Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Drug Discov Today ; 26(9): 2173-2181, 2021 09.
Article in English | MEDLINE | ID: mdl-33845218

ABSTRACT

The increasing prevalence of multidrug-resistant (MDR) bacterial infections has created a crucial need for new therapeutics that avoid or minimize existing resistance mechanisms. In this review, we describe the development of novel classes of small-molecule adjunctive agents targeting either a bacterial virulence factor, the Pseudomonas aeruginosa type III secretion system (T3SS), or an intrinsic resistance factor, resistance-nodulation-cell division superfamily (RND) efflux pumps of the Enterobacteriaceae. These agents are designed to be administered with antibacterials to improve their efficacy. T3SS inhibition rescues host innate immune system cells from injection with bacterial toxins, whereas RND efflux pump inhibition increases antibiotic susceptibility, in both cases improving the efficacy of the combined antibacterial.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Drug Resistance, Multiple, Bacterial/drug effects , Gram-Negative Bacterial Infections/drug therapy , Membrane Transport Proteins/metabolism , Type III Secretion Systems/antagonists & inhibitors , Animals , Bacterial Proteins/metabolism , Humans , Type III Secretion Systems/metabolism
2.
J Bacteriol ; 202(18)2020 08 25.
Article in English | MEDLINE | ID: mdl-32601072

ABSTRACT

The Pseudomonas aeruginosa type III secretion system (T3SS) needle comprised of multiple PscF subunits is essential for the translocation of effector toxins into human cells, facilitating the establishment and dissemination of infection. Mutations in the pscF gene provide resistance to the phenoxyacetamide (PhA) series of T3SS inhibitory chemical probes. To better understand PscF functions and interactions with PhA, alleles of pscF with 71 single mutations altering 49 of the 85 residues of the encoded protein were evaluated for their effects on T3SS phenotypes. Of these, 37% eliminated and 63% maintained secretion, with representatives of both evenly distributed across the entire protein. Mutations in 14 codons conferred a degree of PhA resistance without eliminating secretion, and all but one were in the alpha-helical C-terminal 25% of PscF. PhA-resistant mutants exhibited no cross-resistance to two T3SS inhibitors with different chemical scaffolds. Two mutations caused constitutive T3SS secretion. The pscF allele at its native locus, whether wild type (WT), constitutive, or PhA resistant, was dominant over other pscF alleles expressed from nonnative loci and promoters, but mixed phenotypes were observed in chromosomal ΔpscF strains with both WT and mutant alleles at nonnative loci. Some PhA-resistant mutants exhibited reduced translocation efficiency that was improved in a PhA dose-dependent manner, suggesting that PhA can bind to those resistant needles. In summary, these results are consistent with a direct interaction between PhA inhibitors and the T3SS needle, suggest a mechanism of blocking conformational changes, and demonstrate that PscF affects T3SS regulation, as well as carrying out secretion and translocation.IMPORTANCEP. aeruginosa effector toxin translocation into host innate immune cells is critical for the establishment and dissemination of P. aeruginosa infections. The medical need for new anti-P. aeruginosa agents is evident by the fact that P. aeruginosa ventilator-associated pneumonia is associated with a high mortality rate (40 to 69%) and recurs in >30% of patients, even with standard-of-care antibiotic therapy. The results described here confirm roles for the PscF needle in T3SS secretion and translocation and suggest that it affects regulation, possibly by interaction with T3SS regulatory proteins. The results also support a model of direct interaction of the needle with PhA and suggest that, with further development, members of the PhA series may prove useful as drugs for P. aeruginosa infection.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/metabolism , Pseudomonas aeruginosa/drug effects , Type III Secretion Systems/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Drug Resistance, Microbial/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mutation , Phenoxyacetates/pharmacology , Pseudomonas aeruginosa/genetics , Structure-Activity Relationship
3.
ACS Infect Dis ; 6(3): 489-502, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31899866

