Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Antimicrob Agents Chemother ; 59(5): 2807-16, 2015 May.
Article in English | MEDLINE | ID: mdl-25733511

ABSTRACT

Methylation of bacterial 16S rRNA within the ribosomal decoding center confers exceptionally high resistance to aminoglycoside antibiotics. This resistance mechanism is exploited by aminoglycoside producers for self-protection while functionally equivalent methyltransferases have been acquired by human and animal pathogenic bacteria. Here, we report structural and functional analyses of the Sorangium cellulosum So ce56 aminoglycoside resistance-conferring methyltransferase Kmr. Our results demonstrate that Kmr is a 16S rRNA methyltransferase acting at residue A1408 to confer a canonical aminoglycoside resistance spectrum in Escherichia coli. Kmr possesses a class I methyltransferase core fold but with dramatic differences in the regions which augment this structure to confer substrate specificity in functionally related enzymes. Most strikingly, the region linking core ß-strands 6 and 7, which forms part of the S-adenosyl-l-methionine (SAM) binding pocket and contributes to base flipping by the m(1)A1408 methyltransferase NpmA, is disordered in Kmr, correlating with an exceptionally weak affinity for SAM. Kmr is unexpectedly insensitive to substitutions of residues critical for activity of other 16S rRNA (A1408) methyltransferases and also to the effects of by-product inhibition by S-adenosylhomocysteine (SAH). Collectively, our results indicate that adoption of a catalytically competent Kmr conformation and binding of the obligatory cosubstrate SAM must be induced by interaction with the 30S subunit substrate.


Subject(s)
Aminoglycosides/pharmacology , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , RNA, Ribosomal, 16S/genetics , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/genetics
2.
PLoS One ; 8(1): e53217, 2013.
Article in English | MEDLINE | ID: mdl-23359797

ABSTRACT

Targeting DNA double-strand breaks is a powerful strategy for gene inactivation applications. Without the use of a repair plasmid, targeted mutagenesis can be achieved through Non-Homologous End joining (NHEJ) pathways. However, many of the DNA breaks produced by engineered nucleases may be subject to precise re-ligation without loss of genetic information and thus are likely to be unproductive. In this study, we combined engineered endonucleases and DNA-end processing enzymes to increase the efficiency of targeted mutagenesis, providing a robust and efficient method to (i) greatly improve targeted mutagenesis frequency up to 30-fold, and; (ii) control the nature of mutagenic events using meganucleases in conjunction with DNA-end processing enzymes in human primary cells.


Subject(s)
DNA End-Joining Repair , DNA/metabolism , Endonucleases/metabolism , Mutagenesis , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , DNA/genetics , DNA Primers , HEK293 Cells , Humans
3.
J Biol Chem ; 287(46): 38427-32, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23019344

ABSTRACT

Within the past 2 years, transcription activator-like effector (TALE) DNA binding domains have emerged as the new generation of engineerable platform for production of custom DNA binding domains. However, their recently described sensitivity to cytosine methylation represents a major bottleneck for genome engineering applications. Using a combination of biochemical, structural, and cellular approaches, we were able to identify the molecular basis of such sensitivity and propose a simple, drug-free, and universal method to overcome it.


Subject(s)
Cytosine/chemistry , DNA Methylation , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Base Sequence , CHO Cells , Cricetinae , DNA/genetics , Epigenesis, Genetic , Gene Silencing , Genetic Engineering/methods , Genetic Therapy/methods , HEK293 Cells , Humans , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Engineering/methods , Protein Structure, Tertiary , Recombinant Proteins/chemistry
4.
J Biol Chem ; 287(36): 30139-50, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22740697

ABSTRACT

In this study, we asked whether CpG methylation could influence the DNA binding affinity and activity of meganucleases used for genome engineering applications. A combination of biochemical and structural approaches enabled us to demonstrate that CpG methylation decreases I-CreI DNA binding affinity and inhibits its endonuclease activity in vitro. This inhibition depends on the position of the methylated cytosine within the DNA target and was almost total when it is located inside the central tetrabase. Crystal structures of I-CreI bound to methylated cognate target DNA suggested a molecular basis for such inhibition, although the precise mechanism still has to be specified. Finally, we demonstrated that the efficacy of engineered meganucleases can be diminished by CpG methylation of the targeted endogenous site, and we proposed a rational design of the meganuclease DNA binding domain to alleviate such an effect. We conclude that although activity and sequence specificity of engineered meganucleases are crucial parameters, target DNA epigenetic modifications need to be considered for successful gene editions.


