Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
PLoS Negl Trop Dis ; 13(8): e0007113, 2019 08.
Article in English | MEDLINE | ID: mdl-31425525

ABSTRACT

Buruli Ulcer (BU) is a cutaneous disease caused by Mycobacterium ulcerans. The pathogenesis of this disease is closely related to the secretion of the toxin mycolactone that induces extensive destruction of the skin and soft tissues. Currently, there are no effective measures to prevent the disease and, despite availability of antibiotherapy and surgical treatments, these therapeutic options are often associated with severe side effects. Therefore, it is important to develop alternative strategies for the treatment of BU. Endolysins (lysins) are phage encoded enzymes that degrade peptidoglycan of bacterial cell walls. Over the past years, lysins have been emerging as alternative antimicrobial agents against bacterial infections. However, mycobacteria have an unusual outer membrane composed of mycolylarabinogalactan-peptidoglycan. To overcome this complex barrier, some mycobacteriophages encode a lipolytic enzyme, Lysin B (LysB). In this study, we demonstrate for the first time that recombinant LysB displays lytic activity against M. ulcerans isolates. Moreover, using a mouse model of M. ulcerans footpad infection, we show that subcutaneous treatment with LysB prevented further bacterial proliferation, associated with IFN-γ and TNF production in the draining lymph node. These findings highlight the potential use of lysins as a novel therapeutic approach against this neglected tropical disease.


Subject(s)
Buruli Ulcer/drug therapy , Endopeptidases/administration & dosage , Mycobacteriophages/enzymology , Mycobacterium ulcerans/drug effects , Animals , Bacteriolysis , Buruli Ulcer/pathology , Disease Models, Animal , Endopeptidases/pharmacology , Female , Interferon-gamma/analysis , Lymph Nodes/immunology , Mice, Inbred BALB C , Mycobacterium ulcerans/virology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Treatment Outcome , Tumor Necrosis Factor-alpha/analysis
2.
J Med Microbiol ; 67(3): 296-307, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29458674

ABSTRACT

PURPOSE: P128, a phage-derived lysin, exerts antibacterial activity on staphylococci by cleaving the pentaglycine-bridge of peptidoglycan. We sought to determine whether the presence of P128 could re-sensitize drug-resistant bacteria to antibiotics by virtue of its cell wall degrading property. METHODOLOGY: P128 was tested in combination with standard-of-care (SoC) drugs by chequerboard assays on planktonic cells and biofilms of strains individually resistant to these drugs. The bactericidal effect of P128 and drug combinations on planktonic cells and biofilms was measured by c.f.u. reduction assays. A mouse model of MRSA bacteraemia was used to test the efficacy of P128 and oxacillin in combination. RESULTS: A combination of sub-MIC P128 (0.025-0.20 µg ml-1) and 0.5 µg ml-1 of oxacillin resulted in inhibition of bacterial growth in four MRSA strains. Similar results were seen with all the other drugs tested, wherein sub-MIC of P128 re-sensitized S. aureus and CoNS strains to SoC drugs. The chequerboard assays on strains of S. aureus and CoNS showed that combinations of P128 and antibiotics consistently inhibited bacterial growth on biofilms. Data from scanning electron microscopy and c.f.u. reduction assays on drug-resistant S. aureus and CoNS demonstrated that sub-MICs of P128 and SoC antibiotics could kill biofilm-embedded bacteria. In vivo, a combination of sub-therapeutic doses of P128 and oxacillin could help protect animals from fatal bacteraemia. CONCLUSION: The ability of P128 to re-sensitize bacteria to SoC drugs suggests that combinations of P128 and SoC antibiotics can potentially be developed to treat infections caused by drug-resistant strains of staphylococci.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Recombinant Fusion Proteins/pharmacology , Staphylococcus aureus/drug effects , Animals , Biofilms/drug effects , Biofilms/growth & development , Disease Models, Animal , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Microbial Sensitivity Tests , Oxacillin/pharmacology , Recombinant Fusion Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development
3.
Drug Discov Today ; 23(4): 848-856, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29326076