ABSTRACT

Marburg virus (MARV) causes severe disease in humans and is known to activate nuclear factor erythroid 2-related factor 2 (Nrf2), the major transcription factor of the antioxidant response. Canonical activation of Nrf2 involves oxidative or electrophilic stress that prevents Kelch-like ECH-associated protein 1 (Keap1) targeted degradation of Nrf2, leading to Nrf2 stabilization and activation of the antioxidant response. MARV activation of Nrf2 is noncanonical with the MARV VP24 protein (mVP24) interacting with Keap1, freeing Nrf2 from degradation. A high-throughput screening (HTS) assay was developed to identify inhibitors of mVP24-induced Nrf2 activity and used to screen more than 55,000 compounds. Hit compounds were further screened against secondary HTS assays for the inhibition of antioxidant activity induced by additional canonical and noncanonical mechanisms. This pipeline identified 14 compounds that suppress the response, dependent on the inducer, with 50% inhibitory concentrations below 5 µM and selectivity index values greater than 10. Notably, several of the identified compounds specifically inhibit mVP24-induced Nrf2 activity.


Subject(s)
Gene Expression/drug effects , Marburgvirus/drug effects , NF-E2-Related Factor 2/antagonists & inhibitors , Oxidation-Reduction/drug effects , Small Molecule Libraries/pharmacology , Antioxidants , Gene Expression Regulation , HEK293 Cells , High-Throughput Screening Assays , Humans , NF-E2-Related Factor 2/genetics , Protein Binding , Viral Proteins/metabolism
4.
Article in English | MEDLINE | ID: mdl-28807906

ABSTRACT

Pseudomonas aeruginosa is a leading cause of intra-abdominal infections, wound infections, and community-acquired folliculitis, each of which may involve macro- or microabscess formation. The rising incidence of multidrug resistance among P. aeruginosa isolates has increased both the economic burden and the morbidity and mortality associated with P. aeruginosa disease and necessitates a search for novel therapeutics. Previous work from our group detailed novel phenoxyacetamide inhibitors that block type III secretion and injection into host cells in vitro In this study, we used a mouse model of P. aeruginosa abscess formation to test the in vivo efficacy of these compounds against the P. aeruginosa type III secretion system (T3SS). Bacteria used the T3SS to intoxicate infiltrating neutrophils to establish abscesses. Despite this antagonism, sufficient numbers of functioning neutrophils remained for proper containment of the abscesses, as neutrophil depletion resulted in an increased abscess size, the formation of dermonecrotic lesions on the skin, and the dissemination of P. aeruginosa to internal organs. Consistent with the specificity of the T3SS-neutrophil interaction, P. aeruginosa bacteria lacking a functional T3SS were fully capable of causing abscesses in a neutropenic host. Phenoxyacetamide inhibitors attenuated abscess formation and aided in the immune clearance of the bacteria. Finally, a P. aeruginosa strain resistant to the phenoxyacetamide compound was fully capable of causing abscess formation even in the presence of the T3SS inhibitors. Together, our results further define the role of type III secretion in murine abscess formation and demonstrate the in vivo efficacy of phenoxyacetamide inhibitors in P. aeruginosa infection.


Subject(s)
Abscess/microbiology , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Abscess/drug therapy , Abscess/pathology , Animals , Anti-Bacterial Agents/chemistry , Disease Models, Animal , Host-Pathogen Interactions , Mice, Inbred C57BL , Neutropenia/microbiology , Neutrophils/pathology , Phenoxyacetates/chemistry , Pseudomonas Infections/complications , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Type III Secretion Systems , Virulence Factors/metabolism
5.
ACS Infect Dis ; 3(3): 190-198, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28152588

