Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 14: 1353682, 2024.
Article in English | MEDLINE | ID: mdl-38590438

ABSTRACT

Introduction: Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is a disease endemic in many tropical countries globally. Clinical presentation is highly variable, ranging from asymptomatic to fatal septicemia, and thus the outcome of infection can depend on the host immune responses. The aims of this study were to firstly, characterize the macrophage immune response to B. pseudomallei and secondly, to determine whether the immune response was modified in the presence of novel inhibitors targeting the virulence factor, the macrophage infectivity potentiator (Mip) protein. We hypothesized that inhibition of Mip in B. pseudomallei would disarm the bacteria and result in a host beneficial immune response. Methods: Murine macrophage J774A.1 cells were infected with B. pseudomallei K96243 in the presence of small-molecule inhibitors targeting the Mip protein. RNA-sequencing was performed on infected cells four hours post-infection. Secreted cytokines and lactose dehydrogenase were measured in cell culture supernatants 24 hours post-infection. Viable, intracellular B. pseudomallei in macrophages were also enumerated 24 hours post-infection. Results: Global transcriptional profiling of macrophages infected with B. pseudomallei by RNA-seq demonstrated upregulation of immune-associated genes, in particular a significant enrichment of genes in the TNF signaling pathway. Treatment of B. pseudomallei-infected macrophages with the Mip inhibitor, AN_CH_37 resulted in a 5.3-fold reduction of il1b when compared to cells treated with DMSO, which the inhibitors were solubilized in. A statistically significant reduction in IL-1ß levels in culture supernatants was seen 24 hours post-infection with AN_CH_37, as well as other pro-inflammatory cytokines, namely IL-6 and TNF-α. Treatment with AN_CH_37 also reduced the survival of B. pseudomallei in macrophages after 24 hours which was accompanied by a significant reduction in B. pseudomallei-induced cytotoxicity as determined by lactate dehydrogenase release. Discussion: These data highlight the potential to utilize Mip inhibitors in reducing potentially harmful pro-inflammatory responses resulting from B. pseudomallei infection in macrophages. This could be of significance since overstimulation of pro-inflammatory responses can result in immunopathology, tissue damage and septic shock.


Subject(s)
Burkholderia pseudomallei , Melioidosis , Animals , Mice , Burkholderia pseudomallei/metabolism , Melioidosis/microbiology , Macrophages/microbiology , Cytokines/metabolism , Signal Transduction
2.
Arch Pharm (Weinheim) ; : e2400032, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687906

ABSTRACT

Due to increasing antibiotic resistance, the development of anti-infectives with new mechanisms of action is crucial. Virulence factors such as the "macrophage infectivity potentiator" (Mip) protein, which catalyzes the folding of proline-containing proteins by means of their cis-trans isomerase (PPIase) activity, have come into focus as a potential new target. Since the inhibition of Mip by small molecules has been shown to lead to reduced virulence and survival in vitro, especially of Gram-negative bacteria such as Burkholderia pseudomallei (Bp), Neisseria meningitidis (Nm), and Neisseria gonorrhoeae (Ng), or Coxiella burnetii (Cb), among many others, a library of Mip inhibitors was developed. As drug metabolism has a significant impact on the overall therapeutic outcome, this report describes the biotransformation of the most potent Mip inhibitors. Therefore, the anti-infectives were treated using human liver microsomes in vitro. Liquid chromatography with tandem mass spectrometry (LC/MS-MS) methods were applied to identify the metabolites and quantify the metabolic degradation of the hit compounds. Active metabolites, N-oxides, were found, leading to new opportunities for further drug development.