Subject(s)
CpG Islands , DNA Methylation , DNA Restriction Enzymes/chemistry , DNA/chemistry , Epigenesis, Genetic , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , DNA Restriction Enzymes/genetics , DNA Restriction Enzymes/metabolism , HEK293 Cells , Humans , Protein Structure, Tertiary
5.
Protein Expr Purif ; 75(1): 89-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20667473

ABSTRACT

High-level resistance to a broad spectrum of aminoglycoside antibiotics can arise through either N7-methyl guanosine 1405 (m7G1405) or N1-methyl adenosine 1408 (m¹A1408) modifications at the drug binding site in the bacterial 30S ribosomal subunit decoding center. Two distinct families of 16S ribosomal RNA (rRNA) methyltransferases that incorporate these modifications were first identified in aminoglycoside-producing bacteria but were more recently identified in both human and animal pathogens. These resistance determinants thus pose a new threat to the usefulness of aminoglycosides as antibiotics, demanding urgent characterization of their structures and activities. Here, we describe approaches to cloning, heterologous expression in Escherichia coli, and purification of two A1408 rRNA methyltransferases: KamB from the aminoglycoside-producer Streptoalloteichus tenebrarius and NpmA identified in a clinical isolate of pathogenic E. coli ARS3. Antibiotic minimum inhibitory concentration (MIC) assays and in vitro analysis of KamB and NpmA using circular dichroism (CD) spectroscopy, S-adenosyl-l-methionine (SAM) binding by isothermal titration calorimetry and 30S subunit methylation assays showed both enzymes were soluble, folded and active. Finally, crystals of each enzyme complexed with SAM were obtained, including selenomethionine-derived KamB, that will facilitate high-resolution X-ray crystallographic analyses of these important bacterial antibiotic-resistance determinants.


Subject(s)
Bacterial Proteins/genetics , Cloning, Molecular/methods , Drug Resistance, Microbial/genetics , Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Methyltransferases/genetics , Streptomyces/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Crystallography, X-Ray , Drug Resistance, Bacterial , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Methyltransferases/chemistry , Methyltransferases/isolation & purification , Methyltransferases/metabolism , RNA, Ribosomal, 16S/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solubility , Streptomyces/chemistry , Streptomyces/genetics
6.
Nucleic Acids Res ; 38(21): 7791-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20639535

ABSTRACT

X-ray crystal structures were determined of the broad-spectrum aminoglycoside-resistance A1408 16S rRNA methyltransferases KamB and NpmA, from the aminoglycoside-producer Streptoalloteichus tenebrarius and human pathogenic Escherichia coli, respectively. Consistent with their common function, both are Class I methyltransferases with additional highly conserved structural motifs that embellish the core SAM-binding fold. In overall structure, the A1408 rRNA methyltransferase were found to be most similar to a second family of Class I methyltransferases of distinct substrate specificity (m(7)G46 tRNA). Critical residues for A1408 rRNA methyltransferase activity were experimentally defined using protein mutagenesis and bacterial growth assays with kanamycin. Essential residues for SAM coenzyme binding and an extended protein surface that likely interacts with the 30S ribosomal subunit were thus revealed. The structures also suggest potential mechanisms of A1408 target nucleotide selection and positioning. We propose that a dynamic extended loop structure that is positioned adjacent to both the bound SAM and a functionally critical structural motif may mediate concerted conformational changes in rRNA and protein that underpin the specificity of target selection and activation of methyltransferase activity. These new structures provide important new insights that may provide a starting point for strategies to inhibit these emerging causes of pathogenic bacterial resistance to aminoglycosides.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli Proteins/chemistry , Methyltransferases/chemistry , Actinomycetales/enzymology , Bacterial Proteins/metabolism , Crystallography, X-Ray , Drug Resistance, Microbial , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Methyltransferases/metabolism , Models, Molecular , Protein Binding , S-Adenosylhomocysteine/chemistry , S-Adenosylmethionine/chemistry , Sequence Analysis, Protein , Substrate Specificity , tRNA Methyltransferases/chemistry
7.
J Bacteriol ; 190(17): 5855-61, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18586937

