Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Article in English | MEDLINE | ID: mdl-30150481

ABSTRACT

Alpha toxin (AT) is a cytolytic pore-forming toxin that plays a key role in Staphylococcus aureus pathogenesis; consequently, extensive research was undertaken to understand the AT mechanism of action and its utility as a target for novel prophylaxis and treatment strategies against S. aureus infections. MEDI4893 (suvratoxumab) is a human anti-AT IgG1 monoclonal antibody (MAb) that targets AT and is currently in phase 2 clinical development. As shown previously, the MEDI4893-binding epitope on AT is comprised of the highly conserved amino acid regions 177 to 200 and 261 to 271, suggesting these amino acids are important for AT function. To test this hypothesis and gain insight into the effect of mutations in the epitope on AT neutralization by MEDI4893, nine MEDI4893 contact residues in AT were individually mutated to alanine. Consistent with our hypothesis, 8 out of 9 mutants exhibited >2-fold loss in lytic activity resulting from a defect in cell binding and pore formation. MEDI4893 binding affinity was reduced >2-fold (2- to 27-fold) for 7 out of 9 mutants, and no binding was detected for the W187A mutant. MEDI4893 effectively neutralized all of the lytic mutants in vitro and in vivo When the defective mutants were introduced into an S. aureus clinical isolate, the mutant-expressing strains exhibited less severe disease in mouse models and were effectively neutralized by MEDI4893. These results indicate the MEDI4893 epitope is highly conserved due in part to its role in AT pore formation and bacterial fitness, thereby decreasing the likelihood for the emergence of MAb-resistant variants.


Subject(s)
Alanine/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/genetics , Mutagenesis/genetics , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , A549 Cells , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal, Humanized , Broadly Neutralizing Antibodies , Epitopes/genetics , Epitopes/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology
2.
J Infect Dis ; 218(12): 1983-1994, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30016475

ABSTRACT

Background: Bispecific antibody MEDI3902, targeting the Pseudomonas aeruginosa type 3 secretion system (PcrV) and Psl exopolysaccharide, is currently in phase 2b development for prevention of nosocomial pneumonia in patients undergoing mechanical ventilation. We surveyed a diverse collection of isolates to study MEDI3902 epitope conservation and protective activity. Methods: P. aeruginosa clinical isolates (n = 913) were collected from diverse patients and geographic locations during 2003-2014. We conducted whole-genome sequencing; performed PcrV and Psl expression analyses via immunoblotting and enzyme-linked immunosorbent assay, respectively; performed crystallography to determine the MEDI3902 PcrV epitope, using anti-PcrV Fab and PcrV components (resolved at 2.8 Å); and evaluated MEDI3902 protective activity against select isolates in vitro and in vivo. Results: Intact psl operon and pcrV genes were present in 94% and 99% of isolates, respectively, and 99.9% of isolates contained at least one of the genetic elements. Anti-Psl binding was confirmed in tested isolates harboring a complete Psl operon or lacking nonessential psl genes. We identified 46 PcrV variant sequences, and MEDI3902-PcrV contact residues were preserved. MEDI3902 maintained potent in vivo activity against various strains, including strains expressing only a single target. Conclusions: Psl and PcrV are highly prevalent in global clinical isolates, suggesting MEDI3902 can mediate broad coverage against P. aeruginosa.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Conserved Sequence , Pseudomonas aeruginosa/metabolism , Antibodies, Bispecific , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Epitopes , Humans , Models, Molecular , Operon , Protein Conformation , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Whole Genome Sequencing
3.
Article in English | MEDLINE | ID: mdl-28584141