3.
Environ Microbiol ; 26(1): e16571, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38178319

ABSTRACT

Burkholderia pseudomallei is a saprophytic Gram-negative bacillus that can cause the disease melioidosis. Although B. pseudomallei is a recognised member of terrestrial soil microbiomes, little is known about its contribution to the saprophytic degradation of polysaccharides within its niche. For example, while chitin is predicted to be abundant within terrestrial soils the chitinolytic capacity of B. pseudomallei is yet to be defined. This study identifies and characterises a putative glycoside hydrolase, bpsl0500, which is expressed by B. pseudomallei K96243. Recombinant BPSL0500 was found to exhibit activity against substrate analogues and GlcNAc disaccharides relevant to chitinolytic N-acetyl-ß-d-hexosaminidases. In B. pseudomallei, bpsl0500 was found to be essential for both N-acetyl-ß-d-hexosaminidase activity and chitooligosaccharide metabolism. Furthermore, bpsl0500 was also observed to significantly affect biofilm deposition. These observations led to the identification of BPSL0500 activity against model disaccharide linkages that are present in biofilm exopolysaccharides, a feature that has not yet been described for chitinolytic enzymes. The results in this study indicate that chitinolytic N-acetyl-ß-d-hexosaminidases like bpsl0500 may facilitate biofilm disruption as well as chitin assimilation, providing dual functionality for saprophytic bacteria such as B. pseudomallei within the competitive soil microbiome.


Subject(s)
Burkholderia pseudomallei , Chitosan , Melioidosis , Oligosaccharides , Humans , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/metabolism , Soil , Biofilms , Chitin/metabolism , Hexosaminidases/genetics , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism , Melioidosis/microbiology
4.
Antibiotics (Basel) ; 12(9)2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37760679

ABSTRACT

Antibiotic resistance caused by multidrug-resistant (MDR) bacteria is a major challenge to global public health. Polymyxins are increasingly being used as last-in-line antibiotics to treat MDR Gram-negative bacterial infections, but resistance development renders them ineffective for empirical therapy. The main mechanism that bacteria use to defend against polymyxins is to modify the lipid A headgroups of the outer membrane by adding phosphoethanolamine (PEA) moieties. In addition to lipid A modifying PEA transferases, Gram-negative bacteria possess PEA transferases that decorate proteins and glycans. This review provides a comprehensive overview of the function, structure, and mechanism of action of PEA transferases identified in pathogenic Gram-negative bacteria. It also summarizes the current drug development progress targeting this enzyme family, which could reverse antibiotic resistance to polymyxins to restore their utility in empiric therapy.

5.
PLoS Pathog ; 19(7): e1011491, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37399210

ABSTRACT

Coxiella burnetii is a Gram-negative intracellular pathogen that causes the debilitating disease Q fever, which affects both animals and humans. The only available human vaccine, Q-Vax, is effective but has a high risk of severe adverse reactions, limiting its use as a countermeasure to contain outbreaks. Therefore, it is essential to identify new drug targets to treat this infection. Macrophage infectivity potentiator (Mip) proteins catalyse the folding of proline-containing proteins through their peptidyl prolyl cis-trans isomerase (PPIase) activity and have been shown to play an important role in the virulence of several pathogenic bacteria. To date the role of the Mip protein in C. burnetii pathogenesis has not been investigated. This study demonstrates that CbMip is likely to be an essential protein in C. burnetii. The pipecolic acid derived compounds, SF235 and AN296, which have shown utility in targeting other Mip proteins from pathogenic bacteria, demonstrate inhibitory activities against CbMip. These compounds were found to significantly inhibit intracellular replication of C. burnetii in both HeLa and THP-1 cells. Furthermore, SF235 and AN296 were also found to exhibit antibiotic properties against both the virulent (Phase I) and avirulent (Phase II) forms of C. burnetii Nine Mile Strain in axenic culture. Comparative proteomics, in the presence of AN296, revealed alterations in stress responses with H2O2 sensitivity assays validating that Mip inhibition increases the sensitivity of C. burnetii to oxidative stress. In addition, SF235 and AN296 were effective in vivo and significantly improved the survival of Galleria mellonella infected with C. burnetii. These results suggest that unlike in other bacteria, Mip in C. burnetii is required for replication and that the development of more potent inhibitors against CbMip is warranted and offer potential as novel therapeutics against this pathogen.