ABSTRACT

The 2014 Ebola outbreak in West Africa, the largest outbreak on record, highlighted the need for novel approaches to therapeutics targeting Ebola virus (EBOV). Within the EBOV replication complex, the interaction between polymerase cofactor, viral protein 35 (VP35), and nucleoprotein (NP) is critical for viral RNA synthesis. We recently identified a peptide at the N-terminus of VP35 (termed NPBP) that is sufficient for interaction with NP and suppresses EBOV replication, suggesting that the NPBP binding pocket can serve as a potential drug target. Here we describe the development and validation of a sensitive high-throughput screen (HTS) using a fluorescence polarization assay. Initial hits from this HTS include the FDA-approved compound tolcapone, whose potency against EBOV infection was validated in a nonfluorescent secondary assay. High conservation of the NP-VP35 interface among filoviruses suggests that this assay has the capacity to identify pan-filoviral inhibitors for development as antivirals.


Subject(s)
Antiviral Agents/pharmacology , Filoviridae/physiology , Nucleoproteins/metabolism , Viral Regulatory and Accessory Proteins/chemistry , Amino Acid Sequence , Binding Sites/drug effects , Conserved Sequence , Drug Evaluation, Preclinical , Filoviridae/drug effects , Filoviridae/genetics , Fluorescence Polarization , High-Throughput Screening Assays , In Vitro Techniques , Models, Molecular , Protein Binding/drug effects , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication/drug effects
6.
Antimicrob Agents Chemother ; 60(12): 7067-7076, 2016 12.
Article in English | MEDLINE | ID: mdl-27620482

ABSTRACT

We previously reported the synthesis and biological activity of a series of cationic bis-indoles with potent, broad-spectrum antibacterial properties. Here, we describe mechanism of action studies to test the hypothesis that these compounds bind to DNA and that this target plays an important role in their antibacterial outcome. The results reported here indicate that the bis-indoles bind selectively to DNA at A/T-rich sites, which is correlated with the inhibition of DNA and RNA synthesis in representative Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli) organisms. Further, exposure of E. coli and S. aureus to representative bis-indoles resulted in induction of the DNA damage-inducible SOS response. In addition, the bis-indoles were found to be potent inhibitors of cell wall biosynthesis; however, they do not induce the cell wall stress stimulon in S. aureus, suggesting that this pathway is inhibited by an indirect mechanism. In light of these findings, the most likely basis for the observed activities of these compounds is their ability to bind to the minor groove of DNA, resulting in the inhibition of DNA and RNA synthesis and other secondary effects.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA/metabolism , Indoles/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Escherichia coli/drug effects , HeLa Cells/drug effects , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/pharmacology , Indoles/chemistry , Indoles/metabolism , Microbial Sensitivity Tests , Microscopy, Fluorescence , Molecular Targeted Therapy , SOS Response, Genetics/drug effects , Staphylococcus aureus/drug effects
7.
Clin Infect Dis ; 63(1): 89-95, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27025826

ABSTRACT

The utility of conventional antibiotics for the treatment of bacterial infections has become increasingly strained due to increased rates of resistance coupled with reduced rates of development of new agents. As a result, multidrug-resistant, extensively drug-resistant, and pandrug-resistant bacterial strains are now frequently encountered. This has led to fears of a "postantibiotic era" in which many bacterial infections will be untreatable. Alternative nonantibiotic treatment strategies need to be explored to ensure that a robust pipeline of effective therapies is available to clinicians. In this review, we highlight some of the recent developments in this area, such as the targeting of bacterial virulence factors, utilization of bacteriophages to kill bacteria, and manipulation of the microbiome to combat infections.