ABSTRACT

Secreted alpha-toxin and surface-localized clumping factor A (ClfA) are key virulence determinants in Staphylococcus aureus bloodstream infections. We previously demonstrated that prophylaxis with a multimechanistic monoclonal antibody (MAb) combination against alpha-toxin (MEDI4893*) and ClfA (11H10) provided greater strain coverage and improved efficacy in an S. aureus lethal bacteremia model. Subsequently, 11H10 was found to exhibit reduced affinity and impaired inhibition of fibrinogen binding to ClfA002 expressed by members of a predominant hospital-associated methicillin-resistant S. aureus (MRSA) clone, ST5. Consequently, we identified another anti-ClfA MAb (SAR114) from human tonsillar B cells with >100-fold increased affinity for three prominent ClfA variants, including ClfA002, and potent inhibition of bacterial agglutination by 112 diverse clinical isolates. We next constructed bispecific Abs (BiSAbs) comprised of 11H10 or SAR114 as IgG scaffolds and grafted anti-alpha-toxin (MEDI4893*) single-chain variable fragment to the amino or carboxy terminus of the anti-ClfA heavy chains. Although the BiSAbs exhibited in vitro potencies similar to those of the parental MAbs, only 11H10-BiSAb, but not SAR114-BiSAb, showed protective activity in murine infection models comparable to the respective MAb combination. In vivo activity with SAR114-BiSAb was observed in infection models with S. aureus lacking ClfA. Our data suggest that high-affinity binding to ClfA sequesters the SAR114-BiSAb to the bacterial surface, thereby reducing both alpha-toxin neutralization and protection in vivo These results indicate that a MAb combination targeting ClfA and alpha-toxin is more promising for future development than the corresponding BiSAb.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Bacteremia/drug therapy , Bacterial Toxins/immunology , Coagulase/immunology , Hemolysin Proteins/immunology , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Bacteremia/microbiology , Broadly Neutralizing Antibodies , Female , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Staphylococcal Infections/immunology , Virulence Factors
4.
mBio ; 7(3)2016 06 28.
Article in English | MEDLINE | ID: mdl-27353753

ABSTRACT

UNLABELLED: Staphylococcus aureus produces numerous virulence factors, each contributing different mechanisms to bacterial pathogenesis in a spectrum of diseases. Alpha toxin (AT), a cytolytic pore-forming toxin, plays a key role in skin and soft tissue infections and pneumonia, and a human anti-AT monoclonal antibody (MAb), MEDI4893*, has been shown to reduce disease severity in dermonecrosis and pneumonia infection models. However, interstrain diversity and the complex pathogenesis of S. aureus bloodstream infections suggests that MEDI4893* alone may not provide adequate protection against S. aureus sepsis. Clumping factor A (ClfA), a fibrinogen binding protein, is an important virulence factor facilitating S. aureus bloodstream infections. Herein, we report on the identification of a high-affinity anti-ClfA MAb, 11H10, that inhibits ClfA binding to fibrinogen, prevents bacterial agglutination in human plasma, and promotes opsonophagocytic bacterial killing (OPK). 11H10 prophylaxis reduced disease severity in a mouse bacteremia model and was dependent on Fc effector function and OPK. Additionally, prophylaxis with 11H10 in combination with MEDI4893* provided enhanced strain coverage in this model and increased survival compared to that obtained with the individual MAbs. The MAb combination also reduced disease severity in murine dermonecrosis and pneumonia models, with activity similar to that of MEDI4893* alone. These results indicate that an MAb combination targeting multiple virulence factors provides benefit over a single MAb neutralizing one virulence mechanism by providing improved efficacy, broader strain coverage, and protection against multiple infection pathologies. IMPORTANCE: Alternative strategies to broad-spectrum antibiotics are required to combat the antibiotic resistance epidemic. Previous attempts at active or passive immunization against Staphylococcus aureus targeting single antigens have failed in clinical trials despite positive preclinical data. To provide broad disease and isolate coverage, an effective immunization strategy likely must target multiple virulence mechanisms of the pathogen. Herein, we tested a multimechanistic MAb combination targeting alpha toxin (AT) and clumping factor A (ClfA) that neutralizes AT-mediated cytotoxicity, blocks fibrinogen binding by ClfA, prevents bacterial agglutination, targets the bacteria for opsonophagocytic killing, and provides broad isolate coverage in a lethal-bacteremia model. Although each MAb alone was effective in bacteremia against some individual isolates, the MAb combination provided improved protection against other isolates. These results illustrate the importance of targeting multiple virulence mechanisms and highlight the potential for an MAb combination targeting AT and ClfA to effectively prevent S. aureus disease.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Bacterial Toxins/immunology , Coagulase/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Virulence Factors/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/therapeutic use , Bacterial Load , Disease Models, Animal , HL-60 Cells , Humans , Immunization, Passive/methods , Mice , Phagocytosis , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology
5.
Antimicrob Agents Chemother ; 59(8): 4526-32, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25987629

