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











Publication year range
1.
J Ethnopharmacol ; 267: 113533, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33137433

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Secondary metabolites play a critical role in plant defense against disease and are of great importance to ethnomedicine. Bacterial efflux pumps are active transport proteins that bacterial cells use to protect themselves against multiple toxic compounds, including many antimicrobials. Efflux pump inhibitors from plants can block these efflux pumps, increasing the potency of antimicrobial compounds. This study demonstrates that efflux pump inhibition against the Gram-positive bacterial pathogen Staphylococcus aureus is widespread in extracts prepared from individual species throughout the land plant lineage. It therefore suggests a general mechanism by which plants used by indigenous species may be effective as a topical treatment for some bacterial infections. AIM OF THE STUDY: The goal of this research was to evaluate the distribution of efflux pump inhibitors in nine plant extracts with an ethnobotanical use suggestive of an antimicrobial function for the presence of efflux pump inhibitory activity against Staphylococcus aureus. MATERIALS AND METHODS: Plants were collected, dried, extracted, and vouchers submitted to the Herbarium of the University of North Carolina Chapel Hill (NCU). The extracts were analyzed by quantitative mass spectrometry (UPLC-MS) to determine the presence and concentration of flavonoids with known efflux pump inhibitory activity. A mass spectrometry-based assay was employed to measure efflux pump inhibition for all extracts against Staphylococcus aureus. The assay relies on UPLC-MS measurement of changes in ethidium concentration in the spent culture broth when extracts are incubated with bacteria. RESULTS: Eight of these nine plant extracts inhibited toxic compound efflux at concentrations below the MIC (minimum inhibitory concentration) value for the same extract. The most active extracts were those prepared from Osmunda claytoniana L. and Pinus strobes L., which both demonstrated IC50 values for efflux inhibition of 19 ppm. CONCLUSIONS: Our findings indicate that efflux pump inhibitors active against Staphylococcus aureus are common in land plants. By extension, this activity is likely to be important in many plant-derived antimicrobial extracts, including those used in traditional medicine, and evaluation of efflux pump inhibition may often be valuable when studying natural product efficacy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Secretion Systems/drug effects , Membrane Transport Modulators/pharmacology , Membrane Transport Proteins/drug effects , Plants, Medicinal , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/isolation & purification , Bacterial Proteins/metabolism , Membrane Transport Modulators/isolation & purification , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Phytotherapy , Plants, Medicinal/chemistry , Plants, Medicinal/classification , Staphylococcus aureus/metabolism
2.
Sci Rep ; 10(1): 21109, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273542

ABSTRACT

Porphyromonas gingivalis, a periodontal pathogen, translocates many virulence factors including the cysteine proteases referred to as gingipains to the cell surface via the type IX secretion system (T9SS). Expression of the T9SS component proteins is regulated by the tandem signaling of the PorXY two-component system and the ECF sigma factor SigP. However, the details of this regulatory pathway are still unknown. We found that one of the T9SS conserved C-terminal domain-containing proteins, PGN_0123, which we have designated PorA, is involved in regulating expression of genes encoding T9SS structural proteins and that PorA can be translocated onto the cell surface without the T9SS translocation machinery. X-ray crystallography revealed that PorA has a domain similar to the mannose-binding domain of Escherichia coli FimH, the tip protein of Type 1 pilus. Mutations in the cytoplasmic domain of the sensor kinase PorY conferred phenotypic recovery on the ΔporA mutant. The SigP sigma factor, which is activated by the PorXY two-component system, markedly decreased in the ΔporA mutant. These results strongly support a potential role for PorA in relaying a signal from the cell surface to the PorXY-SigP signaling pathway.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Secretion Systems , Porphyromonas gingivalis/metabolism , Signal Transduction , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Secretion Systems/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Gain of Function Mutation , Gingipain Cysteine Endopeptidases/pharmacology , Hemagglutination/drug effects , Lipopolysaccharides/metabolism , Models, Biological , Peptide Mapping , Pigmentation/drug effects , Porphyromonas gingivalis/drug effects , Protein Domains , Protein Transport/drug effects , Signal Transduction/drug effects
3.
Mol Plant Pathol ; 20(5): 716-730, 2019 05.
Article in English | MEDLINE | ID: mdl-30912619