ABSTRACT

Bacterial biofilms are highly resistant to the action of antibiotics. Presence of persisters, phenotypically resistant populations of bacterial cells, is thought to contribute toward recalcitrance of biofilms. The phage-derived lysins, by virtue of their ability to cleave the peptidoglycan of bacterial cells in an enzymatic manner, have the unique ability to kill dormant cells. Several lysins have shown potent antibiofilm activity in vitro. The fact that lysins have shown better efficacy than conventional drugs in animal models of endocarditis and other infections involving biofilms suggests that the lysins can potentially be developed against difficult-to-treat bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteriophages/metabolism , Biofilms/drug effects , Mucoproteins/pharmacology , Animals , Bacterial Infections/drug therapy , Humans
4.
Article in English | MEDLINE | ID: mdl-28559263

ABSTRACT

Coagulase-negative staphylococci (CoNS) are the major causative agents of foreign-body-related infections, including catheter-related bloodstream infections. Because of the involvement of biofilms, foreign-body-related infections are difficult to treat. P128, a chimeric recombinant phage-derived ectolysin, has been shown to possess bactericidal activity on strains of Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA). We tested the killing potential of P128 on three clinically significant species of CoNS, S. epidermidis, S. haemolyticus, and S. lugdunensis, under a variety of physiological conditions representing growing and nongrowing states. The MIC90 and minimum bactericidal concentration at which 90% of strains tested are killed (MBC90) of P128 on 62 clinical strains of CoNS were found to be 16 and 32 µg/ml (0.58 and 1.16 µM), respectively, demonstrating the bactericidal nature of P128 on CoNS strains. Serum showed a potentiating effect on P128 inhibition, as indicated by 4- to 32-fold lower MIC values observed in serum. P128 caused a rapid loss of viability in all CoNS strains tested. Persisters of CoNS that were enriched in the presence of vancomycin or daptomycin were killed by P128 at 1× the MIC in a rapid manner. Low concentrations of P128 caused a 2- to 5-log reduction in CFU in stationary-phase or poorly metabolizing CoNS cultures. P128 at low concentrations eliminated CoNS biofilms in microtiter plates and on the surface of catheters. Combinations of P128 and standard-of-care (SoC) antibiotics were highly synergistic in inhibiting growth in preformed biofilms. Potent activity on planktonic cells, persisters, and biofilms of CoNS suggests that P128 is a promising candidate for the clinical development of treatments for foreign-body-related and other CoNS infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Recombinant Fusion Proteins/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus epidermidis/drug effects , Staphylococcus haemolyticus/drug effects , Staphylococcus lugdunensis/drug effects , Catheter-Related Infections/drug therapy , Catheter-Related Infections/microbiology , Coagulase/metabolism , Daptomycin/pharmacology , Drug Synergism , Drug Therapy, Combination , Foreign-Body Reaction/drug therapy , Foreign-Body Reaction/microbiology , Humans , Microbial Sensitivity Tests , Staphylococcus epidermidis/enzymology , Staphylococcus haemolyticus/enzymology , Staphylococcus lugdunensis/enzymology , Vancomycin/pharmacology
5.
Crit Care ; 21(1): 99, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28468638
6.
Antimicrob Agents Chemother ; 60(12): 7280-7289, 2016 12.
Article in English | MEDLINE | ID: mdl-27671070

ABSTRACT

P128 is an antistaphylococcal protein, comprising a cell wall-degrading enzymatic region and a Staphylococcus-specific binding region, which possesses specific and potent bactericidal activity against sensitive and drug-resistant strains of Staphylococcus aureus To explore P128's ability to kill S. aureus in a range of environments relevant to clinical infection, we investigated the anti-S. aureus activity of P128 alone and in combination with standard-of-care antibiotics on planktonic and biofilm-embedded cells. P128 was found to have potent antibiofilm activity on preformed S. aureus biofilms as detected by CFU reduction and a colorimetric minimum biofilm inhibitory concentration (MBIC) assay. Scanning electron microscopic images of biofilms formed on the surfaces of microtiter plates and on catheters showed that P128 at low concentrations could destroy the biofilm structure and lyse the cells. When it was tested in combination with antibiotics which are known to be poor inhibitors of S. aureus in biofilms, such as vancomycin, gentamicin, ciprofloxacin, linezolid, and daptomycin, P128 showed highly synergistic antibiofilm activity that resulted in much reduced MBIC values for P128 and the individual antibiotics. The synergistic effect was seen for both sensitive and resistant isolates of S. aureus Additionally, in an in vitro mixed-biofilm model mimicking the wound infection environment, P128 was able to prevent biofilm formation by virtue of its anti-Staphylococcus activity. The potent S. aureus biofilm-inhibiting activity of P128 both alone and in combination with antibiotics is an encouraging sign for the development of P128 for treatment of complicated S. aureus infections involving biofilms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Recombinant Fusion Proteins/pharmacology , Staphylococcus aureus/drug effects , Biofilms/growth & development , Ciprofloxacin/pharmacology , Daptomycin/pharmacology , Drug Combinations , Drug Resistance, Multiple, Bacterial , Gentamicins/pharmacology , Linezolid/pharmacology , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Staphylococcus aureus/growth & development , Vancomycin/pharmacology
7.
PLoS One ; 10(8): e0134562, 2015.
Article in English | MEDLINE | ID: mdl-26247874