ABSTRACT

Immunocompromised individuals are at increased risk of Staphylococcus aureus pneumonia. Neutralization of alpha-toxin (AT) with the monoclonal antibody (MAb) MEDI4893* protects normal mice from S. aureus pneumonia; however, the effects of the MAb in immunocompromised mice have not been reported. In this study, passive immunization with MEDI4893* increased survival rates and reduced bacterial numbers in the lungs in an immunocompromised murine S. aureus pneumonia model. Lungs from infected mice exhibited alveolar epithelial damage, protein leakage, and bacterial overgrowth, whereas lungs from mice passively immunized with MEDI4893* retained a healthy architecture, with an intact epithelial barrier. Adjunctive therapy or prophylaxis with a subtherapeutic MEDI4893* dose combined with subtherapeutic doses of vancomycin or linezolid improved survival rates, compared with the monotherapies. Furthermore, coadministration of MEDI4893* with vancomycin or linezolid extended the antibiotic treatment window. These data suggest that MAb-mediated neutralization of AT holds promise in strategies for prevention and adjunctive therapy among immunocompromised patients.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Immunocompromised Host/drug effects , Pneumonia, Staphylococcal/drug therapy , Staphylococcus aureus/drug effects , Animals , Antibodies, Monoclonal, Humanized , Broadly Neutralizing Antibodies , Female , Linezolid/pharmacology , Lung/microbiology , Mice , Mice, Inbred C57BL , Survival Rate , Vancomycin/pharmacology
6.
Antimicrob Agents Chemother ; 58(2): 1108-17, 2014.
Article in English | MEDLINE | ID: mdl-24295977

ABSTRACT

Alpha-toxin (AT) is a major virulence factor in the disease pathogenesis of Staphylococcus aureus. We previously identified a monoclonal antibody (MAb) against AT that reduced disease severity in a mouse dermonecrosis model. Here, we evaluate the activity of an affinity-optimized variant, LC10, in a mouse model of S. aureus pneumonia. Passive immunization with LC10 increased survival and reduced bacterial numbers in the lungs and kidneys of infected mice and showed protection against diverse S. aureus clinical isolates. The lungs of S. aureus-infected mice exhibited bacterial pneumonia, including widespread inflammation, whereas the lungs of mice that received LC10 exhibited minimal inflammation and retained healthy architecture. Consistent with reduced immune cell infiltration, LC10-treated animals had significantly lower (P < 0.05) proinflammatory cytokine and chemokine levels in the bronchoalveolar lavage fluid than did those of the control animals. This reduction in inflammation and damage to the LC10-treated animals resulted in reduced vascular protein leakage and CO2 levels in the blood. LC10 was also assessed for its therapeutic activity in combination with vancomycin or linezolid. Treatment with a combination of LC10 and vancomycin or linezolid resulted in a significant increase (P < 0.05) in survival relative to the monotherapies and was deemed additive to synergistic by isobologram analysis. Consistent with improved survival, the lungs of animals treated with antibiotic plus LC10 exhibited less inflammatory tissue damage than those that received monotherapy. These data provide insight into the mechanisms of protection provided by AT inhibition and support AT as a promising target for immunoprophylaxis or adjunctive therapy against S. aureus pneumonia.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Bacterial Toxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Lung/drug effects , Pneumonia, Staphylococcal/drug therapy , Acetamides/pharmacology , Animals , Bacterial Toxins/immunology , Bronchoalveolar Lavage Fluid/chemistry , Chemokines/antagonists & inhibitors , Chemokines/biosynthesis , Drug Synergism , Drug Therapy, Combination , Female , Hemolysin Proteins/immunology , Immunization, Passive , Kidney/drug effects , Kidney/immunology , Kidney/microbiology , Linezolid , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Oxazolidinones/pharmacology , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/mortality , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Survival Analysis , Vancomycin/pharmacology
7.
Clin Vaccine Immunol ; 19(3): 377-85, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22237895