ABSTRACT

In a number of compatible plant-bacterium interactions, a rise in apoplastic Ca2+ levels is observed, suggesting that Ca2+ represents an important environmental clue, as reported for bacteria infecting mammalians. We demonstrate that Ca2+ entry in Pseudomonas savastanoi pv. savastanoi (Psav) strain DAPP-PG 722 is mediated by a Na+ /Ca2+ exchanger critical for virulence. Using the fluorescent Ca2+ probe Fura 2-AM, we demonstrate that Ca2+ enters Psav cells foremost when they experience low levels of energy, a situation mimicking the apoplastic fluid. In fact, Ca2+ entry was suppressed in the presence of high concentrations of glucose, fructose, sucrose or adenosine triphosphate (ATP). Since Ca2+ entry was inhibited by nifedipine and LiCl, we conclude that the channel for Ca2+ entry is a Na+ /Ca2+ exchanger. In silico analysis of the Psav DAPP-PG 722 genome revealed the presence of a single gene coding for a Na+ /Ca2+ exchanger (cneA), which is a widely conserved and ancestral gene within the P. syringae complex based on gene phylogeny. Mutation of cneA compromised not only Ca2+ entry, but also compromised the Hypersensitive response (HR) in tobacco leaves and blocked the ability to induce knots in olive stems. The expression of both pathogenicity (hrpL, hrpA and iaaM) and virulence (ptz) genes was reduced in this Psav-cneA mutant. Complementation of the Psav-cneA mutation restored both Ca2+ entry and pathogenicity in olive plants, but failed to restore the HR in tobacco leaves. In conclusion, Ca2+ entry acts as a 'host signal' that allows and promotes Psav pathogenicity on olive plants.


Subject(s)
Bacterial Proteins/metabolism , Olea/microbiology , Pseudomonas/pathogenicity , Sodium-Calcium Exchanger/metabolism , Bacterial Proteins/genetics , Bacterial Secretion Systems/drug effects , Biofilms/growth & development , Calcium/metabolism , Chromosomes, Bacterial/genetics , Cytosol/metabolism , Gene Expression Regulation, Bacterial/drug effects , Indoleacetic Acids/pharmacology , Mutation/genetics , Olea/drug effects , Phenotype , Plant Diseases/microbiology , Plant Growth Regulators/metabolism , Pseudomonas/drug effects , Nicotiana/microbiology , Virulence/drug effects
4.
EcoSal Plus ; 8(2)2019 03.
Article in English | MEDLINE | ID: mdl-30873935

ABSTRACT

The chaperone-usher (CU) pathway is a conserved secretion system dedicated to the assembly of a superfamily of virulence-associated surface structures by a wide range of Gram-negative bacteria. Pilus biogenesis by the CU pathway requires two specialized assembly components: a dedicated periplasmic chaperone and an integral outer membrane assembly and secretion platform termed the usher. The CU pathway assembles a variety of surface fibers, ranging from thin, flexible filaments to rigid, rod-like organelles. Pili typically act as adhesins and function as virulence factors that mediate contact with host cells and colonization of host tissues. Pilus-mediated adhesion is critical for early stages of infection, allowing bacteria to establish a foothold within the host. Pili are also involved in modulation of host cell signaling pathways, bacterial invasion into host cells, and biofilm formation. Pili are critical for initiating and sustaining infection and thus represent attractive targets for the development of antivirulence therapeutics. Such therapeutics offer a promising alternative to broad-spectrum antibiotics and provide a means to combat antibiotic resistance and treat infection while preserving the beneficial microbiota. A number of strategies have been taken to develop antipilus therapeutics, including vaccines against pilus proteins, competitive inhibitors of pilus-mediated adhesion, and small molecules that disrupt pilus biogenesis. Here we provide an overview of the function and assembly of CU pili and describe current efforts aimed at interfering with these critical virulence structures.


Subject(s)
Bacterial Secretion Systems/drug effects , Fimbriae, Bacterial/drug effects , Gram-Negative Bacterial Infections/drug therapy , Molecular Chaperones/metabolism , Adhesins, Bacterial/metabolism , Clinical Trials as Topic , Escherichia coli Proteins/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Gram-Negative Bacteria/drug effects , Humans , Models, Molecular , Uropathogenic Escherichia coli , Virulence
5.
EMBO Rep ; 20(3)2019 03.
Article in English | MEDLINE | ID: mdl-30683680

ABSTRACT

Extracellular vesicles (EVs) have been shown to carry microbial components and function in the host defense against infections. In this study, we demonstrate that Mycobacterium tuberculosis (M.tb) RNA is delivered into macrophage-derived EVs through an M.tb SecA2-dependent pathway and that EVs released from M.tb-infected macrophages stimulate a host RIG-I/MAVS/TBK1/IRF3 RNA sensing pathway, leading to type I interferon production in recipient cells. These EVs also promote, in a RIG-I/MAVS-dependent manner, the maturation of M.tb-containing phagosomes through a noncanonical LC3 pathway, leading to increased bacterial killing. Moreover, treatment of M.tb-infected macrophages or mice with a combination of moxifloxacin and EVs, isolated from M.tb-infected macrophages, significantly lowered bacterial burden relative to either treatment alone. We hypothesize that EVs, which are preferentially removed by macrophages in vivo, can be combined with effective antibiotics as a novel approach to treat drug-resistant TB.