ABSTRACT

Conditional expression strains serve as a valuable tool to study the essentiality and to establish the vulnerability of a target under investigation in a drug discovery program. While essentiality implies an absolute requirement of a target function, vulnerability provides valuable information on the extent to which a target function needs to be depleted to achieve bacterial growth inhibition followed by cell death. The critical feature of an ideal conditional expression system is its ability to tightly regulate gene expression to achieve the full spectrum spanning from a high level of expression in order to support growth and near zero level of expression to mimic conditions of gene knockout. A number of bacterial conditional expression systems have been reported for use in mycobacteria. The utility of an isopropylthiogalactoside (IPTG) inducible system in mycobacteria has been reported for protein overexpression and anti-sense gene expression from a replicating multi-copy plasmid. Herein, we report the development of a versatile set of non-replicating IPTG inducible vectors for mycobacteria which can be used for generation of conditional expression strains through homologous recombination. The role of a single lac operator versus a double lac operator to regulate gene expression was evaluated by monitoring the expression levels of ß-galactosidase in Mycobacterium smegmatis. These studies indicated a significant level of leaky expression from the vector with a single lac operator but none from the vector with double lac operator. The significance of the double lac operator vector for target validation was established by monitoring the growth kinetics of an inhA, a rpoB and a ftsZ conditional expression strain grown in the presence of different concentrations of IPTG. The utility of this inducible system in identifying target specific inhibitors was established by screening a focussed library of small molecules using an inhA and a rpoB conditional expression strain.


Subject(s)
Gene Expression Regulation, Bacterial/drug effects , Isopropyl Thiogalactoside/pharmacology , Mycobacterium smegmatis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Lac Operon/genetics , Mycobacterium smegmatis/growth & development , Oxidoreductases/genetics , Oxidoreductases/metabolism , Phenotype , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
8.
Tuberculosis (Edinb) ; 95(5): 589-98, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26073894

ABSTRACT

DNA topoisomerases perform the essential function of maintaining DNA topology in prokaryotes. DNA gyrase, an essential enzyme that introduces negative supercoils, is a clinically validated target. However, topoisomerase I (Topo I), an enzyme responsible for DNA relaxation has received less attention as an antibacterial target, probably due to the ambiguity over its essentiality in many organisms. The Mycobacterium tuberculosis genome harbors a single topA gene with no obvious redundancy in its function suggesting an essential role. The topA gene could be inactivated only in the presence of a complementing copy of the gene in M. tuberculosis. Furthermore, down-regulation of topA in a genetically engineered strain of M. tuberculosis resulted in loss of bacterial viability which correlated with a concomitant depletion of intracellular Topo I levels. The topA knockdown strain of M. tuberculosis failed to establish infection in a murine model of TB and was cleared from lungs in two months post infection. Phenotypic screening of a Topo I overexpression strain led to the identification of an inhibitor, thereby providing chemical validation of this target. Thus, our work confirms the attractiveness of Topo I as an anti-mycobacterial target.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , DNA Topoisomerases, Type I , Drug Discovery , Mycobacterium tuberculosis/drug effects , Topoisomerase I Inhibitors/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Gene Expression Regulation, Bacterial , Gene Knockdown Techniques , Genotype , Humans , Microbial Viability , Molecular Targeted Therapy , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/pathogenicity , Phenotype , Time Factors
9.
Gene ; 555(2): 269-76, 2015 Jan 25.
Article in English | MEDLINE | ID: mdl-25447907