Subject(s)
Coxiella burnetii , Q Fever , Animals , Humans , Peptidylprolyl Isomerase/metabolism , Bacterial Proteins/metabolism , Hydrogen Peroxide/metabolism , Bacteria/metabolism , Macrophages/metabolism
6.
J Med Chem ; 66(13): 8876-8895, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37389560

ABSTRACT

The macrophage infectivity potentiator (Mip) protein is a promising target for developing new drugs to combat antimicrobial resistance. New rapamycin-derived Mip inhibitors have been designed that may be able to combine two binding modes to inhibit the Mip protein of Burkholderia pseudomallei (BpMip). These novel compounds are characterized by an additional substituent in the middle chain linking the lateral pyridine to the pipecoline moiety, constituting different stereoisomers. These compounds demonstrated high affinity for the BpMip protein in the nanomolar range and high anti-enzymatic activity and ultimately resulted in significantly reduced cytotoxicity of B. pseudomallei in macrophages. They also displayed strong anti-enzymatic activity against the Mip proteins of Neisseria meningitidis and Neisseria gonorrhoeae and substantially improved the ability of macrophages to kill the bacteria. Hence, the new Mip inhibitors are promising, non-cytotoxic candidates for further testing against a broad spectrum of pathogens and infectious diseases.


Subject(s)
Burkholderia pseudomallei , Neisseria meningitidis , Bacterial Proteins , Burkholderia pseudomallei/metabolism , Macrophages/metabolism , Neisseria gonorrhoeae/metabolism , Neisseria meningitidis/metabolism , Structure-Activity Relationship
7.
Virulence ; 13(1): 1945-1965, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36271712

ABSTRACT

The soil saprophyte, Burkholderia pseudomallei, is the causative agent of melioidosis, a disease endemic in South East Asia and northern Australia. Exposure to B. pseudomallei by either inhalation or inoculation can lead to severe disease. B. pseudomallei rapidly shifts from an environmental organism to an aggressive intracellular pathogen capable of rapidly spreading around the body. The expression of multiple virulence factors at every stage of intracellular infection allows for rapid progression of infection. Following invasion or phagocytosis, B. pseudomallei resists host-cell killing mechanisms in the phagosome, followed by escape using the type III secretion system. Several secreted virulence factors manipulate the host cell, while bacterial cells undergo a shift in energy metabolism allowing for overwhelming intracellular replication. Polymerisation of host cell actin into "actin tails" propels B. pseudomallei to the membranes of host cells where the type VI secretion system fuses host cells into multinucleated giant cells (MNGCs) to facilitate cell-to-cell dissemination. This review describes the various mechanisms used by B. pseudomallei to survive within cells.


Subject(s)
Burkholderia pseudomallei , Melioidosis , Humans , Virulence , Actins/metabolism , Melioidosis/microbiology , Virulence Factors/metabolism
8.
J Antimicrob Chemother ; 77(9): 2441-2447, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35770844

ABSTRACT

OBJECTIVES: Neisseria gonorrhoeae is an exclusively human pathogen that commonly infects the urogenital tract resulting in gonorrhoea. Empirical treatment of gonorrhoea with antibiotics has led to multidrug resistance and the need for new therapeutics. Inactivation of lipooligosaccharide phosphoethanolamine transferase A (EptA), which attaches phosphoethanolamine to lipid A, results in attenuation of the pathogen in infection models. Small molecules that inhibit EptA are predicted to enhance natural clearance of gonococci via the human innate immune response. METHODS: A library of small-fragment compounds was tested for the ability to enhance susceptibility of the reference strain N. gonorrhoeae FA1090 to polymyxin B. The effect of these compounds on lipid A synthesis and viability in models of infection were tested. RESULTS: Three compounds, 135, 136 and 137, enhanced susceptibility of strain FA1090 to polymyxin B by 4-fold. Pre-treatment of bacterial cells with all three compounds resulted in enhanced killing by macrophages. Only lipid A from bacterial cells exposed to compound 137 showed a 17% reduction in the level of decoration of lipid A with phosphoethanolamine by MALDI-TOF MS analysis and reduced stimulation of cytokine responses in THP-1 cells. Binding of 137 occurred with higher affinity to purified EptA than the starting material, as determined by 1D saturation transfer difference NMR. Treatment of eight MDR strains with 137 increased susceptibility to polymyxin B in all cases. CONCLUSIONS: Small molecules have been designed that bind to EptA, inhibit addition of phosphoethanolamine to lipid A and can sensitize N. gonorrhoeae to killing by macrophages.