Subject(s)
Bacterial Infections , Microbiota , Phage Therapy/methods , Virulence Factors/therapeutic use , Animals , Anti-Bacterial Agents , Bacterial Infections/microbiology , Bacterial Infections/therapy , Bacterial Secretion Systems , Bacteriophages , Biomedical Research , Humans , Mice
8.
J Infect Dis ; 212 Suppl 2: S425-34, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26206510

ABSTRACT

BACKGROUND: The current Ebola virus (EBOV) outbreak has highlighted the troubling absence of available antivirals or vaccines to treat infected patients and stop the spread of EBOV. The EBOV glycoprotein (GP) plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a potential target for the development of anti-EBOV drugs. We report the identification of 2 novel EBOV inhibitors targeting viral entry. METHODS: To identify small molecule inhibitors of EBOV entry, we carried out a cell-based high-throughput screening using human immunodeficiency virus-based pseudotyped viruses expressing EBOV-GP. Two compounds were identified, and mechanism-of-action studies were performed using immunoflourescence, AlphaLISA, and enzymatic assays for cathepsin B inhibition. RESULTS: We report the identification of 2 novel entry inhibitors. These inhibitors (1) inhibit EBOV infection (50% inhibitory concentration, approximately 0.28 and approximately 10 µmol/L) at a late stage of entry, (2) induce Niemann-Pick C phenotype, and (3) inhibit GP-Niemann-Pick C1 (NPC1) protein interaction. CONCLUSIONS: We have identified 2 novel EBOV inhibitors, MBX2254 and MBX2270, that can serve as starting points for the development of an anti-EBOV therapeutic agent. Our findings also highlight the importance of NPC1-GP interaction in EBOV entry and the attractiveness of NPC1 as an antifiloviral therapeutic target.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Small Molecule Libraries/pharmacology , Animals , Carrier Proteins/metabolism , Cell Line , Chlorocebus aethiops , Glycoproteins/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Niemann-Pick C1 Protein , Protein Binding/drug effects , Vero Cells , Virus Internalization/drug effects
9.
Antimicrob Agents Chemother ; 59(9): 5775-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26169404

ABSTRACT

Novel, cellular, gain-of-signal, bioluminescent reporter assays for fatty acid synthesis type II (FASII) inhibitors were constructed in an efflux-deficient strain of Pseudomonas aeruginosa and based on the discovery that FASII genes in P. aeruginosa are coordinately upregulated in response to pathway disruption. A screen of 115,000 compounds identified a series of sulfonamidobenzamide (SABA) analogs, which generated strong luminescent signals in two FASII reporter strains but not in four control reporter strains designed to respond to inhibitors of pathways other than FASII. The SABA analogs selectively inhibited lipid biosynthesis in P. aeruginosa and exhibited minimal cytotoxicity to mammalian cells (50% cytotoxic concentration [CC50] ≥ 80 µM). The most potent SABA analogs had MICs of 0.5 to 7.0 µM (0.2 to 3.0 µg/ml) against an efflux-deficient Escherichia coli (ΔtolC) strain but had no detectable MIC against efflux-proficient E. coli or against P. aeruginosa (efflux deficient or proficient). Genetic, molecular genetic, and biochemical studies revealed that SABA analogs target the enzyme (AccC) catalyzing the biotin carboxylase half-reaction of the acetyl coenzyme A (acetyl-CoA) carboxylase step in the initiation phase of FASII in E. coli and P. aeruginosa. These results validate the capability and the sensitivity of this novel bioluminescent reporter screen to identify inhibitors of E. coli and P. aeruginosa FASII.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fatty Acid Synthase, Type II/antagonists & inhibitors , Acetyl Coenzyme A/metabolism , Escherichia coli/drug effects , Escherichia coli/enzymology , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology
10.
Bioorg Med Chem ; 23(5): 1027-43, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25638499

ABSTRACT

The increasing prevalence of drug-resistant bacterial infections is driving the discovery and development not only of new antibiotics, but also of inhibitors of virulence factors that are crucial for in vivo pathogenicity. One such virulence factor is the type III secretion system (T3SS), which plays a critical role in the establishment and dissemination of Pseudomonas aeruginosa infections. We have recently described the discovery and characterization of a series of inhibitors of P. aeruginosa T3SS based on a phenoxyacetamide scaffold. To better characterize the factors involved in potent T3SS inhibition, we have conducted a systematic exploration of this structure, revealing several highly responsive structure-activity relationships indicative of interaction with a specific target. Most of the structural features contributing to potency were additive, and combination of those features produced optimized inhibitors with IC50 values <1µM.