ABSTRACT

Glutamate racemase (MurI) converts l-glutamate into d-glutamate which is an essential component of peptidoglycan in bacteria. The gene encoding glutamate racemase, murI has been shown to be essential for the growth of a number of bacterial species including Escherichia coli. However, in some Gram-positive species d-amino acid transaminase (Dat) can also convert l-glutamate into d-glutamate thus rendering MurI non-essential for growth. In a recent study the murI gene of Mycobacterium tuberculosis was shown to be non-essential. As d-glutamate is an essential component of peptidoglycan of M. tuberculosis, either Dat or MurI has to be essential for its survival. Since, a Dat encoding gene has not been reported in M. tuberculosis genome sequence, the reported non-essentiality of murI was unexplainable. In order to resolve this dilemma we tried to knockout murI in the presence of single and two copies of murI, in wild type and merodiploid strains respectively. It was found that murI could not be inactivated in the wild type background indicating that it could be an essential gene. Also, inactivation of murI could not be achieved in the presence of externally supplied d-glutamate in 7H9 medium suggesting that M. tuberculosis is unable to take up d-glutamate under the conditions tested. However we could generate murI knockout strains at high frequency when two copies of the gene were present indicating that at least one murI gene is required for cellular viability. The essential nature of MurI in M. tuberculosis H37Rv suggests that it could be a potential drug target.


Subject(s)
Amino Acid Isomerases/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Amino Acid Isomerases/genetics , Amino-Acid N-Acetyltransferase/genetics , Bacterial Proteins/genetics , Cell Wall/chemistry , Computational Biology , Electroporation , Gene Deletion , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Genetic , Mutation , Mycobacterium tuberculosis/genetics , Peptidoglycan/chemistry , Promoter Regions, Genetic , Recombination, Genetic , Transgenes
10.
Gene ; 550(1): 110-6, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128581

ABSTRACT

Most bacteria are able to generate sufficient amounts of ATP from substrate level phosphorylation, thus rendering the respiratory oxidative phosphorylation non-critical. In mycobacteria, including Mycobacterium tuberculosis, ATP generation by oxidative phosphorylation is an essential process. Of the two types of NADH dehydrogenases (type I and type II), the type II NADH dehydrogenase (Ndh) which is inhibited by phenothiazines has been thought to be essential. In M. tuberculosis there are two Ndh isozymes (Ndh and NdhA) coded by ndh and ndhA genes respectively. Ndh and NdhA share a high degree of amino acid similarity. Both the enzymes have been shown to be enzymatically active and are inhibited by phenothiazines, suggesting a functional similarity between the two. We attempted gene knockout of ndh and ndhA genes in wild type and merodiploid backgrounds. It was found that ndh gene cannot be inactivated in a wild type background, though it was possible to do so when an additional copy of ndh was provided. This showed that in spite of its apparent functional equivalence, NdhA cannot complement the loss of Ndh in M. tuberculosis. We also showed that NdhA is not essential in M. tuberculosis as the ndhA gene could be deleted in a wild type strain of M. tuberculosis without causing any adverse effects in vitro. RT-PCR analysis of in vitro grown M. tuberculosis showed that ndhA gene is actively transcribed. This study suggests that despite being biochemically similar, Ndh and NdhA play different roles in the physiology of M. tuberculosis.


Subject(s)
Bacterial Proteins/genetics , Microbial Viability/genetics , Mycobacterium tuberculosis/genetics , NADH Dehydrogenase/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockout Techniques , Isoenzymes/genetics , Isoenzymes/metabolism , Mycobacterium tuberculosis/enzymology , NADH Dehydrogenase/metabolism , Phenothiazines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
11.
Microbiology (Reading) ; 160(Pt 10): 2157-2169, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25023246