Subject(s)
Gonorrhea , Neisseria gonorrhoeae , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Peptides , Drug Resistance, Bacterial , Ethanolaminephosphotransferase/metabolism , Ethanolamines , Gonorrhea/drug therapy , Humans , Lipid A/chemistry , Microbial Sensitivity Tests , Polymyxin B/pharmacology
9.
J Antimicrob Chemother ; 77(6): 1625-1634, 2022 05 29.
Article in English | MEDLINE | ID: mdl-35245364

ABSTRACT

BACKGROUND: The macrophage infectivity potentiator (Mip) protein, which belongs to the immunophilin superfamily, is a peptidyl-prolyl cis/trans isomerase (PPIase) enzyme. Mip has been shown to be important for virulence in a wide range of pathogenic microorganisms. It has previously been demonstrated that small-molecule compounds designed to target Mip from the Gram-negative bacterium Burkholderia pseudomallei bind at the site of enzymatic activity of the protein, inhibiting the in vitro activity of Mip. OBJECTIVES: In this study, co-crystallography experiments with recombinant B. pseudomallei Mip (BpMip) protein and Mip inhibitors, biochemical analysis and computational modelling were used to predict the efficacy of lead compounds for broad-spectrum activity against other pathogens. METHODS: Binding activity of three lead compounds targeting BpMip was verified using surface plasmon resonance spectroscopy. The determination of crystal structures of BpMip in complex with these compounds, together with molecular modelling and in vitro assays, was used to determine whether the compounds have broad-spectrum antimicrobial activity against pathogens. RESULTS: Of the three lead small-molecule compounds, two were effective in inhibiting the PPIase activity of Mip proteins from Neisseria meningitidis, Klebsiella pneumoniae and Leishmania major. The compounds also reduced the intracellular burden of these pathogens using in vitro cell infection assays. CONCLUSIONS: These results indicate that Mip is a novel antivirulence target that can be inhibited using small-molecule compounds that prove to be promising broad-spectrum drug candidates in vitro. Further optimization of compounds is required for in vivo evaluation and future clinical applications.


Subject(s)
Bacterial Proteins , Gram-Negative Bacteria , Leishmania major , Peptidylprolyl Isomerase , Protozoan Proteins , Bacterial Proteins/antagonists & inhibitors , Gram-Negative Bacteria/drug effects , Leishmania major/drug effects , Macrophages/metabolism , Neisseria meningitidis , Peptidylprolyl Isomerase/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Recombinant Proteins
10.
Antibiotics (Basel) ; 10(2)2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33494538

ABSTRACT

While antimicrobial resistance (AMR) is seen in both Neisseria gonorrhoeae and Neisseria meningitidis, the former has become resistant to commonly available over-the-counter antibiotic treatments. It is imperative then to develop new therapies that combat current AMR isolates whilst also circumventing the pathways leading to the development of AMR. This review highlights the growing research interest in developing anti-virulence therapies (AVTs) which are directed towards inhibiting virulence factors to prevent infection. By targeting virulence factors that are not essential for gonococcal survival, it is hypothesized that this will impart a smaller selective pressure for the emergence of resistance in the pathogen and in the microbiome, thus avoiding AMR development to the anti-infective. This review summates the current basis of numerous anti-virulence strategies being explored for N. gonorrhoeae.