Subject(s)
Acetates/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa/drug effects , Acetates/chemistry , Amides/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Pseudomonas aeruginosa/metabolism , Structure-Activity Relationship
11.
Antimicrob Agents Chemother ; 58(4): 2211-20, 2014.
Article in English | MEDLINE | ID: mdl-24468789

ABSTRACT

The type III secretion system (T3SS) is a clinically important virulence mechanism in Pseudomonas aeruginosa that secretes and translocates effector toxins into host cells, impeding the host's rapid innate immune response to infection. Inhibitors of T3SS may be useful as prophylactic or adjunctive therapeutic agents to augment the activity of antibiotics in P. aeruginosa infections, such as pneumonia and bacteremia. One such inhibitor, the phenoxyacetamide MBX 1641, exhibits very responsive structure-activity relationships, including striking stereoselectivity, in its inhibition of P. aeruginosa T3SS. These features suggest interaction with a specific, but unknown, protein target. Here, we identify the apparent molecular target by isolating inhibitor-resistant mutants and mapping the mutation sites by deep sequencing. Selection and sequencing of four independent mutants resistant to the phenoxyacetamide inhibitor MBX 2359 identified the T3SS gene pscF, encoding the needle apparatus, as the only locus of mutations common to all four strains. Transfer of the wild-type and mutated alleles of pscF, together with its chaperone and cochaperone genes pscE and pscG, to a ΔpscF P. aeruginosa strain demonstrated that each of the single-codon mutations in pscF is necessary and sufficient to provide secretion and translocation that is resistant to a variety of phenoxyacetamide inhibitor analogs but not to T3SS inhibitors with different chemical scaffolds. These results implicate the PscF needle protein as an apparent new molecular target for T3SS inhibitor discovery and suggest that three other chemically distinct T3SS inhibitors interact with one or more different targets or a different region of PscF.


Subject(s)
Carrier Proteins/metabolism , Pseudomonas aeruginosa/drug effects , Carrier Proteins/genetics , Immunoblotting , Intercellular Signaling Peptides and Proteins , Mutation , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/metabolism , Structure-Activity Relationship , Virulence/genetics
12.
J Virol ; 88(3): 1447-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24198411

ABSTRACT

Influenza viruses are a major public health threat worldwide, and options for antiviral therapy are limited by the emergence of drug-resistant virus strains. The influenza virus glycoprotein hemagglutinin (HA) plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a potential target for the development of anti-influenza drugs. Using pseudotype virus-based high-throughput screens, we have identified several new small molecules capable of inhibiting influenza virus entry. We prioritized two novel inhibitors, MBX2329 and MBX2546, with aminoalkyl phenol ether and sulfonamide scaffolds, respectively, that specifically inhibit HA-mediated viral entry. The two compounds (i) are potent (50% inhibitory concentration [IC50] of 0.3 to 5.9 µM); (ii) are selective (50% cytotoxicity concentration [CC(50)] of >100 µM), with selectivity index (SI) values of >20 to 200 for different influenza virus strains; (iii) inhibit a wide spectrum of influenza A viruses, which includes the 2009 pandemic influenza virus A/H1N1/2009, highly pathogenic avian influenza (HPAI) virus A/H5N1, and oseltamivir-resistant A/H1N1 strains; (iv) exhibit large volumes of synergy with oseltamivir (36 and 331 µM(2) % at 95% confidence); and (v) have chemically tractable structures. Mechanism-of-action studies suggest that both MBX2329 and MBX2546 bind to HA in a nonoverlapping manner. Additional results from HA-mediated hemolysis of chicken red blood cells (cRBCs), competition assays with monoclonal antibody (MAb) C179, and mutational analysis suggest that the compounds bind in the stem region of the HA trimer and inhibit HA-mediated fusion. Therefore, MBX2329 and MBX2546 represent new starting points for chemical optimization and have the potential to provide valuable future therapeutic options and research tools to study the HA-mediated entry process.