ABSTRACT

P128 is an anti-staphylococcal protein consisting of the Staphylococcus aureus phage-K-derived tail-associated muralytic enzyme (TAME) catalytic domain (Lys16) fused with the cell-wall-binding SH3b domain of lysostaphin. In order to understand the mechanism of action and emergence of resistance to P128, we isolated mutants of Staphylococcus spp., including meticillin-resistant Staphylococcus aureus (MRSA), resistant to P128. In addition to P128, the mutants also showed resistance to Lys16, the catalytic domain of P128. The mutants showed loss of fitness as shown by reduced rate of growth in vitro. One of the mutants tested was found to show reduced virulence in animal models of S. aureus septicaemia suggesting loss of fitness in vivo as well. Analysis of the antibiotic sensitivity pattern showed that the mutants derived from MRSA strains had become sensitive to meticillin and other ß-lactams. Interestingly, the mutant cells were resistant to the lytic action of phage K, although the phage was able to adsorb to these cells. Sequencing of the femA gene of three P128-resistant mutants showed either a truncation or deletion in femA, suggesting that improper cross-bridge formation in S. aureus could be causing resistance to P128. Using glutathione S-transferase (GST) fusion peptides as substrates it was found that both P128 and Lys16 were capable of cleaving a pentaglycine sequence, suggesting that P128 might be killing S. aureus by cleaving the pentaglycine cross-bridge of peptidoglycan. Moreover, peptides corresponding to the reported cross-bridge of Staphylococcus haemolyticus (GGSGG, AGSGG), which were not cleaved by lysostaphin, were cleaved efficiently by P128. This was also reflected in high sensitivity of S. haemolyticus to P128. This showed that in spite of sharing a common mechanism of action with lysostaphin, P128 has unique properties, which allow it to act on certain lysostaphin-resistant Staphylococcus strains.


Subject(s)
Cell Wall/metabolism , Peptide Hydrolases/metabolism , Peptidoglycan/metabolism , Staphylococcus Phages/enzymology , Staphylococcus/drug effects , Animals , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/metabolism , Disease Models, Animal , Drug Resistance, Bacterial , Peptide Hydrolases/genetics , Peptide Hydrolases/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sepsis/microbiology , Staphylococcus/growth & development , Staphylococcus/isolation & purification , Virulence
12.
J Med Chem ; 57(11): 4889-905, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24809953

ABSTRACT

DNA gyrase is a clinically validated target for developing drugs against Mycobacterium tuberculosis (Mtb). Despite the promise of fluoroquinolones (FQs) as anti-tuberculosis drugs, the prevalence of pre-existing resistance to FQs is likely to restrict their clinical value. We describe a novel class of N-linked aminopiperidinyl alkyl quinolones and naphthyridones that kills Mtb by inhibiting the DNA gyrase activity. The mechanism of inhibition of DNA gyrase was distinct from the fluoroquinolones, as shown by their ability to inhibit the growth of fluoroquinolone-resistant Mtb. Biochemical studies demonstrated this class to exert its action via single-strand cleavage rather than double-strand cleavage, as seen with fluoroquinolones. The compounds are highly bactericidal against extracellular as well as intracellular Mtb. Lead optimization resulted in the identification of potent compounds with improved oral bioavailability and reduced cardiac ion channel liability. Compounds from this series are efficacious in various murine models of tuberculosis.


Subject(s)
Antitubercular Agents/chemical synthesis , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Piperidines/chemical synthesis , Topoisomerase II Inhibitors/chemical synthesis , Acute Disease , Administration, Oral , Animals , Antitubercular Agents/pharmacokinetics , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Availability , Chronic Disease , DNA Gyrase/genetics , DNA Gyrase/metabolism , Drug Resistance, Bacterial , ERG1 Potassium Channel , Fluoroquinolones/pharmacology , Humans , Macrophages/drug effects , Macrophages/microbiology , Mice, Inbred BALB C , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutation , Mycobacterium tuberculosis/enzymology , Piperidines/pharmacokinetics , Piperidines/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Stereoisomerism , Structure-Activity Relationship , Topoisomerase II Inhibitors/pharmacokinetics , Topoisomerase II Inhibitors/pharmacology , Tuberculosis, Pulmonary/drug therapy
13.
Tuberculosis (Edinb) ; 92(6): 521-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22954585