11.
J Med Chem ; 63(22): 13355-13388, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32786507

ABSTRACT

Infectious diseases are a major cause of morbidity and mortality worldwide, exacerbated by increasing antibiotic resistance in many bacterial species. The development of drugs with new modes of action is essential. A leading strategy is antivirulence, with the aim to target bacterial proteins that are important in disease causation and progression but do not affect growth, resulting in reduced selective pressure for resistance. Immunophilins, a superfamily of peptidyl-prolyl cis-trans isomerase (PPIase) enzymes have been shown to be important for virulence in a broad-spectrum of pathogenic bacteria. This Perspective will provide an overview of the recent advances made in understanding the role of each immunophilin family, cyclophilins, FK506 binding proteins (FKBPs), and parvulins in bacteria. Inhibitor design and medicinal chemistry strategies for development of novel drugs against bacterial FKBPs will be discussed. Furthermore, drugs against human cyclophilins and parvulins will be reviewed in their current indication as antiviral and anticancer therapies.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Drug Delivery Systems/methods , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Protein Folding/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Bacterial Infections/drug therapy , Bacterial Infections/metabolism , Bacterial Proteins/chemistry , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/chemistry , Gram-Positive Bacteria/drug effects , Humans , Protein Structure, Secondary , Protein Structure, Tertiary
12.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31331957

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, a disease endemic to Southeast Asia and northern Australia. Mortality rates in these areas are high even with antimicrobial treatment, and there are few options for effective therapy. Therefore, there is a need to identify antibacterial targets for the development of novel treatments. Cyclophilins are a family of highly conserved enzymes important in multiple cellular processes. Cyclophilins catalyze the cis-trans isomerization of xaa-proline bonds, a rate-limiting step in protein folding which has been shown to be important for bacterial virulence. B. pseudomallei carries a putative cyclophilin B gene, ppiB, the role of which was investigated. A B. pseudomalleiΔppiB (BpsΔppiB) mutant strain demonstrates impaired biofilm formation and reduced motility. Macrophage invasion and survival assays showed that although the BpsΔppiB strain retained the ability to infect macrophages, it had reduced survival and lacked the ability to spread cell to cell, indicating ppiB is essential for B. pseudomallei virulence. This is reflected in the BALB/c mouse infection model, demonstrating the requirement of ppiB for in vivo disease dissemination and progression. Proteomic analysis demonstrates that the loss of PpiB leads to pleiotropic effects, supporting the role of PpiB in maintaining proteome homeostasis. The loss of PpiB leads to decreased abundance of multiple virulence determinants, including flagellar machinery and alterations in type VI secretion system proteins. In addition, the loss of ppiB leads to increased sensitivity toward multiple antibiotics, including meropenem and doxycycline, highlighting ppiB inhibition as a promising antivirulence target to both treat B. pseudomallei infections and increase antibiotic efficacy.


Subject(s)
Bacterial Proteins/genetics , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/pathogenicity , Cyclophilins/genetics , Melioidosis/microbiology , Proteome/genetics , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Burkholderia pseudomallei/drug effects , Burkholderia pseudomallei/metabolism , Cell Line , Cyclophilins/deficiency , Female , Gene Deletion , Gene Expression , Homeostasis/genetics , Macrophages/microbiology , Melioidosis/drug therapy , Melioidosis/mortality , Melioidosis/pathology , Mice , Mice, Inbred BALB C , Microbial Viability/drug effects , Proteome/classification , Proteome/metabolism , Survival Analysis , Virulence
13.
Methods Mol Biol ; 1969: 123-133, 2019.
Article in English | MEDLINE | ID: mdl-30877674

ABSTRACT

Neisseria meningitidis and Neisseria gonorrhoeae are obligate pathogens of the human host. Due to their adaptation to the human host, many factors required for infection are specialized for the human host to the point that natural infection processes are difficult to replicate in animal models. Immortalized human cell lines have been used to identify the host factors necessary for successful colonization of human mucosal surfaces. One such model is the Detroit 562 pharyngeal immortalized cell monolayer model which is used to measure the rate of attachment to and invasion of N. meningitidis and N. gonorrhoeae into epithelial cells. The methodology of this assay, as well as the maintenance of Detroit 562 cells necessary for the experiment, will be described.