ABSTRACT

The 16S rRNA methyltransferase Sgm from "Micromonospora zionensis" confers resistance to aminoglycoside antibiotics by specific modification of the 30S ribosomal A site. Sgm is a member of the FmrO family, distant relatives of the S-adenosyl-L-methionine (SAM)-dependent RNA subfamily of methyltransferase enzymes. Using amino acid conservation across the FmrO family, seven putative key amino acids were selected for mutation to assess their role in forming the SAM cofactor binding pocket or in methyl group transfer. Each mutated residue was found to be essential for Sgm function, as no modified protein could effectively support bacterial growth in liquid media containing gentamicin or methylate 30S subunits in vitro. Using isothermal titration calorimetry, Sgm was found to bind SAM with a K(D) (binding constant) of 17.6 microM, and comparable values were obtained for one functional mutant (N179A) and four proteins modified at amino acids predicted to be involved in catalysis in methyl group transfer. In contrast, none of the G135, D156, or D182 Sgm mutants bound the cofactor, confirming their role in creating the SAM binding pocket. These results represent the first functional characterization of any FmrO methyltransferase and may provide a basis for a further structure-function analysis of these aminoglycoside resistance determinants.


Subject(s)
Aminoglycosides/pharmacology , Bacterial Proteins/metabolism , Methyltransferases/metabolism , Micromonospora/drug effects , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Calorimetry , Catalysis , Circular Dichroism , Drug Resistance, Bacterial , Methylation , Methyltransferases/chemistry , Methyltransferases/genetics , Microbial Sensitivity Tests , Micromonospora/genetics , Micromonospora/metabolism , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Ribosome Subunits, Small, Bacterial/genetics , Ribosome Subunits, Small, Bacterial/metabolism , Sequence Homology, Amino Acid , Structural Homology, Protein
8.
Nucleic Acids Res ; 34(19): 5577-84, 2006.
Article in English | MEDLINE | ID: mdl-17028102

ABSTRACT

We have determined the structure of a catalytically inactive D70N variant of the Escherichia coli RusA resolvase bound to a duplex DNA substrate that reveals critical protein-DNA interactions and permits a much clearer understanding of the interaction of the enzyme with a Holliday junction (HJ). The RusA enzyme cleaves HJs, the fourway DNA branchpoints formed by homologous recombination, by introducing symmetrical cuts in the phosphodiester backbone in a Mg2+ dependent reaction. Although, RusA shows a high level of selectivity for DNA junctions, preferring to bind fourway junctions over other substrates in vitro, it has also been shown to have appreciable affinity for duplex DNA. However, RusA does not show DNA cleavage activity with duplex substrates. Our structure suggests the possible basis for structural selectivity as well as sources of the sequence specificity observed for DNA cleavage by RusA.


Subject(s)
DNA, Cruciform/chemistry , Escherichia coli Proteins/chemistry , Holliday Junction Resolvases/chemistry , Models, Molecular , Amino Acid Substitution , Binding Sites , DNA/chemistry , DNA, Cruciform/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Holliday Junction Resolvases/genetics , Holliday Junction Resolvases/metabolism , Substrate Specificity
9.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 8): 1067-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16041071

ABSTRACT

Equine P2 protein has been isolated from horse spinal cord and its structure determined to 2.1 A. Since equine myelin is a viable alternative to bovine tissue for large-scale preparations, characterization of the proteins from equine spinal cord myelin has been initiated. There is an unusually high amount of P2 protein in equine CNS myelin compared with other species. The structure was determined by molecular replacement and subsequently refined to an R value of 0.187 (Rfree=0.233). The structure contains a molecule of the detergent LDAO and HEPES buffer in the binding cavity and is otherwise analogous to other cellular retinol-binding proteins.


Subject(s)
Myelin P2 Protein/chemistry , Spinal Cord/chemistry , Amino Acid Sequence , Animals , Binding Sites , Computer Simulation , Crystallography, X-Ray/methods , Detergents/chemistry , Dimethylamines/chemistry , HEPES/chemistry , Horses , Ligands , Retinol-Binding Proteins/chemistry , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...