ABSTRACT

Staphylococcus aureus alpha toxin (AT) is an important virulence determinant and may be a valid target for immunoprophylaxis against staphylococcal disease. Here we report the identification of potent inhibitory anti-AT monoclonal antibodies (MAbs) derived using B-cell hybridoma technology from VelocImmune mice engineered to produce IgG with a human variable domain. A small panel of inhibitory MAbs blocked AT-mediated lysis of rabbit red blood cells, A549 human lung epithelial cells, and THP-1 human monocytic cells, in a dose-dependent manner. Binding studies indicated that these MAbs recognize a similar epitope on AT and exhibit dissociation constants (K(D)) ranging from 0.50 to 15 nM. In an S. aureus dermonecrosis model, mice passively immunized with anti-AT inhibitory MAbs exhibited significant reductions of lesion size relative to mice treated with an irrelevant IgG control. Interestingly, there was a correlation between MAb affinity for a single epitope, the 50% inhibitory concentration (IC(50)) in the AT hemolytic assay, and lesion size reduction in the dermonecrosis model. A representative high-affinity MAb, 2A3.1, was demonstrated to significantly reduce lesion size following infection with three different clinical isolates (USA300, CC30, and CC5). Taken together, these results indicate that in vitro potency of anti-AT MAbs predicts in vivo potency in this model, supporting their continued preclinical evaluation as molecules for immunoprophylaxis against staphylococcal skin and soft tissue infections caused by diverse clinical isolates.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antitoxins/administration & dosage , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/toxicity , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/toxicity , Necrosis/prevention & control , Staphylococcal Skin Infections/prevention & control , Animals , Antibodies, Monoclonal/pharmacology , Antitoxins/pharmacology , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Epithelial Cells/drug effects , Erythrocytes/drug effects , Female , Humans , Immunization, Passive , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Necrosis/pathology , Rabbits , Staphylococcal Skin Infections/pathology
8.
Antimicrob Agents Chemother ; 53(4): 1314-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19075058

ABSTRACT

Oxazolidinone antibiotics have activity against Mycobacterium tuberculosis. Linezolid, the only marketed oxazolidinone, has been used off-label in combination regimens to treat multidrug-resistant tuberculosis, but its precise contribution to the efficacy of such combinations is unclear. Another oxazolidinone, PNU-100480, has been demonstrated to have more potent activity in vitro and in a murine model of tuberculosis. In this study, we compared the pharmacokinetics and the antituberculosis activities of these two oxazolidinones over a range of doses and found that linezolid has limited activity at clinically relevant doses in the murine model compared to that of PNU-100480, which has potent bactericidal activity, even at lower drug exposures. These findings were unexpected, given the similar in vitro activities of PNU-100480, its major metabolites, and linezolid. Moreover, the incorporation of PNU-100480 dramatically improved the bactericidal activities of regimens containing current first-line antituberculosis drugs and moxifloxacin. For example, the addition of PNU-100480 (100 mg/kg of body weight/day) to the standard daily regimen of rifampin (rifampicin), isoniazid, and pyrazinamide resulted in an additional 2.0-log(10)-unit reduction in lung CFU counts during the first 2 months of treatment. The combination of PNU-100480, moxifloxacin, and pyrazinamide, which does not contain either rifampin or isoniazid, was also more active than rifampin, isoniazid, and pyrazinamide. These results suggest that PNU-100480 may have the potential to significantly shorten the duration of therapy for drug-susceptible as well as multidrug-resistant tuberculosis.


Subject(s)
Acetamides/therapeutic use , Antitubercular Agents/therapeutic use , Oxazolidinones/therapeutic use , Tuberculosis/drug therapy , Acetamides/pharmacokinetics , Acetamides/pharmacology , Animals , Antitubercular Agents/pharmacokinetics , Antitubercular Agents/pharmacology , Colony Count, Microbial , Disease Models, Animal , Drug Therapy, Combination , Female , Linezolid , Lung/microbiology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Oxazolidinones/pharmacokinetics , Oxazolidinones/pharmacology , Tuberculosis/microbiology
10.
Nature ; 406(6799): 959-64, 2000 Aug 31.
Article in English | MEDLINE | ID: mdl-10984043

ABSTRACT

Pseudomonas aeruginosa is a ubiquitous environmental bacterium that is one of the top three causes of opportunistic human infections. A major factor in its prominence as a pathogen is its intrinsic resistance to antibiotics and disinfectants. Here we report the complete sequence of P. aeruginosa strain PAO1. At 6.3 million base pairs, this is the largest bacterial genome sequenced, and the sequence provides insights into the basis of the versatility and intrinsic drug resistance of P. aeruginosa. Consistent with its larger genome size and environmental adaptability, P. aeruginosa contains the highest proportion of regulatory genes observed for a bacterial genome and a large number of genes involved in the catabolism, transport and efflux of organic compounds as well as four potential chemotaxis systems. We propose that the size and complexity of the P. aeruginosa genome reflect an evolutionary adaptation permitting it to thrive in diverse environments and resist the effects of a variety of antimicrobial substances.


