Site-directed mutation of amino acids in contact with this water molecule in the "sensitive" form of PBP2x produces mutants similar, in terms of beta-lactam affinity and substrate hydrolysis, to altered PBP2x produced in resistant clinical isolates. A reverse mutation in a PBP2x variant from a clinically important resistant clone increases the acylation efficiency for beta-lactams and substrate analogues. Furthermore, amino acid residues in contact with the structural water molecule are conserved in the equivalent H-PBPs of pathogenic Gram-positive cocci. We suggest that, probably via a local structural modification, the partial or complete loss of this water molecule reduces the acylation efficiency of PBP2x substrates to a point at which cell wall synthesis still occurs, but the sensitivity to therapeutic concentrations of beta-lactam antibiotics is lost.


Subject(s)
Bacterial Proteins , Carrier Proteins/chemistry , Carrier Proteins/genetics , Hexosyltransferases , Muramoylpentapeptide Carboxypeptidase/chemistry , Muramoylpentapeptide Carboxypeptidase/genetics , Peptidyl Transferases , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , beta-Lactam Resistance/genetics , Amino Acid Substitution , Mutagenesis, Site-Directed , Penicillin-Binding Proteins
18.
FEMS Microbiol Lett ; 164(2): 427-31, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9682492

ABSTRACT

The DNA sequences of two related plasmids pPR1 and pPR3 described previously in Streptococcus pneumoniae isolates from Germany and Spain were now determined. Both plasmids belong to a family of rolling circle (RC) plasmids found in a variety of bacteria. Their GC content with 32% is lower than that of the S. pneumoniae chromosomal DNA. The plasmid pPR3 has a molecular size of 3160 bp with four putative open reading frames, whereas pPR1 contained a deletion of 313 bp that included the 5'-part of ORF2 and upstream regions and differed by three bp from pPR3. The predicted protein of ORF1 showed high similarity to replication proteins of RC plasmids with 74% identical amino acids to RepA of Streptococcus thermophilus plasmids. Sequences similar to the plus origin of replication of ssDNA plasmids were present in both plasmids. They also contained a 152-bp region with over 83% identity to the minus origin of replication of the Streptococcus agalacticae plasmid pMV158.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Plasmids/genetics , Streptococcus pneumoniae/genetics , Trans-Activators , Base Sequence , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , Humans , Molecular Sequence Data , Open Reading Frames , Polymerase Chain Reaction , Proteins/genetics , Sequence Analysis, DNA
19.
Cell Mol Life Sci ; 54(4): 332-40, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9614969

ABSTRACT

Low-affinity penicillin-binding proteins (PBPs), which participate in the beta-lactam resistance of several pathogenic bacteria, have different origins. Natural transformation and recombination events with DNA acquired from neighbouring intrinsically resistant organisms are responsible for the appearance of mosaic genes encoding two or three low-affinity PBPs in highly resistant strains of transformable microorganisms such as Neisseria and Streptococcus pneumoniae. Methicillin-resistant Staphylococcus aureus and coagulase-negative staphylococcal strains possess the mecA determinant gene, which probably evolved within the Staphylococcus genus from a closely related and physiologically functional gene that was modified by point mutations. The expression of mecA is either inducible or constitutive. A stable high-level resistant phenotype requires the synthesis of a normally constituted peptidoglycan. Enterococci have a natural low susceptibility to beta-lactams related to the presence of an intrinsic low-affinity PBP. Highly resistant enterococcal strains overexpress this PBP and/or reduce its affinity.


Subject(s)
Bacterial Proteins , Carrier Proteins/genetics , Hexosyltransferases , Muramoylpentapeptide Carboxypeptidase/genetics , Penicillin Resistance , Penicillins/pharmacology , Peptidyl Transferases , Amino Acid Sequence , Enterococcus faecium/genetics , Molecular Sequence Data , Neisseria gonorrhoeae/genetics , Penicillin-Binding Proteins , Staphylococcus aureus/genetics , Streptococcus pneumoniae/genetics
20.
Electrophoresis ; 19(4): 597-601, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9588809

ABSTRACT

Penicillin resistance in clinical isolates of Streptococcus pneumoniae is mediated by mosaic genes encoding altered penicillin binding proteins. Mosaic sequence blocks are the result of a genetic exchange between related streptococcal species. It is likely that resistance has emerged in commensal streptococci before being transferred into the pneumococcus. Closely related mosaic genes are found in different pneumococcal clones and in different streptococcal species isolated worldwide since the first reports on such strains in the late 70s, demonstrating the importance of commensal streptococci for the spread of selectable markers in naturally transforming pathogens.


Subject(s)
Bacterial Proteins , Genes, Bacterial , Hexosyltransferases , Penicillin Resistance/genetics , Peptidyl Transferases , Streptococcus pneumoniae/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mosaicism , Muramoylpentapeptide Carboxypeptidase/genetics , Muramoylpentapeptide Carboxypeptidase/metabolism , Penicillin-Binding Proteins , Streptococcus pneumoniae/drug effects , Transformation, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL
...