Subject(s)
Antiviral Agents/pharmacology , Hemagglutinins, Viral/metabolism , Influenza A virus/drug effects , Influenza in Birds/virology , Influenza, Human/virology , Poultry Diseases/virology , Small Molecule Libraries/pharmacology , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Chickens , Hemagglutinins, Viral/genetics , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/physiology , Influenza A virus/genetics , Influenza A virus/physiology , Small Molecule Libraries/chemistry
13.
Bioorg Med Chem ; 21(24): 7790-806, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24239389

ABSTRACT

The prevalence of drug-resistant bacteria in the clinic has propelled a concerted effort to find new classes of antibiotics that will circumvent current modes of resistance. We have previously described a set of bisamidine antibiotics that contains a core composed of two indoles and a central linker. The first compounds of the series, MBX 1066 and MBX 1090, have potent antibacterial properties against a wide range of Gram-positive and Gram-negative bacteria. We have conducted a systematic exploration of the amidine functionalities, the central linker, and substituents at the indole 3-position to determine the factors involved in potent antibacterial activity. Some of the newly synthesized compounds have even more potent and broad-spectrum activity than MBX 1066 and MBX 1090.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Furans/pharmacology , Imidazoles/pharmacology , Indoles/chemistry , Indoles/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Furans/chemical synthesis , Furans/chemistry , Imidazoles/chemical synthesis , Imidazoles/chemistry , Indoles/chemical synthesis , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
14.
Appl Environ Microbiol ; 79(19): 5830-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23872555

ABSTRACT

The bacterial SOS response is a well-characterized regulatory network encoded by most prokaryotic bacterial species and is involved in DNA repair. In addition to nucleic acid repair, the SOS response is involved in pathogenicity, stress-induced mutagenesis, and the emergence and dissemination of antibiotic resistance. Using high-throughput sequencing technology (SOLiD RNA-Seq), we analyzed the Burkholderia thailandensis global SOS response to the fluoroquinolone antibiotic, ciprofloxacin (CIP), and the DNA-damaging chemical, mitomycin C (MMC). We demonstrate that a B. thailandensis recA mutant (RU0643) is ∼4-fold more sensitive to CIP in contrast to the parental strain B. thailandensis DW503. Our RNA-Seq results show that CIP and MMC treatment (P < 0.01) resulted in the differential expression of 344 genes in B. thailandensis and 210 genes in RU0643. Several genes associated with the SOS response were induced and include lexA, uvrA, dnaE, dinB, recX, and recA. At the genome-wide level, we found an overall decrease in gene expression, especially for genes involved in amino acid and carbohydrate transport and metabolism, following both CIP and MMC exposure. Interestingly, we observed the upregulation of several genes involved in bacterial motility and enhanced transcription of a B. thailandensis genomic island encoding a Siphoviridae bacteriophage designated E264. Using B. thailandensis plaque assays and PCR with B. mallei ATCC 23344 as the host, we demonstrate that CIP and MMC exposure in B. thailandensis DW503 induces the transcription and translation of viable bacteriophage in a RecA-dependent manner. This is the first report of the SOS response in Burkholderia spp. to DNA-damaging agents. We have identified both common and unique adaptive responses of B. thailandensis to chemical stress and DNA damage.