Subject(s)
Genome, Bacterial , Pseudomonas aeruginosa/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Computational Biology , DNA, Bacterial , Drug Resistance, Microbial , Gene Expression Regulation, Bacterial , Humans , Molecular Sequence Data , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Sequence Analysis, DNA , Species Specificity
11.
Nature ; 405(6789): 962-6, 2000 Jun 22.
Article in English | MEDLINE | ID: mdl-10879539

ABSTRACT

Mycobacterium tuberculosis, which causes tuberculosis, is the greatest single infectious cause of mortality worldwide, killing roughly two million people annually. Estimates indicate that one-third of the world population is infected with latent M. tuberculosis. The synergy between tuberculosis and the AIDS epidemic, and the surge of multidrug-resistant clinical isolates of M. tuberculosis have reaffirmed tuberculosis as a primary public health threat. However, new antitubercular drugs with new mechanisms of action have not been developed in over thirty years. Here we report a series of compounds containing a nitroimidazopyran nucleus that possess antitubercular activity. After activation by a mechanism dependent on M. tuberculosis F420 cofactor, nitroimidazopyrans inhibited the synthesis of protein and cell wall lipid. In contrast to current antitubercular drugs, nitroimidazopyrans exhibited bactericidal activity against both replicating and static M. tuberculosis. Lead compound PA-824 showed potent bactericidal activity against multidrugresistant M. tuberculosis and promising oral activity in animal infection models. We conclude that nitroimidazopyrans offer the practical qualities of a small molecule with the potential for the treatment of tuberculosis.


Subject(s)
Antitubercular Agents/therapeutic use , Nitroimidazoles/therapeutic use , Tuberculosis/drug therapy , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/metabolism , Bacterial Proteins/biosynthesis , Drug Resistance, Microbial/genetics , Drug Resistance, Multiple , Guinea Pigs , Lipids/biosynthesis , Metronidazole/chemistry , Metronidazole/therapeutic use , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Nitroimidazoles/chemistry , Nitroimidazoles/metabolism , Nitroimidazoles/pharmacology , Oxazoles/chemistry , Oxazoles/therapeutic use , Structure-Activity Relationship
12.
Biofactors ; 10(2-3): 211-7, 1999.
Article in English | MEDLINE | ID: mdl-10609885

ABSTRACT

The Mycobacterium tuberculosis AhpC is similar to a family of bacterial and eukaryotic antioxidant proteins with alkylhydroperoxidase (Ahp) and thioredoxin-dependent peroxidase (TPx) activities. AhpC expression is associated with resistance to the front-line antitubercular drug isoniazid in the naturally resistant organisms E. coli and M. smegmatis. We identified several isoniazid-resistant M. tuberculosis isolates with ahpC promoter mutations resulting in AhpC overexpression. These strains were more resistant to cumene hydroperoxide than were wild-type strains. However, these strains were unchanged in their sensitivity to isoniazid, refuting a role for AhpC in detoxification of this drug. All the isoniazid-resistant, AhpC-overexpressing strains were also deficient in activity of the mycobacterial catalase-peroxidase KatG. KatG, the only known catalase in M. tuberculosis, is required for activation of isoniazid. We propose that compensatory ahpC promoter mutations are selected from KatG-deficient, isoniazid-resistant M. tuberculosis during infections, to mitigate the added burden imposed by organic peroxides on these strains.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Oxidative Stress , Peroxidases/genetics , Peroxidases/metabolism , Amino Acid Sequence , Antioxidants/metabolism , Drug Resistance, Microbial , Isoniazid/pharmacology , Molecular Sequence Data , Mycobacterium tuberculosis/genetics , Peroxidases/chemistry , Peroxiredoxins , Promoter Regions, Genetic , Sequence Alignment , Sequence Homology, Amino Acid
13.
Antimicrob Agents Chemother ; 43(12): 2975-83, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10582892