Subject(s)
Extracellular Vesicles/metabolism , Host-Pathogen Interactions/immunology , Immunity , Microbial Viability , Mycobacterium tuberculosis/metabolism , RNA, Bacterial/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Secretion Systems/drug effects , DEAD Box Protein 58/metabolism , Extracellular Vesicles/drug effects , Host-Pathogen Interactions/drug effects , Immunity/drug effects , Interferon Type I/biosynthesis , Lysosomes/drug effects , Lysosomes/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Microbial Viability/drug effects , Moxifloxacin/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Myeloid Differentiation Factor 88/metabolism , Phagocytosis/drug effects , Phagosomes/drug effects , Phagosomes/metabolism , Signal Transduction/drug effects
6.
Article in English | MEDLINE | ID: mdl-30524970

ABSTRACT

Dozens of Gram negative pathogens use one or more type III secretion systems (T3SS) to disarm host defenses or occupy a beneficial niche during infection of a host organism. While the T3SS represents an attractive drug target and dozens of compounds with T3SS inhibitory activity have been identified, few T3SS inhibitors have been validated and mode of action determined. One issue is the lack of standardized orthogonal assays following high throughput screening. Using a training set of commercially available compounds previously shown to possess T3SS inhibitory activity, we demonstrate the utility of an experiment pipeline comprised of six distinct assays to assess the stages of type III secretion impacted: T3SS gene copy number, T3SS gene expression, T3SS basal body and needle assembly, secretion of cargo through the T3SS, and translocation of T3SS effector proteins into host cells. We used enteropathogenic Yersinia as the workhorse T3SS-expressing model organisms for this experimental pipeline, as Yersinia is sensitive to all T3SS inhibitors we tested, including those active against other T3SS-expressing pathogens. We find that this experimental pipeline is capable of rapidly distinguishing between T3SS inhibitors that interrupt the process of type III secretion at different points in T3SS assembly and function. For example, our data suggests that Compound 3, a malic diamide, blocks either activity of the assembled T3SS or alters the structure of the T3SS in a way that blocks T3SS cargo secretion but not antibody recognition of the T3SS needle. In contrast, our data predicts that Compound 4, a haloid-containing sulfonamidobenzamide, disrupts T3SS needle subunit secretion or assembly. Furthermore, we suggest that misregulation of copy number control of the pYV virulence plasmid, which encodes the Yersinia T3SS, should be considered as a possible mode of action for compounds with T3SS inhibitory activity against Yersinia.


Subject(s)
Type III Secretion Systems/drug effects , Type III Secretion Systems/metabolism , Yersinia/drug effects , Yersinia/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Bacterial Secretion Systems/drug effects , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/metabolism , Diamide/pharmacology , Gene Dosage , Gene Expression Regulation, Bacterial , Genes, Bacterial , Malates/pharmacology , Plasmids/genetics , Protein Tyrosine Phosphatases/genetics , Type III Secretion Systems/genetics , Virulence/genetics , Yersinia/genetics , Yersinia pseudotuberculosis/metabolism
7.
Microb Pathog ; 110: 225-231, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28687320

ABSTRACT

Chronic burn wound infections caused by Stapyhylococcus aureus (S. aureus) are largely associated with biofilm formation. However, the mechanism by which S. aureus form biofilm in clinical environments is far less understood. In the present study we addressed the association between biofilm formation and membrane vesicle (MV) secretion of S. aureus during vancomycin treatment. A representative methicillin-resistant S. aureus (MRSA) strain BWMR22 obtained from a chronic burn wound was used in this study. Transmission electron microscope was used to observe the MV secretion. Fourier transform infrared spectroscopy was used to analyze the chemical component of MV. Biofilm formation was assayed under conditions of sub-inhibitory concentrations of vancomycin. Functional potencies of MV in surface adhesion and auto-aggregation were assayed in the presence of additional purified MVs. Biofilm formation by S. aureus BWMR22 was enhanced in the presence of sub-inhibitory concentration of vancomycin. Vancomycin treatment caused an increase in the chemical composition of protein relative to carbohydrates of secreted MVs, a property which was highly associated with bacterial hydrophobicity, surface adhesion, and intercellular aggregation. These findings suggest that MV secretion is correlated with biofilm formation by MRSA especially under clinical conditions with improper vancomycin chemotherapy. This study first demonstrates a potential role of MVs in the biofilm formation by S. aureus, stresses on the importance of avoiding low dose of antibiotic therapy in controlling of S. aureus infections, and provides further information to reveal the mechanisms behind MRSA infections.