Subject(s)
Epithelial Cells/microbiology , Gonorrhea/microbiology , Meningococcal Infections/microbiology , Models, Biological , Neisseria gonorrhoeae/pathogenicity , Neisseria meningitidis/pathogenicity , Pharynx/microbiology , Cells, Cultured , Humans
14.
Curr Opin Struct Biol ; 53: 140-150, 2018 12.
Article in English | MEDLINE | ID: mdl-30223251

ABSTRACT

Society has benefitted greatly from the use of antibiotics. Unfortunately, the misuse of these valuable molecules has resulted in increased levels of antibiotic resistance, a major global and public health issue. This resistance and the reliance on a small number of biological targets for the development of antibiotics emphasizes the need for new targets. A critical aspect guiding the development of new antimicrobials through a rational structure-guided approach is to understand the molecular structures of specific biological targets of interest. Here we give an overview of the structures of bacterial virulence enzyme targets involved in protein folding, peptidoglycan biosynthesis and cell wall modification. These include enzymes of the thiol-disulphide oxidoreductase pathway (DSB enzymes), peptidyl-proly cis/trans isomerases (Mips), enzymes from the Mur pathway and enzymes involved in lipopolysaccharide modification (EptA and ArnT). We also present progress towards inhibitor design of these targets for the development of novel anti-virulence therapeutic agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria , Bacterial Proteins , Virulence Factors , Bacteria/enzymology , Bacteria/metabolism , Bacterial Infections/drug therapy , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Drug Design , Hexosyltransferases/metabolism , Humans , Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/metabolism , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Disulfide Reductase (Glutathione)/chemistry , Protein Disulfide Reductase (Glutathione)/metabolism , Protein Domains , Protein Structure, Quaternary , Virulence Factors/chemistry , Virulence Factors/metabolism
15.
J R Soc Interface ; 15(146)2018 09 12.
Article in English | MEDLINE | ID: mdl-30209043

ABSTRACT

Current and reoccurring viral epidemic outbreaks such as those caused by the Zika virus illustrate the need for rapid development of antivirals. Such development would be facilitated by computational approaches that can provide experimentally testable predictions for possible antiviral strategies. To this end, we focus here on the fact that viruses are directly dependent on their host metabolism for reproduction. We develop a stoichiometric, genome-scale metabolic model that integrates human macrophage cell metabolism with the biochemical demands arising from virus production and use it to determine the virus impact on host metabolism and vice versa. While this approach applies to any host-virus pair, we first apply it to currently epidemic viruses Chikungunya, Dengue and Zika in this study. We find that each of these viruses causes specific alterations in the host metabolic flux towards fulfilling their biochemical demands as predicted by their genome and capsid structure. Subsequent analysis of this integrated model allows us to predict a set of host reactions, which, when constrained, inhibit virus production. We show that this prediction recovers known targets of existing antiviral drugs, specifically those targeting nucleotide production, while highlighting a set of hitherto unexplored reactions involving both amino acid and nucleotide metabolic pathways, with either broad or virus-specific antiviral potential. Thus, this computational approach allows rapid generation of experimentally testable hypotheses for novel antiviral targets within a host.