ABSTRACT

Coenzyme A biosynthesis pathway proteins are potential targets for developing inhibitors against bacteria including Mycobacterium tuberculosis. We have evaluated two enzymes in this pathway: phosphopantetheine adenylyltransferase (CoaD) and dephospho CoA kinase (CoaE) for essentiality and selectivity. Based on the previous transposon mutagenesis studies, coaD had been predicted to be a non-essential gene in M. tuberculosis. Our bioinformatics analysis showed that there is no other functional homolog of this enzyme in M. tuberculosis, which suggests that coaD should be an essential gene. In order to get an unambiguous answer on the essentiality of coaD, we attempted inactivation of coaD in wild type and merodiploid backgrounds. It was found that coaD could only be inactivated in the presence of an additional gene copy, confirming it to be an essential gene. Using a similar approach we found that CoaE was also essential for the survival of M. tuberculosis. RT-PCR analysis showed that both coaD and coaE were transcribed in M. tuberculosis. Amino acids alignment and phylogenetic analysis showed CoaD to be distantly related to the human counterpart while CoaE was found to be relatively similar to the human enzyme. Analysis of CoaD and CoaE structures at molecular level allowed us to identify unique residues in the Mtb proteins, thus providing a selectivity handle. The essentiality and selectivity analysis combined with the published biochemical characterization of CoaD and CoaE makes them suitable targets for developing inhibitors against M. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Coenzyme A/biosynthesis , Mycobacterium tuberculosis/immunology , Nucleotidyltransferases/biosynthesis , Transferases/biosynthesis , Tuberculosis, Multidrug-Resistant/immunology , Coenzyme A/genetics , Computational Biology , Humans , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/genetics
14.
Antimicrob Agents Chemother ; 56(5): 2643-51, 2012 May.
Article in English | MEDLINE | ID: mdl-22314527

ABSTRACT

Active efflux of drugs mediated by efflux pumps that confer drug resistance is one of the mechanisms developed by bacteria to counter the adverse effects of antibiotics and chemicals. To understand these efflux mechanisms in Mycobacterium tuberculosis, we generated knockout (KO) mutants of four efflux pumps of the pathogen belonging to different classes. We measured the MICs and kill values of two different compound classes on the wild type (WT) and the efflux pump (EP) KO mutants in the presence and absence of the efflux inhibitors verapamil and l-phenylalanyl-l-arginyl-ß-naphthylamide (PAßN). Among the pumps studied, the efflux pumps belonging to the ABC (ATP-binding cassette) class, encoded by Rv1218c, and the SMR (small multidrug resistance) class, encoded by Rv3065, appear to play important roles in mediating the efflux of different chemical classes and antibiotics. Efflux pumps encoded by Rv0849 and Rv1258c also mediate the efflux of these compounds, but to a lesser extent. Increased killing is observed in WT M. tuberculosis cells by these compounds in the presence of either verapamil or PAßN. The efflux pump KO mutants were more susceptible to these compounds in the presence of efflux inhibitors. We have shown that these four efflux pumps of M. tuberculosis play a vital role in mediating efflux of different chemical scaffolds. Inhibitors of one or several of these efflux pumps could have a significant impact in the treatment of tuberculosis. The identification and characterization of Rv0849, a new efflux pump belonging to the MFS (major facilitator superfamily) class, are reported.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Mycobacterium tuberculosis/metabolism , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Culture Media , Dipeptides/pharmacology , Drug Combinations , Drug Resistance, Multiple, Bacterial/drug effects , Drug Synergism , Gene Knockout Techniques , Homologous Recombination , Humans , Microbial Sensitivity Tests , Mutation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Plasmids , Pyrazolones/pharmacology , Pyrroles/pharmacology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology , Verapamil/pharmacology
15.
Microbiology (Reading) ; 158(Pt 2): 319-327, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075031

ABSTRACT

Alanine racemase (Alr) is an essential enzyme in most bacteria; however, some species (e.g. Listeria monocytogenes) can utilize d-amino acid transaminase (Dat) to generate d-alanine, which renders Alr non-essential. In addition to the conflicting reports on gene knockout of alr in Mycobacterium smegmatis, a recent study concluded that depletion of Alr does not affect the growth of M. smegmatis. In order to get an unambiguous answer on the essentiality of Alr in Mycobacterium tuberculosis and validate it as a drug target in vitro and in vivo, we have inactivated the alr gene of M. tuberculosis and found that it was not possible to generate an alr knockout in the absence of a complementing gene copy or d-alanine in the growth medium. The growth kinetics of the alr mutant revealed that M. tuberculosis requires very low amounts of d-alanine (5-10 µg ml(-1)) for optimum growth. Survival kinetics of the mutant in the absence of d-alanine indicated that depletion of this amino acid results in rapid loss of viability. The alr mutant was found to be defective for growth in macrophages. Analysis of phenotype in mice suggested that non-availability of d-alanine in mice leads to clearance of bacteria followed by stabilization of bacterial number in lungs and spleen. Additionally, reversal of d-cycloserine inhibition in the presence of d-alanine in M. tuberculosis suggested that Alr is the primary target of d-cycloserine. Thus, Alr of M. tuberculosis is a valid drug target and inhibition of Alr alone should result in loss of viability in vitro and in vivo.