Subject(s)
Bacterial Secretion Systems/drug effects , Biofilms/drug effects , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/metabolism , Bacterial Secretion Systems/ultrastructure , Cell Aggregation/drug effects , Cell Communication/drug effects , Cell Culture Techniques , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Vancomycin/administration & dosage
8.
Mol Plant Microbe Interact ; 30(1): 28-34, 2017 01.
Article in English | MEDLINE | ID: mdl-27918247

ABSTRACT

In this study, we investigated genetic elements of the type IV secretion system (T4SS) found in Sinorhizobium spp. and the role they play in symbiosis. Sinorhizobium meliloti and S. medicae each contain a putative T4SS similar to that used by Agrobacterium tumefaciens during pathogenesis. The Cre reporter assay for translocation system was used to validate potential effector proteins. Both S. meliloti and S. medicae contained the effector protein TfeA, which was translocated into the host plant. Sequence analysis revealed the presence of a nod box involved in transcriptional activation of symbiosis-related genes, upstream of the transcriptional regulator (virG) in the Sinorhizobium T4SS. Replicate quantitative reverse transcription-polymerase chain reaction analyses indicated that luteolin, released by roots and seeds of Medicago truncatula, upregulated transcription of tfeA and virG. Mutations in the T4SS apparatus or tfeA alone resulted in reduced numbers of nodules formed on M. truncatula genotypes. In addition, S. meliloti KH46c, which contains a deletion in the T4SS, was less competitive for nodule formation when coinoculated with an equal number of cells of the wild-type strain. To our knowledge, TfeA is the first T4SS effector protein identified in Sinorhizobium spp. Our results indicate that Sinorhizobium i) uses a T4SS during initiation of symbiosis with Medicago spp., and ii) alters Medicago cells in planta during symbiosis. This study also offers additional bioinformatic evidence that several different rhizobial species may use the T4SS in symbiosis with other legumes.


Subject(s)
Bacterial Secretion Systems , Medicago truncatula/microbiology , Sinorhizobium/physiology , Symbiosis , Arabidopsis/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Secretion Systems/drug effects , Bacterial Secretion Systems/genetics , Gene Deletion , Genes, Bacterial , Genotype , Luteolin/pharmacology , Medicago truncatula/drug effects , Medicago truncatula/genetics , Phenotype , Reproducibility of Results , Root Nodules, Plant/drug effects , Root Nodules, Plant/metabolism , Sinorhizobium/drug effects , Sinorhizobium/genetics , Symbiosis/drug effects , Symbiosis/genetics , Synteny/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Cell Rep ; 15(5): 951-958, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117415

ABSTRACT

Type VI secretion systems (T6SSs) are nanomachines used for interbacterial killing and intoxication of eukaryotes. Although Vibrio cholerae is a model organism for structural studies on T6SSs, the underlying regulatory network is less understood. A recent study showed that the T6SS is part of the natural competence regulon in V. cholerae and is activated by the regulator TfoX. Here, we identify the TfoX homolog TfoY as a second activator of the T6SS. Importantly, despite inducing the same T6SS core machinery, the overall regulons differ significantly for TfoX and TfoY. We show that TfoY does not contribute to competence induction. Instead, TfoY drives the production of T6SS-dependent and T6SS-independent toxins, together with an increased motility phenotype. Hence, we conclude that V. cholerae uses its sole T6SS in response to diverse cues and for distinctive outcomes: either to kill for the prey's DNA, leading to horizontal gene transfer, or as part of a defensive escape reaction.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Vibrio cholerae/metabolism , Bacterial Proteins/genetics , Bacterial Secretion Systems/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Models, Biological , Sequence Homology, Amino Acid , Vibrio cholerae/drug effects , Vibrio cholerae/genetics
10.
PLoS One ; 11(2): e0148387, 2016.
Article in English | MEDLINE | ID: mdl-26828590

ABSTRACT

BteA is one of the effectors secreted from the Bordetella bronchiseptica type III secretion system. It has been reported that BteA induces necrosis in mammalian cells; however, the roles of BteA during the infection process are largely unknown. In order to investigate the BteA functions, morphological changes of the cells infected with the wild-type B. bronchiseptica were examined by time-lapse microscopy. L2 cells, a rat lung epithelial cell line, spread at 1.6 hours after B. bronchiseptica infection. Membrane ruffles were observed at peripheral parts of infected cells during the cell spreading. BteA-dependent cytotoxicity and cell detachment were inhibited by addition of cytochalasin D, an actin polymerization inhibitor. Domain analyses of BteA suggested that two separate amino acid regions, 200-312 and 400-658, were required for the necrosis induction. In order to examine the intra/intermolecular interactions of BteA, the amino- and the carboxyl-terminal moieties were purified as recombinant proteins from Escherichia coli. The amino-terminal moiety of BteA appeared to interact with the carboxyl-terminal moiety in the pull-down assay in vitro. When we measured the amounts of bacteria phagocytosed by J774A.1, a macrophage-like cell line, the phagocytosed amounts of B. bronchiseptica strains that deliver BteA into the host cell cytoplasm were significantly lower than those of strains that lost the ability to translocate BteA into the host cell cytoplasm. These results suggest that B. bronchiseptica induce necrosis by exploiting the actin polymerization signaling pathway and inhibit macrophage phagocytosis.