ABSTRACT

Pseudomonas aeruginosa can employ many distinct mechanisms of resistance to aminoglycoside antibiotics; however, in cystic fibrosis patients, more than 90% of aminoglycoside-resistant P. aeruginosa isolates are of the impermeability phenotype. The precise molecular mechanisms that produce aminoglycoside impermeability-type resistance are yet to be elucidated. A subtractive hybridization technique was used to reveal gene expression differences between PAO1 and isogenic, spontaneous aminoglycoside-resistant mutants of the impermeability phenotype. Among the many genes found to be up-regulated in these laboratory mutants were the amrAB genes encoding a recently discovered efflux system. The amrAB genes appear to be the same as the recently described mexXY genes; however, the resistance profile that we see in P. aeruginosa is very different from that described for Escherichia coli with mexXY. Direct evidence for AmrAB involvement in aminoglycoside resistance was provided by the deletion of amrB in the PAO1-derived laboratory mutant, which resulted in the restoration of aminoglycoside sensitivity to a level nearly identical to that of the parent strain. Furthermore, transcription of the amrAB genes was shown to be up-regulated in P. aeruginosa clinical isolates displaying the impermeability phenotype compared to a genotypically matched sensitive clinical isolate from the same patient. This suggests the possibility that AmrAB-mediated efflux is a clinically relevant mechanism of aminoglycoside resistance. Although it is unlikely that hyperexpression of AmrAB is the sole mechanism conferring the impermeability phenotype, we believe that the Amr efflux system can contribute to a complex interaction of molecular events resulting in the aminoglycoside impermeability-type resistance phenotype.


Subject(s)
Anti-Bacterial Agents/metabolism , Pseudomonas aeruginosa/metabolism , Anti-Bacterial Agents/pharmacology , Blotting, Southern , Chromosome Mapping , Culture Media , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Drug Resistance, Microbial , Electrophoresis, Polyacrylamide Gel , Humans , Microbial Sensitivity Tests , Mutation/genetics , Permeability , Phenotype , Plasmids/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tobramycin/pharmacology , Transcriptional Activation/physiology , Up-Regulation/drug effects , Up-Regulation/genetics
14.
Vaccine ; 17(7-8): 904-14, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10067697

ABSTRACT

This phase I clinical trial was designed to determine the feasibility of using rBCG as a live bacterial vaccine vector for the outer surface protein A (OspA) of Borrelia burgdorferi and as model for other vaccines based on a rBCG vector. To construct the vaccine, a signal peptide derived from a mycobacterial lipoprotein was used to direct the export, and membrane-associated surface expression, of OspA in a standard strain of BCG (Connaught). The rBCG OspA vaccine was safe and immunogenic in several animal species, and protective in a mouse model of Lyme borreliosis. An intradermal injection (0.1 ml) of rBCG OspA was administered to 24 healthy adult volunteers sequentially at one of four dose levels, ranging from 2.0 x 10(4) CFU to 2 x 10(7) CFU, using a dose-escalation design. All volunteers were initially PPD-skin test and OspA antibody negative, and they were monitored for 2 years after immunization. Three volunteers had mild flu-like reactions 1-2 days after vaccination. Local ulceration and drainage at the site of injection, which occurred in 50% and 83% of volunteers in the two highest dose groups, persisted for 1-70 days before the ulcers healed. Most of the drainage samples yielded rBCG colonies that contained the OspA plasmid. Thirteen of 24 vaccinees, principally in the two highest dose groups, converted their PPD skin tests from negative to positive. None of the 24 volunteers developed OspA antibody. In conclusion, the current rBCG vaccine construct, the first such construct tested in humans, had a safety profile comparable to that of licensed BCG, but it did not elicit primary humoral responses to the vectored antigen.


Subject(s)
Antigens, Surface/adverse effects , Antigens, Surface/immunology , BCG Vaccine/adverse effects , BCG Vaccine/immunology , Bacterial Outer Membrane Proteins/adverse effects , Bacterial Outer Membrane Proteins/immunology , Borrelia burgdorferi Group/immunology , Lipoproteins , Lyme Disease/prevention & control , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Adolescent , Adult , Animals , Antigens, Surface/genetics , BCG Vaccine/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines , Borrelia burgdorferi Group/growth & development , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Tuberculin Test , Vaccines, Synthetic/genetics
15.
Infect Immun ; 67(2): 740-4, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9916085