Subject(s)
Chikungunya virus/pathogenicity , Dengue Virus/pathogenicity , Host-Pathogen Interactions , Macrophages/metabolism , Zika Virus Infection/drug therapy , Zika Virus/pathogenicity , Adenosine Triphosphate/metabolism , Algorithms , Antiviral Agents/therapeutic use , Biomass , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Dengue/drug therapy , Dengue Virus/drug effects , Genome, Viral , Humans , Macrophages/virology , Models, Theoretical , Virus Replication , Zika Virus/drug effects
16.
Infect Immun ; 86(5)2018 05.
Article in English | MEDLINE | ID: mdl-29440370

ABSTRACT

The naturally antibiotic-resistant bacterium Burkholderia pseudomallei is the causative agent of melioidosis, a disease with stubbornly high mortality and a complex, protracted treatment regimen. The worldwide incidence of melioidosis is likely grossly underreported, though it is known to be highly endemic in northern Australia and Southeast Asia. Bacterial disulfide bond (DSB) proteins catalyze the oxidative folding and isomerization of disulfide bonds in substrate proteins. In the present study, we demonstrate that B. pseudomallei membrane protein disulfide bond protein B (BpsDsbB) forms a functional redox relay with the previously characterized virulence mediator B. pseudomallei disulfide bond protein A (BpsDsbA). Genomic analysis of diverse B. pseudomallei clinical isolates demonstrated that dsbB is a highly conserved core gene. Critically, we show that DsbB is required for virulence in B. pseudomallei A panel of B. pseudomalleidsbB deletion strains (K96243, 576, MSHR2511, MSHR0305b, and MSHR5858) were phenotypically diverse according to the results of in vitro assays that assess hallmarks of virulence. Irrespective of their in vitro virulence phenotypes, two deletion strains were attenuated in a BALB/c mouse model of infection. A crystal structure of a DsbB-derived peptide complexed with BpsDsbA provides the first molecular characterization of their interaction. This work contributes to our broader understanding of DSB redox biology and will support the design of antimicrobial drugs active against this important family of bacterial virulence targets.


Subject(s)
Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/pathogenicity , Melioidosis/pathology , Membrane Proteins/immunology , Mice, Inbred BALB C/immunology , Oxidoreductases/immunology , Virulence/genetics , Animals , Australia , Burkholderia pseudomallei/immunology , Disease Models, Animal , Melioidosis/genetics , Melioidosis/microbiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Oxidoreductases/genetics , Oxidoreductases/metabolism , Virulence/immunology
17.
Nat Commun ; 8(1): 1206, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089518

ABSTRACT

Erythromycin, avermectin and rapamycin are clinically useful polyketide natural products produced on modular polyketide synthase multienzymes by an assembly-line process in which each module of enzymes in turn specifies attachment of a particular chemical unit. Although polyketide synthase encoding genes have been successfully engineered to produce novel analogues, the process can be relatively slow, inefficient, and frequently low-yielding. We now describe a method for rapidly recombining polyketide synthase gene clusters to replace, add or remove modules that, with high frequency, generates diverse and highly productive assembly lines. The method is exemplified in the rapamycin biosynthetic gene cluster where, in a single experiment, multiple strains were isolated producing new members of a rapamycin-related family of polyketides. The process mimics, but significantly accelerates, a plausible mechanism of natural evolution for modular polyketide synthases. Detailed sequence analysis of the recombinant genes provides unique insight into the design principles for constructing useful synthetic assembly-line multienzymes.


Subject(s)
Biosynthetic Pathways/genetics , Evolution, Molecular , Genetic Variation , Multigene Family , Bioengineering , Polyketide Synthases/genetics , Sirolimus/chemistry , Sirolimus/metabolism
18.
Anal Biochem ; 536: 59-68, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28803887