Subject(s)
Actin Cytoskeleton/metabolism , Bacterial Secretion Systems , Bordetella bronchiseptica/physiology , Macrophages/metabolism , Macrophages/microbiology , Phagocytosis , Signal Transduction , Actin Cytoskeleton/drug effects , Amino Acids/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Secretion Systems/drug effects , COS Cells , Cell Shape/drug effects , Chlorocebus aethiops , Cytochalasin B/pharmacology , Endocytosis/drug effects , Gentamicins/pharmacology , L-Lactate Dehydrogenase/metabolism , Macrophages/drug effects , Mice , Mutant Proteins/metabolism , Necrosis , Phagocytes/metabolism , Phagocytes/microbiology , Phagocytosis/drug effects , Protein Multimerization/drug effects , Protein Structure, Tertiary , Rats , Signal Transduction/drug effects , Time-Lapse Imaging
11.
Antimicrob Agents Chemother ; 60(4): 2028-38, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26824945

ABSTRACT

Many bacterial pathogens assemble surface fibers termed pili or fimbriae that facilitate attachment to host cells and colonization of host tissues. The chaperone/usher (CU) pathway is a conserved secretion system that is responsible for the assembly of virulence-associated pili by many different Gram-negative bacteria. Pilus biogenesis by the CU pathway requires a dedicated periplasmic chaperone and an integral outer membrane (OM) assembly and secretion platform termed the usher. Nitazoxanide (NTZ), an antiparasitic drug, was previously shown to inhibit the function of aggregative adherence fimbriae and type 1 pili assembled by the CU pathway in enteroaggregativeEscherichia coli, an important causative agent of diarrhea. We show here that NTZ also inhibits the function of type 1 and P pili from uropathogenicE. coli(UPEC). UPEC is the primary causative agent of urinary tract infections, and type 1 and P pili mediate colonization of the bladder and kidneys, respectively. By analysis of the different stages of the CU pilus biogenesis pathway, we show that treatment of bacteria with NTZ causes a reduction in the number of usher molecules in the OM, resulting in a loss of pilus assembly on the bacterial surface. In addition, we determine that NTZ specifically prevents proper folding of the usher ß-barrel domain in the OM. Our findings demonstrate that NTZ is a pilicide with a novel mechanism of action and activity against diverse CU pathways. This suggests that further development of the NTZ scaffold may lead to new antivirulence agents that target the usher to prevent pilus assembly.


Subject(s)
Antiparasitic Agents/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Fimbriae, Bacterial/chemistry , Molecular Chaperones/antagonists & inhibitors , Protein Subunits/antagonists & inhibitors , Thiazoles/pharmacology , Uropathogenic Escherichia coli/chemistry , Animals , Bacterial Secretion Systems/drug effects , Cloning, Molecular , Erythrocytes/drug effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fimbriae, Bacterial/drug effects , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Gene Expression , Guinea Pigs , Hemagglutination Tests , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nitro Compounds , Plasmids/chemistry , Plasmids/metabolism , Protein Conformation, beta-Strand , Protein Folding/drug effects , Protein Multimerization/drug effects , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/metabolism
12.
Plant Physiol ; 167(3): 1117-35, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25635112

ABSTRACT

AvrE family type III effector proteins share the ability to suppress host defenses, induce disease-associated cell death, and promote bacterial growth. However, despite widespread contributions to numerous bacterial diseases in agriculturally important plants, the mode of action of these effectors remains largely unknown. WtsE is an AvrE family member required for the ability of Pantoea stewartii ssp. stewartii (Pnss) to proliferate efficiently and cause wilt and leaf blight symptoms in maize (Zea mays) plants. Notably, when WtsE is delivered by a heterologous system into the leaf cells of susceptible maize seedlings, it alone produces water-soaked disease symptoms reminiscent of those produced by Pnss. Thus, WtsE is a pathogenicity and virulence factor in maize, and an Escherichia coli heterologous delivery system can be used to study the activity of WtsE in isolation from other factors produced by Pnss. Transcriptional profiling of maize revealed the effects of WtsE, including induction of genes involved in secondary metabolism and suppression of genes involved in photosynthesis. Targeted metabolite quantification revealed that WtsE perturbs maize metabolism, including the induction of coumaroyl tyramine. The ability of mutant WtsE derivatives to elicit transcriptional and metabolic changes in susceptible maize seedlings correlated with their ability to promote disease. Furthermore, chemical inhibitors that block metabolic flux into the phenylpropanoid pathways targeted by WtsE also disrupted the pathogenicity and virulence activity of WtsE. While numerous metabolites produced downstream of the shikimate pathway are known to promote plant defense, our results indicate that misregulated induction of phenylpropanoid metabolism also can be used to promote pathogen virulence.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Pantoea/metabolism , Propanols/metabolism , Zea mays/metabolism , Zea mays/microbiology , Bacterial Secretion Systems/drug effects , Biological Assay , Gene Expression Regulation, Plant/drug effects , Gene Ontology , Genome, Plant , Models, Biological , Mutation/genetics , Pantoea/drug effects , Pantoea/growth & development , Pantoea/pathogenicity , Phenylalanine Ammonia-Lyase/metabolism , Seedlings/drug effects , Seedlings/genetics , Seedlings/microbiology , Shikimic Acid/metabolism , Transcription, Genetic/drug effects , Tyramine , Virulence/drug effects , Zea mays/drug effects , Zea mays/genetics
13.
Infect Immun ; 83(2): 456-69, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25404026