ABSTRACT

Staphylococcus aureus causes a wide variety of invasive human infections. However, delineation of the genes which are essential for the in vivo survival of this pathogen has not been accomplished to date. Using signature tag mutagenesis techniques and large mutant pool screens, previous investigators identified several major gene classes as candidate essential gene loci for in vivo survival; these include genes for amino acid transporters, oligopeptide transporters, and lantibiotic synthesis (W. R. Schwan, S. N. Coulter, E. Y. W. Ng, M. H. Langhorne, H. D. Ritchie, L. L. Brody, S. Westbrock-Wadman, A. S. Bayer, K. R. Folger, and C. K. Stover, Infect. Immun. 66:567-572, 1998). In this study, we directly compared the virulence of four such isogenic signature tag mutants with that of the parental strain (RN6390) by using a prototypical model of invasive S. aureus infection, experimental endocarditis (IE). The oligonucleotide signature tag (OST) mutant with insertional inactivation of the gene (putP) which encodes the high-affinity transporter for proline uptake exhibited significantly reduced virulence in the IE model across three challenge inocula (10(4) to 10(6) CFU) in terms of achievable intravegetation densities (P, <0.05). The negative impact of putP inactivation on in vivo survival in the IE model was confirmed by simultaneous challenge with the original putP mutant and the parental strain as well as by challenge with a putP mutant in which this genetic inactivation was transduced into a distinct parental strain (S6C). In contrast, inactivation of loci encoding an oligopeptide transporter, a purine repressor, and lantibiotic biosynthesis had no substantial impact on the capacity of OST mutants to survive within IE vegetations. Thus, genes encoding the uptake of essential amino acids may well represent novel targets for new drug development. These data also confirm the utility of the OST technique as an important screening methodology for identifying candidate genes as requisite loci for the in vivo survival of S. aureus.


Subject(s)
Amino Acid Transport Systems, Neutral , Endocarditis, Bacterial/microbiology , Membrane Transport Proteins/genetics , Staphylococcus aureus/enzymology , Animals , Disease Models, Animal , Genes, Bacterial , Membrane Transport Proteins/physiology , Rabbits , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence
16.
J Mol Microbiol Biotechnol ; 1(2): 289-93, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10943558

ABSTRACT

We herein describe all genes encoding constituents of the phosphoenolpyruvate:sugar phosphotransferase system (PTS) in the 6Mbp genome of the opportunistic human pathogen, Pseudomonas aeruginosa. Only four gene clusters were found to encode identifiable PTS homologues. These genes clusters encode novel multidomain proteins, two complete sugar-specific PTS phosphoryl transfer chains for the metabolism of fructose and N-acetylglucosamine, and a complex regulatory system that may function to coordinate carbon and nitrogen metabolism. No previously characterized organism has been shown to exhibit such a novel and restricted complement of PTS proteins.


Subject(s)
Phosphoenolpyruvate Sugar Phosphotransferase System/genetics , Pseudomonas aeruginosa/enzymology , Genome, Bacterial , Humans , Multigene Family , Operon , Phosphotransferases (Nitrogenous Group Acceptor)/genetics , Pseudomonas aeruginosa/genetics
17.
Mol Microbiol ; 30(2): 393-404, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9791183

ABSTRACT

The Gram-positive bacterium Staphylococcus aureus infects diverse tissues and causes a wide spectrum of diseases, suggesting that it possesses a repertoire of distinct molecular mechanisms promoting bacterial survival in disparate in vivo environments. Signature-tag transposon mutagenesis screening of a 1520-member library identified numerous S. aureus genetic loci affecting growth and survival in four complementary animal infection models including mouse abscess, bacteraemia and wound and rabbit endocarditis. Of a total of 237 in vivo attenuated mutants identified by the murine models, less than 10% showed attenuation in all three models, emphasizing the advantage of screening in diverse disease environments. The largest gene class identified by these analyses encoded peptide and amino acid transporters, some of which were important for S. aureus survival in all animal infection models tested. The identification of staphylococcal loci affecting growth, persistence and virulence in multiple tissue environments provides insight into the complexities of human infection and on the molecular mechanisms that could be targeted by new antibacterial therapies.