Subject(s)
Burkholderia/physiology , SOS Response, Genetics , Transcriptome , Anti-Bacterial Agents/pharmacology , Bacteriophages/genetics , Burkholderia/drug effects , Burkholderia/genetics , Ciprofloxacin/pharmacology , Mitomycin/pharmacology , Mutagens , Protein Biosynthesis , Sequence Analysis, DNA , Siphoviridae/genetics , Transcription, Genetic
15.
Bioorg Med Chem Lett ; 23(12): 3481-6, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23664213

ABSTRACT

Benzobisthiazole derivatives were identified as novel helicase inhibitors through high throughput screening against purified Staphylococcus aureus (Sa) and Bacillus anthracis (Ba) replicative helicases. Chemical optimization has produced compound 59 with nanomolar potency against the DNA duplex strand unwinding activities of both B. anthracis and S. aureus helicases. Selectivity index (SI=CC50/IC50) values for 59 were greater than 500. Kinetic studies demonstrated that the benzobisthiazole-based bacterial helicase inhibitors act competitively with the DNA substrate. Therefore, benzobisthiazole helicase inhibitors represent a promising new scaffold for evaluation as antibacterial agents.


Subject(s)
Bacterial Proteins/genetics , Benzothiazoles/pharmacology , DNA Helicases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacillus anthracis/enzymology , Benzothiazoles/chemistry , DNA Helicases/metabolism , DNA, Bacterial/metabolism , Enzyme Inhibitors/chemistry , Microbial Sensitivity Tests/methods , Staphylococcus aureus/enzymology , Structure-Activity Relationship
17.
J Med Chem ; 55(24): 10896-908, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23231076

ABSTRACT

The increasing prevalence of drug-resistant bacterial infections demands the development of new antibacterials that are not subject to existing mechanisms of resistance. Previously, we described coumarin-based inhibitors of an underexploited bacterial target, namely the replicative helicase. Here we report the synthesis and evaluation of optimized coumarin-based inhibitors with 9-18-fold increased potency against Staphylococcus aureus (Sa) and Bacillus anthracis (Ba) helicases. Compounds 20 and 22 provided the best potency, with IC(50) values of 3 and 1 µM, respectively, against the DNA duplex strand-unwinding activities of both B. anthracis and S. aureus helicases without affecting the single strand DNA-stimulated ATPase activity. Selectivity index (SI = CC(50)/MIC) values against S. aureus and B. anthracis for compound 20 were 33 and 66 and for compound 22 were 20 and 40, respectively. In addition, compounds 20 and 22 demonstrated potent antibacterial activity against multiple ciprofloxacin-resistant MRSA strains, with MIC values ranging between 0.5 and 4.2 µg/mL.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Bacillus anthracis/enzymology , Coumarins/chemical synthesis , DnaB Helicases/antagonists & inhibitors , Staphylococcus aureus/enzymology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Ciprofloxacin/pharmacology , Coumarins/chemistry , Coumarins/pharmacology , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DnaB Helicases/chemistry , DnaB Helicases/metabolism , Drug Resistance, Bacterial , Enzyme Assays , Fluorescence Resonance Energy Transfer , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Structure-Activity Relationship
18.
Curr Opin Pharmacol ; 12(5): 535-44, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22841284

ABSTRACT

Several novel chemical classes of antibiotics are currently in human clinical studies. While most are narrow spectrum agents that inhibit unexploited targets, the susceptible pathogens are clinically important, including staphylococci, pseudomonads, and mycobacteria. Given the paucity of antibacterial agents consisting of novel chemical scaffolds that act on established targets, these new antibacterial scaffolds, which are active against new targets, represent an important advance in the battle against antibiotic resistance. Indeed, most of these compounds are unlikely to be subject to existing compound-based or target-based resistance mechanisms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Animals , Anti-Bacterial Agents/classification , Clinical Trials as Topic , Humans
19.
J Biomol Screen ; 16(7): 694-705, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21602485