ABSTRACT

In recent years, chemical biology and chemical genomics have been increasingly applied to the field of microbiology to uncover new potential therapeutics as well as to probe virulence mechanisms in pathogens. The approach offers some clear advantages, as identified compounds (i) can serve as a proof of principle for the applicability of drugs to specific targets; (ii) can serve as conditional effectors to explore the function of their targets in vitro and in vivo; (iii) can be used to modulate gene expression in otherwise genetically intractable organisms; and (iv) can be tailored to a narrow or broad range of bacteria. This review highlights recent examples from the literature to illustrate how the use of small molecules has advanced discovery of novel potential treatments and has been applied to explore biological mechanisms underlying pathogenicity. We also use these examples to discuss practical considerations that are key to establishing a screening or discovery program. Finally, we discuss the advantages and challenges of different approaches and the methods that are emerging to address these challenges.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Drug Design , Drug Evaluation, Preclinical , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Small Molecule Libraries/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Secretion Systems/drug effects , Bacterial Toxins/biosynthesis , Bacterial Toxins/toxicity , Butyrates/pharmacology , Fimbriae, Bacterial/drug effects , Gene Expression , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/pathogenicity , Naphthalimides/pharmacology , Small Molecule Libraries/chemistry
14.
J Biomed Sci ; 21: 96, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25296847

ABSTRACT

BACKGROUND: Hyperglycemia increases the risk of gastric cancer in H. pylori-infected patients. High glucose could increase endothelial permeability and cancer-associated signaling. These suggest high glucose may affect H. pylori or its infected status.We used two strains to investigate whether H. pylori growth, viability, adhesion and CagA-phosphorylation level in the infected-AGS cells were influenced by glucose concentration (100, 150, and 200 mg/dL). RESULTS: The growth curves of both strains in 200 mg/dL of glucose were maintained at the highest optimal density after 48 h and the best viability of both strains were retained in the same glucose condition at 72 h. Furthermore, adhesion enhancement of H. pylori was significantly higher in 200 mg/dL of glucose as compared to that in 100 and 150 mg/dL (p < 0.05). CagA protein also increased in higher glucose condition. The cell-associated CagA and phosphorylated-CagA was significantly increased in 150 and 200 mg/dL of glucose concentrations as compared to that of 100 mg/dL (p < 0.05), which were found to be dose-dependent. CONCLUSION: Higher glucose could maintain H. pylori growth and viability after 48 h. H. pylori adhesion and CagA increased to further facilitate the enhancement of cell-associated CagA and phosphorylated CagA in higher glucose conditions.


Subject(s)
Bacterial Adhesion/drug effects , Bacterial Secretion Systems/drug effects , Glucose/pharmacology , Helicobacter pylori/metabolism , Helicobacter pylori/pathogenicity , Sweetening Agents/pharmacology , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line, Tumor , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Humans , Phosphorylation/drug effects
15.
Article in English | MEDLINE | ID: mdl-25325019

ABSTRACT

A detailed understanding of host-pathogen interactions provides exciting opportunities to interfere with the infection process. Anti-virulence compounds aim to modulate or pacify pathogenesis by reducing expression of critical virulence determinants. In particular, prevention of attachment by inhibiting adhesion mechanisms has been the subject of intense research. Whilst it has proven relatively straightforward to develop robust screens for potential anti-virulence compounds, understanding their precise mode of action has proven much more challenging. In this review we illustrate this challenge from our own experiences working with the salicylidene acylhydrazide group of compounds. We aim to provide a useful perspective to guide researchers interested in this field and to avoid some of the obvious pitfalls.