Subject(s)
Bacterial Proteins , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bacteremia/microbiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Division/genetics , Disease Models, Animal , Endocarditis/microbiology , Gene Library , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Molecular Sequence Data , Mutagenesis , Mutation , Rabbits , Staphylococcus aureus/growth & development , Virulence , Wounds and Injuries/microbiology
18.
Infect Immun ; 66(2): 567-72, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453610

ABSTRACT

Staphylococcus aureus is an important pathogen of humans and other animals, causing bacteremia, abscesses, endocarditis, and other infectious syndromes. A signature-tagged mutagenesis (STM) system was adapted for use in studying the genes required for in vivo survival of S. aureus. An STM library was ultimately created in S. aureus RN6390, with Tn917 being used to create the transposon mutations. Pools of S. aureus RN6390 mutants were screened in mouse abscess, bacteremia, and wound infection models for growth attenuation after in vivo passage. One of the mutants that was identified displayed marked attenuation following large-pool screening in all three animal models, which was confirmed in bacteremia and endocarditis models of infection with a smaller pool of mutants. Sequence analysis of the entire open reading frame showed a 99% identity to the high-affinity proline permease (putP) gene characterized in another strain of S. aureus. In wound and murine abscess infection models, the putP mutant was approximately 10-fold more attenuated than was wild-type strain RN6390. Another S. aureus strain transduced with the putP mutation also displayed an attenuated phenotype after passage in the wound model. A [3H]proline uptake assay showed that less proline was specifically transported into the putP mutant than into strain RN6390. The reduced viability of the bacteria possessing the mutation in the S. aureus high-affinity proline permease suggests that proline scavenging by the bacteria is important for in vivo growth and proliferation and that analogs of proline may serve as potential antistaphylococcal therapeutic agents.


Subject(s)
Amino Acid Transport Systems, Neutral , Membrane Transport Proteins/physiology , Staphylococcus aureus/physiology , Animals , DNA Transposable Elements , Mice , Mice, Inbred C57BL , Mutation , Proline/metabolism , Rabbits
19.
Antimicrob Agents Chemother ; 41(3): 570-4, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9055994

ABSTRACT

The object of this study was to investigate the ability of a rapid luciferase assay to detect antimycobacterial activity in plant extracts. Recombinant strains of Mycobacterium bovis BCG (rBCG) and Mycobacterium intracellulare expressing firefly luciferase were used as the test organisms. Assays were conducted in a 96-well minitube format under biosafety level 2 conditions. Control and test wells were sampled immediately after inoculation and after 3 (recombinant M. intracellulare) and 5 (rBCG) days of incubation to measure luminescence with a microplate luminometer, and the relative change in luminescence was calculated as a percentage of control values. As an alternative test method, Alamar blue was added after 12 days of incubation, and changes in color were read visually. A total of 480 extracts were tested. Sixteen extracts were active against rBCG, and of those, seven were also active against recombinant M. intracellulare. With activity defined as a relative change in luminescence of < or = 1% (i.e., > or = 99% inhibition) and a persistence of blue color after addition of Alamar blue, there was 99.0% agreement between the two methods. Our results suggest that the luciferase assay is rapid and accurate and has the potential to greatly accelerate the evaluation of antimycobacterial activity in plant extracts in vitro. With this method, it is possible to screen a large number of samples in a short period of time.


Subject(s)
Luciferases/biosynthesis , Mycobacterium avium Complex/drug effects , Mycobacterium bovis/drug effects , Plant Extracts/pharmacology , Anti-Bacterial Agents/pharmacology , Antitubercular Agents/pharmacology , Colorimetry , Culture Media , Drug Evaluation, Preclinical , Isoniazid/pharmacology , Luminescent Measurements , Mycobacterium avium Complex/enzymology , Mycobacterium bovis/enzymology
20.
J Infect Dis ; 174(5): 1085-90, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8896513

ABSTRACT

An examination of the pattern of lipid biosynthetic responses to isoniazid (INH) treatment of Mycobacterium tuberculosis and Mycobacterium smegmatis suggests that the mode of action of activated INH differs between these 2 organisms. Transformation of M. smegmatis with inhA on a plasmid construct conferred high-level resistance to INH, while the same construct failed to confer resistance upon M. tuberculosis. The inhA region from 2 clinical isolates whose resistance has been attributed to changes in the upstream promoter region has been cloned and was not sufficient to impart INH resistance to the level of the parent strain on sensitive M. tuberculosis. These putative mutant promoter elements appear to elevate expression levels of gene fusion reporter constructs, suggesting some noncausal connection between the observed mutations and the lipid metabolism of drug-resistant organisms. These results suggest that InhA is not the major target for activated INH in M. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Isoniazid/pharmacology , Mycobacterium tuberculosis/drug effects , Oxidoreductases , Biotransformation , Drug Resistance, Microbial , Isoniazid/metabolism , Lipids/biosynthesis , Mutation , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...