ABSTRACT

The type II secretion (T2S) system in gram-negative bacteria comprises the Sec and Tat pathways for translocating proteins into the periplasm and an outer membrane secretin for transporting proteins into the extracellular space. To discover Sec/Tat/T2S pathway inhibitors as potential new therapeutics, the authors used a Pseudomonas aeruginosa bioluminescent reporter strain responsive to SecA depletion and inhibition to screen compound libraries and characterize the hits. The reporter strain placed a luxCDABE operon under regulation of a SecA depletion-responsive upregulated promoter in a secA deletion background complemented with an ectopic lac-regulated secA copy. Bioluminescence was indirectly proportional to the isopropyl-ß-D-thiogalactopyranoside concentration and stimulated by azide, a known SecA ATPase inhibitor. A total of 96 compounds (0.1% of 73,000) were detected as primary hits due to stimulation of luminescence with a z score ≥5. Direct secretion assays of the nine most potent hits, representing five chemical scaffolds, revealed that they do not inhibit SecA-mediated secretion of ß-lactamase into the periplasm but do inhibit T2S-mediated extracellular secretion of elastase with IC(50) values from 5 to 25 µM. In addition, seven of the nine compounds also inhibited the T2S-mediated extracellular secretion of phospholipase C by P. aeruginosa and protease activity by Burkholderia pseudomallei.


Subject(s)
Burkholderia pseudomallei/drug effects , Genes, Reporter/genetics , High-Throughput Screening Assays , Luminescent Proteins/analysis , Pseudomonas aeruginosa/drug effects , Adenosine Triphosphatases/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Burkholderia pseudomallei/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Luminescent Proteins/genetics , Membrane Transport Proteins/genetics , Pseudomonas aeruginosa/genetics , SEC Translocation Channels , SecA Proteins , Secretory Pathway/drug effects , Secretory Pathway/genetics , Small Molecule Libraries/pharmacology , Sodium Azide/pharmacology
20.
J Virol ; 85(7): 3106-19, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270170

ABSTRACT

Ebola virus (EBOV) causes severe hemorrhagic fever, for which therapeutic options are not available. Preventing the entry of EBOV into host cells is an attractive antiviral strategy, which has been validated for HIV by the FDA approval of the anti-HIV drug enfuvirtide. To identify inhibitors of EBOV entry, the EBOV envelope glycoprotein (EBOV-GP) gene was used to generate pseudotype viruses for screening of chemical libraries. A benzodiazepine derivative (compound 7) was identified from a high-throughput screen (HTS) of small-molecule compound libraries utilizing the pseudotype virus. Compound 7 was validated as an inhibitor of infectious EBOV and Marburg virus (MARV) in cell-based assays, with 50% inhibitory concentrations (IC(50)s) of 10 µM and 12 µM, respectively. Time-of-addition and binding studies suggested that compound 7 binds to EBOV-GP at an early stage during EBOV infection. Preliminary Schrödinger SiteMap calculations, using a published EBOV-GP crystal structure in its prefusion conformation, suggested a hydrophobic pocket at or near the GP1 and GP2 interface as a suitable site for compound 7 binding. This prediction was supported by mutational analysis implying that residues Asn69, Leu70, Leu184, Ile185, Leu186, Lys190, and Lys191 are critical for the binding of compound 7 and its analogs with EBOV-GP. We hypothesize that compound 7 binds to this hydrophobic pocket and as a consequence inhibits EBOV infection of cells, but the details of the mechanism remain to be determined. In summary, we have identified a novel series of benzodiazepine compounds that are suitable for optimization as potential inhibitors of filoviral infection.


Subject(s)
Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Ebolavirus/drug effects , Virus Internalization/drug effects , Antiviral Agents/chemistry , Benzodiazepines/isolation & purification , Benzodiazepines/pharmacology , DNA Mutational Analysis , Drug Evaluation, Preclinical/methods , Drug Resistance, Viral , Ebolavirus/physiology , High-Throughput Screening Assays/methods , Humans , Inhibitory Concentration 50 , Marburgvirus/drug effects , Models, Molecular , Mutation, Missense , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...