Subject(s)
Alanine Racemase/genetics , Alanine/metabolism , Bacterial Proteins/genetics , Macrophages/microbiology , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/growth & development , Tuberculosis/microbiology , Alanine Racemase/metabolism , Animals , Bacterial Proteins/metabolism , Cells, Cultured , Humans , Mice , Mice, Inbred BALB C , Microbial Viability , Mutation , Mycobacterium tuberculosis/genetics
16.
Expert Opin Drug Discov ; 6(11): 1171-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22646985

ABSTRACT

INTRODUCTION: Target driven drug discovery is a long and arduous task requiring a huge investment of time, energy and resources. Therefore, it is very important to select targets which provide the maximum chance of obtaining inhibitors that will be efficacious in animal models and finally in tuberculosis (TB) patients. AREAS COVERED: The article discusses the necessity for new targets in Mycobacterium tuberculosis (Mtb) drug discovery and how the functional redundancy of putative targets in Mtb adds a new dimension to the complexity of validation. The article also reviews survival kinetics using conditional knockout (KO) or knockdown (KD) strains and discusses how this has provided crucial information on target vulnerability. Furthermore, the article also comments on how the chemical validation of new targets using specific inhibitors has greatly supplemented the genetic validation efforts. EXPERT OPINION: Because of complexity of pathogenesis of TB, the putative drug targets need to be validated under multiple physiological conditions. Target protein depletion can mimic chemical inhibition and, therefore, will be a valuable tool in predicting the vulnerability of a target. Conditional KO or KD makes it possible to study the phenotypes of Mtb strains under a variety of physiological states. The phenotype of these strains should also be tested in animal models which mimic human TB more closely. Finally, inhibitors with confirmed mode of action can be important tools for validating Mtb drug targets.

17.
Antimicrob Agents Chemother ; 54(12): 5167-72, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20921309

ABSTRACT

Efflux systems are important in determining the efficacy of antibiotics used in the treatment of bacterial infections. In the last decade much attention has been paid to studying the efflux pumps of mycobacteria. New classes of compounds are under investigation for development into potential candidate drugs for the treatment of tuberculosis. Quite often, these have poor bactericidal activities but exhibit excellent target (biochemical) inhibition. Microarray studies conducted in our laboratories for deciphering the mode of action of experimental drugs revealed the presence of putative ABC transporters. Among these transporters, Rv1218c was chosen for studying its physiological relevance in mediating efflux in Mycobacterium tuberculosis. A ΔRv1218c mutant of M. tuberculosis displayed a 4- to 8-fold increase in the inhibitory and bactericidal potency for different classes of compounds. The MICs and MBCs were reversed to wild-type values when the full-length Rv1218c gene was reintroduced into the ΔRv1218c mutant on a multicopy plasmid. Most of the compound classes had significantly better bactericidal activity in the ΔRv1218c mutant than in the wild-type H37Rv, suggesting the involvement of Rv1218c gene product in effluxing these compounds from M. tuberculosis. The implication of these findings on tuberculosis drug discovery is discussed.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/drug effects , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Drug Discovery , Microbial Sensitivity Tests , Mycobacterium tuberculosis/genetics , Pyrazolones/pharmacology , Reserpine/pharmacology , Verapamil/pharmacology
18.
Microbiology (Reading) ; 156(Pt 9): 2691-2701, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20576686