ABSTRACT

Prolyl-peptidyl isomerases (PPIases) are enzymes that are found in all living organisms. They form an essential part of the cellular protein folding homeostasis machinery. PPIases are associated with many important human diseases, e.g. cardiovascular disease, cancer and Alzheimer's. The development of novel PPIase inhibitors has been limited by the lack of a rapid, laboratory-based assay for these enzymes, as their substrates and products are challenging to distinguish. A well described continuous assay, coupled with the hydrolysis of a peptide by chymotrypsin is highly effective, but comparatively slow. To address this, we developed an improved version of the traditional assay using a temperature controlled plate reader. This assay allows semi-automated medium throughput assays in an academic laboratory for 84 samples per day. The assay shows lower errors, with an average Z' of 0.72. We further developed the assay using a fluorogenic peptide-based FRET probe. This provides an extremely sensitive PPIase assay using substrate at 200 nM, which approaches single turnover conditions. The fluorescent probe achieves an excellent quenching efficiency of 98.6%, and initial experiments showed acceptable Z' of 0.31 and 0.30 for cyclophilin A and hFKBP12 respectively. The assays provide an improved toolset for the quantitative, biochemical analysis of PPIases.


Subject(s)
Enzyme Assays/methods , Peptidylprolyl Isomerase/analysis , Peptidylprolyl Isomerase/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Molecular Conformation , Substrate Specificity , Temperature
19.
BMC Microbiol ; 17(1): 163, 2017 Jul 21.
Article in English | MEDLINE | ID: mdl-28732479

ABSTRACT

BACKGROUND: The World Health Organization has categorized plague as a re-emerging disease and the potential for Yersinia pestis to also be used as a bioweapon makes the identification of new drug targets against this pathogen a priority. Environmental temperature is a key signal which regulates virulence of the bacterium. The bacterium normally grows outside the human host at 28 °C. Therefore, understanding the mechanisms that the bacterium used to adapt to a mammalian host at 37 °C is central to the development of vaccines or drugs for the prevention or treatment of human disease. RESULTS: Using a library of over 1 million Y. pestis CO92 random mutants and transposon-directed insertion site sequencing, we identified 530 essential genes when the bacteria were cultured at 28 °C. When the library of mutants was subsequently cultured at 37 °C we identified 19 genes that were essential at 37 °C but not at 28 °C, including genes which encode proteins that play a role in enabling functioning of the type III secretion and in DNA replication and maintenance. Using genome-scale metabolic network reconstruction we showed that growth conditions profoundly influence the physiology of the bacterium, and by combining computational and experimental approaches we were able to identify 54 genes that are essential under a broad range of conditions. CONCLUSIONS: Using an integrated computational-experimental approach we identify genes which are required for growth at 37 °C and under a broad range of environments may be the best targets for the development of new interventions to prevent or treat plague in humans.


Subject(s)
Bacterial Proteins/genetics , Computational Biology/methods , Genes, Essential , Plague/microbiology , Yersinia pestis/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Mutation , Yersinia pestis/growth & development , Yersinia pestis/metabolism
20.
Sci Rep ; 7: 41923, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165493

ABSTRACT

Massively parallel sequencing technology coupled with saturation mutagenesis has provided new and global insights into gene functions and roles. At a simplistic level, the frequency of mutations within genes can indicate the degree of essentiality. However, this approach neglects to take account of the positional significance of mutations - the function of a gene is less likely to be disrupted by a mutation close to the distal ends. Therefore, a systematic bioinformatics approach to improve the reliability of essential gene identification is desirable. We report here a parametric model which introduces a novel mutation feature together with a noise trimming approach to predict the biological significance of Tn5 mutations. We show improved performance of essential gene prediction in the bacterium Yersinia pestis, the causative agent of plague. This method would have broad applicability to other organisms and to the identification of genes which are essential for competitiveness or survival under a broad range of stresses.


Subject(s)
Bacterial Proteins/genetics , Genes, Essential , High-Throughput Nucleotide Sequencing/methods , Mutation , Plague/microbiology , Yersinia pestis/growth & development , Yersinia pestis/genetics , Computational Biology , Gene Expression Regulation, Bacterial , Genome, Bacterial , Phenotype , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...