Subject(s)
Anti-Infective Agents/pharmacology , Drug Discovery , Host-Pathogen Interactions/drug effects , Infections/etiology , Anti-Infective Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/metabolism , Bacterial Infections/microbiology , Bacterial Secretion Systems/drug effects , Humans , Infections/drug therapy , Infections/metabolism , Virulence/drug effects
16.
J Biosci Bioeng ; 118(6): 651-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24994472

ABSTRACT

The purpose of these investigations was to evaluate the reduction capability of phenolic acids (ferulic, chlorogenic, gallic, and p-coumaric acids) on indole acetic acid synthesis by food-associated Pseudomonas fluorescens KM05. Specific genetic primer for the type III secretion system (TTSS) in P. fluorescens KM05 was designed and the influence of phenolic acids on its expression was investigated. In the work the ferulic and chlorogenic acids at the concentration of 0.02 and 0.04 µg/ml affected on bacterial growth pattern and the signal molecules production. The phenolic acids, that were appreciable effective against P. fluorescens KM05 indole acetic acid production, significantly suppressed TTSS gene.


Subject(s)
Bacterial Secretion Systems/drug effects , Bacterial Secretion Systems/genetics , Food Microbiology , Gene Expression Regulation, Bacterial/drug effects , Hydroxybenzoates/pharmacology , Indoleacetic Acids/metabolism , Pseudomonas fluorescens/drug effects , Pseudomonas fluorescens/metabolism , Chlorogenic Acid/pharmacology , Coumaric Acids/pharmacology , Gallic Acid/pharmacology , Gene Expression Regulation, Bacterial/genetics , Oxidation-Reduction , Propionates , Pseudomonas fluorescens/genetics , Pseudomonas fluorescens/growth & development , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Virulence/drug effects
17.
Biochem Biophys Res Commun ; 449(4): 455-9, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24853802

ABSTRACT

Natural small-molecule products are promising lead compounds for developing a generation of novel antimicrobials agents to meet the challenge of antibiotic-resistant pathogens. To facilitate the search for novel anti-virulence agents, we chose a virulence factor of Type Three Secretion System (T3SS) as a drug target to screen candidates from a small-molecule library in our laboratory. This study demonstrated fusaric acid had dramatically inhibitory effects on secretion of Salmonella island 1 (SPI-1) effector proteins and invasion of Salmonella into HeLa cells. Moreover, fusaric acid had no inhibitory effects on bacterial growth and viability of host cells. Protein HilA is a key regulator of SPI-1 in Salmonella, which affects transcription of SPI-1 effectors and SPI-1 apparatus genes. In this study, fusaric acid (FA) did not affect secretion of SPI-1 effectors in HilA over-expressed strain, suggesting it did not affect the transcription of SPI-1. In addition, fusaric acid did not affect the protein level of apparatus protein PrgH in SPI-1 needle complex. As a result, we proposed fusaric acid had an inhibitory effect on SPI-1 probably depending on its influence on SicA/InvF. In summary, fusaric acid is a novel inhibitor of T3SS with potential for further developing novel anti-virulence agents.


Subject(s)
Bacterial Proteins/drug effects , Bacterial Secretion Systems/drug effects , Fusaric Acid/pharmacology , Salmonella typhimurium/pathogenicity , Virulence Factors/antagonists & inhibitors , Bacterial Secretion Systems/physiology , HeLa Cells/microbiology , Humans , Salmonella Infections/drug therapy
18.
Antimicrob Agents Chemother ; 58(7): 4123-30, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24798289

ABSTRACT

The type VI secretion system (T6SS) is the most prevalent bacterial secretion system and an important virulence mechanism utilized by Gram-negative bacteria, either to target eukaryotic cells or to combat other microbes. The components show much variability, but some appear essential for the function, and two homologues, denoted VipA and VipB in Vibrio cholerae, have been identified in all T6SSs described so far. Secretion is dependent on binding of an α-helical region of VipA to VipB, and in the absence of this binding, both components are degraded within minutes and secretion is ceased. The aim of the study was to investigate if this interaction could be blocked, and we hypothesized that such inhibition would lead to abrogation of T6S. A library of 9,600 small-molecule compounds was screened for their ability to block the binding of VipA-VipB in a bacterial two-hybrid system (B2H). After excluding compounds that showed cytotoxicity toward eukaryotic cells, that inhibited growth of Vibrio, or that inhibited an unrelated B2H interaction, 34 compounds were further investigated for effects on the T6SS-dependent secretion of hemolysin-coregulated protein (Hcp) or of phospholipase A1 activity. Two compounds, KS100 and KS200, showed intermediate or strong effects in both assays. Analogues were obtained, and compounds with potent inhibitory effects in the assays and desirable physicochemical properties as predicted by in silico analysis were identified. Since the compounds specifically target a virulence mechanism without affecting bacterial replication, they have the potential to mitigate the virulence with minimal risk for development of resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Bacterial Secretion Systems/drug effects , Hemolysin Proteins/metabolism , Phospholipases A1/antagonists & inhibitors , Vibrio cholerae/drug effects , Microbial Sensitivity Tests , Molecular Sequence Data , Phospholipases A1/metabolism , Vibrio cholerae/pathogenicity , Virulence Factors/antagonists & inhibitors
19.
Mol Microbiol ; 91(5): 976-95, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24397487