ABSTRACT

Pantothenate kinase, an essential enzyme in bacteria and eukaryotes, is involved in catalysing the first step of conversion of pantothenate to coenzyme A (CoA). Three isoforms (type I, II and III) of this enzyme have been reported from various organisms, which can be differentiated from each other on the basis of their biochemical and structural characteristics. Though most bacteria carry only one of the isoforms of pantothenate kinases, some of them possess two isoforms. The physiological relevance of the presence of two types of isozymes in a single organism is not clear. Mycobacterium tuberculosis, an intracellular pathogen, possesses two isoforms of pantothenate kinases (CoaA and CoaX) belonging to type I and III. In order to determine which pantothenate kinase is essential in mycobacteria, we performed gene inactivation of coaA and coaX of M. tuberculosis individually. It was found that coaA could only be inactivated in the presence of an extra copy of the gene, while coaX could be inactivated in the wild-type cells, proving that CoaA is the essential pantothenate kinase in M. tuberculosis. Additionally, the coaA gene of M. tuberculosis was able to complement a temperature-sensitive coaA mutant of Escherichia coli at a non-permissive temperature while coaX could not. The coaX deletion mutant showed no growth defects in vitro, in macrophages or in mice. Taken together, our data suggest that CoaX, which is essential in Bacillus anthracis and thus had been suggested to be a drug target in this organism, might not be a valid target in M. tuberculosis. We have established that the type I isoform, CoaA, is the essential pantothenate kinase in M. tuberculosis and thus can be explored as a drug target.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sequence Deletion , Tuberculosis/microbiology
19.
FEMS Microbiol Rev ; 34(5): 646-57, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20491934

ABSTRACT

During active growth of Escherichia coli, majority of the transcriptional activity is carried out by the housekeeping sigma factor (sigma(70)), whose association with core RNAP is generally favoured because of its higher intracellular level and higher affinity to core RNAP. In order to facilitate transcription by alternative sigma factors during nutrient starvation, the bacterial cell uses multiple strategies by which the transcriptional ability of sigma(70) is diminished in a reversible manner. The facilitators of shifting the balance in favour of alternative sigma factors happen to be as diverse as a small molecule (p)ppGpp (represents ppGpp or pppGpp), proteins (DksA, Rsd) and a species of RNA (6S RNA). Although 6S RNA and (p)ppGpp were known in literature for a long time, their role in transcriptional switching has been understood only in recent years. With the elucidation of function of DksA, a new dimension has been added to the phenomenon of stringent response. As the final outcome of actions of (p)ppGpp, DksA, 6S RNA and Rsd is similar, there is a need to analyse these mechanisms in a collective manner. We review the recent trends in understanding the regulation of sigma(70) by (p)ppGpp, DksA, Rsd and 6S RNA and present a case for evolving a unified model of RNAP redistribution during starvation by modulation of sigma(70) activity in E. coli.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Gene Expression Regulation, Bacterial , Sigma Factor/metabolism , Stress, Physiological/genetics , Transcription, Genetic , DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Guanosine Tetraphosphate/physiology , RNA, Bacterial/physiology , RNA, Untranslated , Repressor Proteins/physiology , Sigma Factor/genetics
20.
Microbiology (Reading) ; 155(Pt 9): 2978-2987, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19542000

ABSTRACT

Acetohydroxyacid synthase (AHAS) is the first enzyme in the branched-chain amino acid biosynthesis pathway in bacteria. Bioinformatics analysis revealed that the Mycobacterium tuberculosis genome contains four genes (ilvB1, ilvB2, ilvG and ilvX) coding for the large catalytic subunit of AHAS, whereas only one gene (ilvN or ilvH) coding for the smaller regulatory subunit of this enzyme was found. In order to understand the physiological role of AHAS in survival of the organism in vitro and in vivo, we inactivated the ilvB1 gene of M. tuberculosis. The mutant strain was found to be auxotrophic for all of the three branched-chain amino acids (isoleucine, leucine and valine), when grown with either C(6) or C(2) carbon sources, suggesting that the ilvB1 gene product is the major AHAS in M. tuberculosis. Depletion of these branched chain amino acids in the medium led to loss of viability of the DeltailvB1 strain in vitro, resulting in a 4-log reduction in colony-forming units after 10 days. Survival kinetics of the mutant strain cultured in macrophages maintained with sub-optimal concentrations of the branched-chain amino acids did not show any loss of viability, indicating either that the intracellular environment was rich in these amino acids or that the other AHAS catalytic subunits were functional under these conditions. Furthermore, the growth kinetics of the DeltailvB1 strain in mice indicated that although this mutant strain showed defective growth in vivo, it could persist in the infected mice for a long time, and therefore could be a potential vaccine candidate.


Subject(s)
Acetolactate Synthase , Amino Acids, Branched-Chain/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/microbiology , Acetolactate Synthase/deficiency , Acetolactate Synthase/genetics , Animals , Cell Culture Techniques , Gene Deletion , Genes, Bacterial , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/genetics , Tuberculosis Vaccines/therapeutic use , Tuberculosis, Pulmonary/prevention & control , Vaccines, Attenuated/therapeutic use , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...