ABSTRACT

Francisella tularensis is a category A biodefence agent that causes a fatal human disease known as tularaemia. The pathogenicity of F. tularensis depends on its ability to persist inside host immune cells primarily by resisting an attack from host-generated reactive oxygen and nitrogen species (ROS/RNS). Based on the ability of F. tularensis to resist high ROS/RNS levels, we have hypothesized that additional unknown factors act in conjunction with known antioxidant defences to render ROS resistance. By screening a transposon insertion library of F. tularensis LVS in the presence of hydrogen peroxide, we have identified an oxidant-sensitive mutant in putative EmrA1 (FTL_0687) secretion protein. The results demonstrate that the emrA1 mutant is highly sensitive to oxidants and several antimicrobial agents, and exhibits diminished intramacrophage growth that can be restored to wild-type F. tularensis LVS levels by either transcomplementation, inhibition of ROS generation or infection in NADPH oxidase deficient (gp91Phox(-/-)) macrophages. The emrA1 mutant is attenuated for virulence, which is restored by infection in gp91Phox(-/-) mice. Further, EmrA1 contributes to oxidative stress resistance by affecting secretion of Francisella antioxidant enzymes SodB and KatG. This study exposes unique links between transporter activity and the antioxidant defence mechanisms of F. tularensis.


Subject(s)
Bacterial Proteins/metabolism , Francisella tularensis/pathogenicity , Macrophages/microbiology , Membrane Fusion , Microbial Viability , Oxidative Stress , Amino Acid Sequence , Animals , Anti-Bacterial Agents/pharmacology , Antioxidants/metabolism , Bacterial Proteins/chemistry , Bacterial Secretion Systems/drug effects , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Francisella tularensis/genetics , Francisella tularensis/metabolism , Genome, Bacterial/genetics , Humans , Hydrogen Peroxide/pharmacology , Macrophages/drug effects , Macrophages/pathology , Membrane Fusion/drug effects , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microbial Viability/drug effects , Molecular Sequence Data , Mutation/genetics , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Tularemia/microbiology , Tularemia/pathology , Virulence/drug effects
20.
Antimicrob Agents Chemother ; 58(2): 1118-26, 2014.
Article in English | MEDLINE | ID: mdl-24295981

ABSTRACT

The type III secretion system (T3SS) is a bacterial appendage used by dozens of Gram-negative pathogens to subvert host defenses and cause disease, making it an ideal target for pathogen-specific antimicrobials. Here, we report the discovery and initial characterization of two related natural products with T3SS-inhibitory activity that were derived from a marine actinobacterium. Bacterial extracts containing piericidin A1 and the piericidin derivative Mer-A 2026B inhibited Yersinia pseudotuberculosis from triggering T3SS-dependent activation of the host transcription factor NF-κB in HEK293T cells but were not toxic to mammalian cells. As the Yersinia T3SS must be functional in order to trigger NF-κB activation, these data indicate that piericidin A1 and Mer-A 2026B block T3SS function. Consistent with this, purified piericidin A1 and Mer-A 2026B dose-dependently inhibited translocation of the Y. pseudotuberculosis T3SS effector protein YopM inside CHO cells. In contrast, neither compound perturbed bacterial growth in vitro, indicating that piericidin A1 and Mer-A 2026B do not function as general antibiotics in Yersinia. In addition, when Yersinia was incubated under T3SS-inducing culture conditions in the absence of host cells, Mer-A 2026B and piericidin A1 inhibited secretion of T3SS cargo as effectively as or better than several previously described T3SS inhibitors, such as MBX-1641 and aurodox. This suggests that Mer-A 2026B and piericidin A1 do not block type III secretion by blocking the bacterium-host cell interaction, but rather inhibit an earlier stage, such as T3SS needle assembly. In summary, the marine-derived natural products Mer-A 2026B and piericidin A1 possess previously uncharacterized activity against the bacterial T3SS.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Secretion Systems/drug effects , NF-kappa B/genetics , Pyridines/pharmacology , Yersinia pseudotuberculosis/drug effects , Actinomycetales/chemistry , Animals , Anti-Bacterial Agents/isolation & purification , Aurodox/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/metabolism , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Gene Expression Regulation , HEK293 Cells , High-Throughput Screening Assays , Humans , NF-kappa B/metabolism , Protein Transport/drug effects , Pyridines/isolation & purification , Yersinia pseudotuberculosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL