Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Microb Pathog ; 139: 103885, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31790793

ABSTRACT

The ability of Rhodococcus equi to survive in macrophages and cause pneumonia in foals depends on vapA and rhbC genes, which produce the virulence-associated protein A (VapA) and the rhequichelin siderophore, respectively. Virulent R. equi acquires Fe from transferrin by unknown mechanisms. Our objectives were to determine the role of GAPDH in Fe homeostasis, to further characterize GAPDH, rhbC, and vapA expression under iron homeostasis, and to document the occurrence of rhbC gene in R. equi isolates. Therefore, vapA + R. equi was cultured under excessive, physiologic, and restricted iron concentrations, and quantitative culture and gene expression were performed. The relative expression of GAPDH, rhbC, and vapA after 48 h of culture were analyzed by qPCR. To determine the rhbC occurrence, total DNA was extracted from R. equi isolated from foals with clinical rhodococcosis (n = 22), healthy horses (feces, n = 16; nasal swab, n = 9), soil (n = 6), and 2 ATCC reference strains. Conventional PCR was performed to identify genus/species, vapA, and rhbC genes. Iron restriction proportionally decreased R. equi growth rates, and induced high expression of both GAPDH and vapA. The putative role of GAPDH in R. equi iron homeostasis should be further investigated. rhbC was significantly up-regulated under both Fe excess and critical starvation. The rhbC gene was identified in all clinical isolates and soil, but it was absent in 2 isolates from healthy horses, suggesting that rhequichelin is not required for R. equi nasal and intestinal colonization.


Subject(s)
Actinomycetales Infections/microbiology , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Iron/metabolism , Rhodococcus equi/genetics , Rhodococcus equi/metabolism , Homeostasis , Rhodococcus equi/growth & development , Virulence/genetics , Virulence Factors/genetics
2.
Appl Environ Microbiol ; 85(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30683740

ABSTRACT

Rhodococcus equi is a leading cause of severe pneumonia in foals. Standard treatment is dual antimicrobial therapy with a macrolide and rifampin, but the emergence of macrolide- and rifampin-resistant R. equi isolates is an increasing problem. The objective of this study was to determine the effect of macrolide and/or rifampin resistance on fitness of R. equi Three unique isogenic sets were created, each consisting of four R. equi strains, as follows: a susceptible parent isolate, strains resistant to macrolides or rifampin, and a dual macrolide- and rifampin-resistant strain. Each isogenic set's bacterial growth curve was generated in enriched medium, minimal medium (MM), and minimal medium without iron (MM-I). Bacterial survival in soil was analyzed over 12 months at -20°C, 4°C, 25°C, and 37°C, and the ability of these strains to retain antimicrobial resistance during sequential subculturing was determined. Insertion of the mobile element conferring macrolide resistance had minimal effect on in vitro growth. However, two of three rpoB mutations conferring rifampin resistance resulted in a decreased growth rate in MM. In soil, macrolide- or rifampin-resistant R. equi strains exhibited limited growth compared to that of the susceptible R. equi isolate at all temperatures except -20°C. During subculturing, macrolide resistance was lost over time, and two of three rpoB mutations reverted to the wild-type form. The growth of rifampin-resistant R. equi colonies is delayed under nutrient restriction. In soil, possession of rifampin or macrolide resistance results in decreased fitness. Both macrolide and rifampin resistance can be lost after repeated subculturing.IMPORTANCE This work advances our understanding of the opportunistic environmental pathogen Rhodococcus equi, a disease agent affecting horses and immunocompromised people. R. equi is one of the most common causes of severe pneumonia in young horses. For decades, the standard treatment for R. equi pneumonia in horses has been dual antimicrobial therapy with a macrolide and rifampin; effective alternatives to this combination are lacking. The World Health Organization classifies these antimicrobial agents as critically important for human medicine. Widespread macrolide and rifampin resistance in R. equi isolates is a major emerging problem for the horse-breeding industry and might also adversely impact human health if resistant strains infect people or transfer resistance mechanisms to other pathogens. This study details the impact of antimicrobial resistance on R. equi fitness, a vital step for understanding the ecology and epidemiology of resistant R. equi isolates, and will support development of novel strategies to combat antimicrobial resistance.


Subject(s)
Drug Resistance, Bacterial/drug effects , Macrolides/pharmacology , Rhodococcus equi/drug effects , Rhodococcus equi/growth & development , Rifampin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , DNA-Directed RNA Polymerases/genetics , Drug Resistance, Bacterial/genetics , Horse Diseases/microbiology , Horses , Humans , Microbial Sensitivity Tests , Rhodococcus equi/genetics
3.
Cell Microbiol ; 21(1): e12958, 2019 01.
Article in English | MEDLINE | ID: mdl-30251327

ABSTRACT

Professional phagocytic cells such as macrophages are a central part of innate immune defence. They ingest microorganisms into membrane-bound compartments (phagosomes), which acidify and eventually fuse with lysosomes, exposing their contents to a microbicidal environment. Gram-positive Rhodococcus equi can cause pneumonia in young foals and in immunocompromised humans. The possession of a virulence plasmid allows them to subvert host defence mechanisms and to multiply in macrophages. Here, we show that the plasmid-encoded and secreted virulence-associated protein A (VapA) participates in exclusion of the proton-pumping vacuolar-ATPase complex from phagosomes and causes membrane permeabilisation, thus contributing to a pH-neutral phagosome lumen. Using fluorescence and electron microscopy, we show that VapA is also transferred from phagosomes to lysosomes where it permeabilises the limiting membranes for small ions such as protons. This permeabilisation process is different from that of known membrane pore formers as revealed by experiments with artificial lipid bilayers. We demonstrate that, at 24 hr of infection, virulent R. equi is contained in a vacuole, which is enriched in lysosome material, yet possesses a pH of 7.2 whereas phagosomes containing a vapA deletion mutant have a pH of 5.8 and those with virulence plasmid-less sister strains have a pH of 5.2. Experimentally neutralising the macrophage endocytic system allows avirulent R. equi to multiply. This observation is mirrored in the fact that virulent and avirulent R. equi multiply well in extracts of purified lysosomes at pH 7.2 but not at pH 5.1. Together these data indicate that the major function of VapA is to generate a pH-neutral and hence growth-promoting intracellular niche. VapA represents a new type of Gram-positive virulence factor by trafficking from one subcellular compartment to another, affecting membrane permeability, excluding proton-pumping ATPase, and consequently disarming host defences.


Subject(s)
Bacterial Proteins/metabolism , Host-Pathogen Interactions , Phagosomes/microbiology , Proton-Translocating ATPases/antagonists & inhibitors , Rhodococcus equi/growth & development , Rhodococcus equi/metabolism , Virulence Factors/metabolism , Animals , Cell Line , Humans , Hydrogen-Ion Concentration , Mice , Microscopy, Electron , Microscopy, Fluorescence , Virulence
4.
Article in English | MEDLINE | ID: mdl-30373803

ABSTRACT

The objective of this study was to determine the prevalence of Rhodococcus equi strains resistant to macrolides and rifampin over time in clinical samples from foals submitted to diagnostic laboratories in central Kentucky. We performed a retrospective observational study of all clinical samples from foals that were submitted to veterinary diagnostic laboratories in Kentucky between January 1995 and December 2017. Samples were included if the R. equi bacterium was cultured and tested for in vitro susceptibility to erythromycin or rifampin. In vitro susceptibility testing to erythromycin was available for 2,169 isolates of R. equi, while susceptibility testing to both erythromycin and rifampin was available for 1,681 isolates. Rifampin resistance was first detected in 2000, and erythromycin resistance was first detected in 2004. Between 1995 and 2006, the proportion of resistant isolates of R. equi was 0.7% for erythromycin and 2.3% for rifampin. There was a significant (P < 0.001) increase in the proportion of resistant R. equi between 2007 and 2017, with 13.6% of isolates being resistant to erythromycin and 16.1% being resistant to rifampin. Between 2007 and 2017, isolates of R. equi resistant to erythromycin or rifampin were significantly less likely to be isolated from feces than from the respiratory tract, other soft tissues, or musculoskeletal infections. The considerable increase in the prevalence of isolates of R. equi resistant to macrolides and rifampin since 2007 is of concern for both human and animal health.


Subject(s)
Actinomycetales Infections/veterinary , Anti-Bacterial Agents/pharmacology , Erythromycin/pharmacology , Musculoskeletal Diseases/veterinary , Rhodococcus equi/drug effects , Rifampin/pharmacology , Soft Tissue Infections/veterinary , Actinomycetales Infections/drug therapy , Actinomycetales Infections/epidemiology , Actinomycetales Infections/microbiology , Animals , Azithromycin/pharmacology , Clarithromycin/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Equidae , Feces/microbiology , Horses , Kentucky/epidemiology , Microbial Sensitivity Tests , Musculoskeletal Diseases/drug therapy , Musculoskeletal Diseases/epidemiology , Musculoskeletal Diseases/microbiology , Prevalence , Respiratory System/drug effects , Respiratory System/microbiology , Rhodococcus equi/genetics , Rhodococcus equi/growth & development , Rhodococcus equi/isolation & purification , Soft Tissue Infections/drug therapy , Soft Tissue Infections/epidemiology , Soft Tissue Infections/microbiology
5.
PLoS One ; 13(10): e0204475, 2018.
Article in English | MEDLINE | ID: mdl-30286098

ABSTRACT

Rhodococcus equi is a facultative intracellular bacterium of macrophages and is an important pathogen of animals and immunocompromised people wherein disease results in abcessation of the lungs and other sites. Prior work has shown that the presence of the major virulence determinant, VapA, encoded on the pVAPA-type plasmid, disrupts normal phagosome development and is essential for bacterial replication within macrophages. pVAPA- type plasmids are typical of R. equi strains derived from foals while strains from pigs carry plasmids of the pVAPB-type, lacking vapA, and those from humans harbor various types of plasmids including pVAPA and pVAPB. Through the creation and analysis of a series of gene deletion mutants, we found that vapK1 or vapK2 is required for optimal intracellular replication of an R. equi isolate carrying a pVAPB plasmid type. Complementation analysis of a ΔvapA R. equi strain with vapK1 or vapK2 showed the VapK proteins of the pVAPB-type plasmid could restore replication capacity to the macrophage growth-attenuated ΔvapA strain. Additionally, in contrast to the intracellular growth capabilities displayed by an equine R. equi transconjugant strain carrying a pVAPB-type plasmid, a transconjugant strain carrying a pVAPB-type plasmid deleted of vapK1 and vapK2 proved incapable of replication in equine macrophages. Cumulatively, these data indicate that VapK1 and K2 are functionally equivalent to VapA.


Subject(s)
Bacterial Proteins/genetics , Macrophages/microbiology , Plasmids , Rhodococcus equi/genetics , Rhodococcus equi/pathogenicity , Virulence Factors/genetics , Actinomycetales Infections/microbiology , Actinomycetales Infections/veterinary , Animals , Cells, Cultured , Female , Horse Diseases/microbiology , Horses , Mice, Inbred BALB C , Mutation , Rhodococcus equi/growth & development , Rhodococcus equi/isolation & purification
6.
Vet Microbiol ; 223: 42-46, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30173750

ABSTRACT

Rhodococcus equi, a soil saprophyte, is a common cause of pneumonia in foals and a frequent opportunistic pathogen in immunosuppressed people. Because it is widespread in the environment, R. equi can be detected in the feces of most horses. However, the exact timing and rate of shedding relative to infection is unknown. The objectives of this study were to quantify shedding of R. equi in mares and foals after experimental infection of foals with 2 different inocula and to determine the effect of composting on concentrations of R. equi in contaminated bedding. Foals were infected intratracheally with virulent R. equi using inocula of 1 × 107 CFU/mL (n = 16) or 1 × 106 CFU/mL (n = 12) at 23 ± 2 days (range 21 to 27 days) of age. Fecal samples were collected from mares and foals prior to infection and on days 3, 7, and 14 post-infection for quantitative culture of total and virulent R. equi. Waste from the horses was composted for 7 days. Concentrations of total and virulent R. equi in foal feces were significantly higher on day 14 post-infection compared to day 0, regardless of inoculum size. Concentration of total R. equi in mare feces was significantly higher on days 3, 7 and 14 compared to day 0 regardless of inoculum size, whereas shedding of virulent R. equi only increased on day 14 post-infection. Composting for 7 days significantly decreased concentrations of total R. equi and virulent R. equi by an average of 1.08 ± 0.21 and 0.59 ± 0.26 log10 CFU/g, respectively.


Subject(s)
Actinomycetales Infections/veterinary , Horse Diseases/microbiology , Rhodococcus equi/growth & development , Actinomycetales Infections/microbiology , Animals , Animals, Newborn , Bedding and Linens/microbiology , Composting , Feces/microbiology , Female , Horses , Rhodococcus equi/pathogenicity , Virulence
7.
Cytokine ; 102: 7-11, 2018 02.
Article in English | MEDLINE | ID: mdl-29245049

ABSTRACT

Rhodococcus equi is a common cause of pneumonia in foals and an opportunistic pathogen in immunosuppressed people. The ability of R. equi to survive and replicate in macrophages is the basis of its pathogenicity. Limited knowledge about the role of cytokines in host defense against R. equi comes from studies in mice and the role of cytokines in intracellular survival of R. equi in equine macrophages is unknown. The objectives of this study were to determine the effect of priming with interferon (IFN)-γ, interleukin (IL)-1ß, IL-4, IL-6, IL-10, or tumor necrosis factor (TNF)-α at various concentrations on intracellular survival of virulent R. equi in equine monocyte-derived macrophages (MDM), and to determine the effects of various combinations of the same cytokines on intracellular survival of R. equi. MDM from 10 adult horses were primed with recombinant equine cytokines at doubling concentrations ranging from 25 to 200 ng/mL prior to infection with virulent R. equi. Priming with IFN-γ, TNF-α, or IL-6 significantly decreased intracellular replication of R. equi compared to unprimed monolayers. In contrast, priming with IL-10 or IL-1ß significantly increased intracellular replication of R. equi. Pairwise combinations of the cytokines listed above did not results in synergism or antagonism. This study demonstrated that IFN-γ, TNF-α, or IL-6 improved equine MDM function against R. equi whereas IL-1ß or IL-10 were detrimental.


Subject(s)
Actinomycetales Infections/microbiology , Interferon-gamma/pharmacology , Interleukins/pharmacology , Macrophages/microbiology , Rhodococcus equi/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Drug Interactions , Horses , Rhodococcus equi/growth & development
8.
J Vet Med Sci ; 79(8): 1323-1326, 2017 Aug 04.
Article in English | MEDLINE | ID: mdl-28690290

ABSTRACT

Rhodococcus equi is a facultative intracellular bacterium that can escape from bactericidal mechanisms associated with phagocytosis. Virulence-associated protein A (VapA), encoded on a virulence-associated plasmid, is essential for intracellular survival in macrophages, but its function is not known. Here, we show that the extracellular addition of recombinant glutathione S-transferase (GST)-VapA fusion protein rescued the intracellular replication defect of a mutant lacking the vapA gene. Furthermore, the virulence-plasmid-cured strain could also multiply to nearly wild-type levels by the addition of GST-VapA. The present data suggest that VapA can alter the intraphagocytic environment, thereby affecting its suitability for the growth of R. equi.


Subject(s)
Rhodococcus equi/genetics , Rhodococcus equi/pathogenicity , Staphylococcal Protein A/genetics , Virulence Factors , Virulence/genetics , Actinomycetales Infections/microbiology , Animals , Cell Line , Gene Deletion , Genes, Bacterial/genetics , Glutathione Transferase , Macrophages/microbiology , Mice , Plasmids , Recombinant Proteins , Rhodococcus equi/growth & development
9.
Infect Immun ; 83(8): 3137-45, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26015480

ABSTRACT

Rhodococcus equi is a facultative intracellular pathogen of macrophages, relying on the presence of a conjugative virulence plasmid harboring a 21-kb pathogenicity island (PAI) for growth in host macrophages. The PAI encodes a family of 6 virulence-associated proteins (Vaps) in addition to 20 other proteins. The contribution of these to virulence has remained unclear. We show that the presence of only 3 virulence plasmid genes (of 73 in total) is required and sufficient for intracellular growth. These include a single vap family member, vapA, and two PAI-located transcriptional regulators, virR and virS. Both transcriptional regulators are essential for wild-type-level expression of vapA, yet vapA expression alone is not sufficient to allow intracellular growth. A whole-genome microarray analysis revealed that VirR and VirS substantially integrate themselves into the chromosomal regulatory network, significantly altering the transcription of 18% of all chromosomal genes. This pathoadaptation involved significant enrichment of select gene ontologies, in particular, enrichment of genes involved in transport processes, energy production, and cellular metabolism, suggesting a major change in cell physiology allowing the bacterium to grow in the hostile environment of the host cell. The results suggest that following the acquisition of the virulence plasmid by an avirulent ancestor of R. equi, coevolution between the plasmid and the chromosome took place, allowing VirR and VirS to regulate the transcription of chromosomal genes in a process that ultimately promoted intracellular growth. Our findings suggest a mechanism for cooption of existing chromosomal traits during the evolution of a pathogenic bacterium from an avirulent saprophyte.


Subject(s)
Actinomycetales Infections/microbiology , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Plasmids/genetics , Rhodococcus equi/physiology , Transcriptome , Adaptation, Physiological , Animals , Bacterial Proteins/metabolism , Humans , Mice , Plasmids/metabolism , Rhodococcus equi/genetics , Rhodococcus equi/growth & development , Transcription, Genetic , Virulence Factors/genetics
10.
Infect Immun ; 83(7): 2725-37, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25895973

ABSTRACT

We report a novel host-associated virulence plasmid in Rhodococcus equi, pVAPN, carried by bovine isolates of this facultative intracellular pathogenic actinomycete. Surprisingly, pVAPN is a 120-kb invertron-like linear replicon unrelated to the circular virulence plasmids associated with equine (pVAPA) and porcine (pVAPB variant) R. equi isolates. pVAPN is similar to the linear plasmid pNSL1 from Rhodococcus sp. NS1 and harbors six new vap multigene family members (vapN to vapS) in a vap pathogenicity locus presumably acquired via en bloc mobilization from a direct predecessor of equine pVAPA. Loss of pVAPN rendered R. equi avirulent in macrophages and mice. Mating experiments using an in vivo transconjugant selection strategy demonstrated that pVAPN transfer is sufficient to confer virulence to a plasmid-cured R. equi recipient. Phylogenetic analyses assigned the vap multigene family complement from pVAPN, pVAPA, and pVAPB to seven monophyletic clades, each containing plasmid type-specific allelic variants of a precursor vap gene carried by the nearest vap island ancestor. Deletion of vapN, the predicted "bovine-type" allelic counterpart of vapA, essential for virulence in pVAPA, abrogated pVAPN-mediated intramacrophage proliferation and virulence in mice. Our findings support a model in which R. equi virulence is conferred by host-adapted plasmids. Their central role is mediating intracellular proliferation in macrophages, promoted by a key vap determinant present in the common ancestor of the plasmid-specific vap islands, with host tropism as a secondary trait selected during coevolution with specific animal species.


Subject(s)
Macrophages/microbiology , Microbial Viability , Plasmids , Rhodococcus equi/physiology , Animals , Cattle , Cluster Analysis , Conjugation, Genetic , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Gene Transfer, Horizontal , Genes, Bacterial , Mice, Inbred BALB C , Molecular Sequence Data , Phylogeny , Rhodococcus equi/genetics , Rhodococcus equi/growth & development , Rhodococcus equi/isolation & purification , Sequence Analysis, DNA , Sequence Homology , Virulence , Virulence Factors/genetics
11.
Microb Cell Fact ; 13: 130, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25201011

ABSTRACT

The gene encoding the putative reductase component (KshB) of 3-ketosteroid 9α-hydroxylase was cloned from Rhodococcus equi USA-18, a cholesterol oxidase-producing strain formerly named Arthrobacter simplex USA-18, by PCR according to consensus amino acid motifs of several bacterial KshB subunits. Deletion of the gene in R. equi USA-18 by a PCR-targeted gene disruption method resulted in a mutant strain that could accumulate up to 0.58 mg/ml 1,4-androstadiene-3,17-dione (ADD) in the culture medium when 0.2% cholesterol was used as the carbon source, indicating the involvement of the deleted enzyme in 9α-hydroxylation of steroids. In addition, this mutant also accumulated 3-oxo-23,24-bisnorchola-1,4-dien-22-oic acid (Δ1,4-BNC). Because both ADD and Δ1,4-BNC are important intermediates for the synthesis of steroid drugs, this mutant derived from R. equi USA-18 may deserve further investigation for its application potential.


Subject(s)
Androstadienes/metabolism , Gene Deletion , Mixed Function Oxygenases/genetics , Oxidoreductases/genetics , Rhodococcus equi/genetics , Steroids/chemistry , Sterols/metabolism , Androstadienes/chemistry , Biotransformation , Cell Line , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Chromosomes, Bacterial/genetics , Gene Knockout Techniques , Genes, Bacterial , Humans , Macrophages/microbiology , Mass Spectrometry , Polymerase Chain Reaction , Reference Standards , Reproducibility of Results , Rhodococcus equi/enzymology , Rhodococcus equi/growth & development , Steroids/metabolism , Sterols/chemistry , Time Factors
12.
PLoS One ; 8(3): e60612, 2013.
Article in English | MEDLINE | ID: mdl-23555995

ABSTRACT

Rhodococcus equi is a facultative intracellular pathogen of macrophages and the causative agent of foal pneumonia. R. equi virulence is usually assessed by analyzing intracellular growth in macrophages by enumeration of bacteria following cell lysis, which is time consuming and does not allow for a high throughput analysis. This paper describes the use of an impedance based real-time method to characterize proliferation of R. equi in macrophages, using virulent and attenuated strains lacking the vapA gene or virulence plasmid. Image analysis suggested that the time-dependent cell response profile (TCRP) is governed by cell size and roundness as well as cytoxicity of infecting R. equi strains. The amplitude and inflection point of the resulting TCRP were dependent on the multiplicity of infection as well as virulence of the infecting strain, thus distinguishing between virulent and attenuated strains.


Subject(s)
Actinomycetales Infections/microbiology , Macrophages/microbiology , Rhodococcus equi/pathogenicity , Actinomycetales Infections/veterinary , Animals , Cell Line , Electric Impedance , Horses/microbiology , Host-Pathogen Interactions , Humans , Macrophages/cytology , Mice , Mutation , Rhodococcus equi/genetics , Rhodococcus equi/growth & development , Rhodococcus equi/physiology
13.
Infect Immun ; 80(12): 4106-14, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22966042

ABSTRACT

We previously showed that the facultative intracellular pathogen Rhodococcus equi produces a nondiffusible and catecholate-containing siderophore (rhequibactin) involved in iron acquisition during saprophytic growth. Here, we provide evidence that the rhbABCDE cluster directs the biosynthesis of a hydroxamate siderophore, rhequichelin, that plays a key role in virulence. The rhbC gene encodes a nonribosomal peptide synthetase that is predicted to produce a tetrapeptide consisting of N(5)-formyl-N(5)-hydroxyornithine, serine, N(5)-hydroxyornithine, and N(5)-acyl-N(5)-hydroxyornithine. The other rhb genes encode putative tailoring enzymes mediating modification of ornithine residues incorporated into the hydroxamate product of RhbC. Transcription of rhbC was upregulated during growth in iron-depleted medium, suggesting that it plays a role in iron acquisition. This was confirmed by deletion of rhbCD, rendering the resulting strain R. equi SID2 unable to grow in the presence of the iron chelator 2,2-dipyridyl. Supernatant of the wild-type strain rescued the phenotype of R. equi SID2. The importance of rhequichelin in virulence was highlighted by the rapid increase in transcription levels of rhbC following infection and the inability of R. equi SID2 to grow within macrophages. Unlike the wild-type strain, R. equi SID2 was unable to replicate in vivo and was rapidly cleared from the lungs of infected mice. Rhequichelin is thus a key virulence-associated factor, although nonpathogenic Rhodococcus species also appear to produce rhequichelin or a structurally closely related compound. Rhequichelin biosynthesis may therefore be considered an example of cooption of a core actinobacterial trait in the evolution of R. equi virulence.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Hydroxamic Acids/metabolism , Iron/metabolism , Oligopeptides/metabolism , Rhodococcus equi/pathogenicity , Siderophores/metabolism , Virulence Factors/metabolism , Animals , Cells, Cultured , Female , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mice, SCID , Peptide Synthases/genetics , Peptide Synthases/metabolism , Rhodococcus equi/genetics , Rhodococcus equi/growth & development , Rhodococcus equi/metabolism , Virulence , Virulence Factors/genetics
14.
PLoS One ; 7(8): e42396, 2012.
Article in English | MEDLINE | ID: mdl-22879963

ABSTRACT

Rhodococcus equi is one of the most widespread causes of disease in foals aged from 1 to 6 months. R. equi possesses antioxidant defense mechanisms to protect it from reactive oxygen metabolites such as hydrogen peroxide (H(2)O(2)) generated during the respiratory burst of phagocytic cells. These defense mechanisms include enzymes such as catalase, which detoxify hydrogen peroxide. Recently, an analysis of the R. equi 103 genome sequence revealed the presence of four potential catalase genes. We first constructed ΔkatA-, ΔkatB-, ΔkatC-and ΔkatD-deficient mutants to study the ability of R. equi to survive exposure to H(2)O(2)in vitro and within mouse peritoneal macrophages. Results showed that ΔkatA and, to a lesser extent ΔkatC, were affected by 80 mM H(2)O(2). Moreover, katA deletion seems to significantly affect the ability of R. equi to survive within murine macrophages. We finally investigated the expression of the four catalases in response to H(2)O(2) assays with a real time PCR technique. Results showed that katA is overexpressed 367.9 times (± 122.6) in response to exposure to 50 mM of H(2)O(2) added in the stationary phase, and 3.11 times (± 0.59) when treatment was administered in the exponential phase. In untreated bacteria, katB, katC and katD were overexpressed from 4.3 to 17.5 times in the stationary compared to the exponential phase. Taken together, our results show that KatA is the major catalase involved in the extreme H(2)O(2) resistance capability of R. equi.


Subject(s)
Catalase/genetics , Genes, Bacterial/genetics , Hydrogen Peroxide/pharmacology , Rhodococcus equi/drug effects , Rhodococcus equi/genetics , Animals , Gene Deletion , Gene Expression Regulation, Bacterial/drug effects , Macrophages/drug effects , Macrophages/microbiology , Male , Mice , Mice, Inbred BALB C , Microbial Viability/drug effects , Phylogeny , Rhodococcus equi/enzymology , Rhodococcus equi/growth & development
15.
Microbiology (Reading) ; 157(Pt 8): 2357-2368, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21565932

ABSTRACT

The virulence plasmid of the pathogenic actinomycete Rhodococcus equi is essential for proliferation of this pathogen in macrophages and the development of disease. The pathogenicity island of this plasmid encodes a family of virulence-associated proteins (Vap), one of which (VapA) is a virulence factor. This paper describes the vcgAB operon (vapA co-expressed gene), located upstream of the vapA operon. Transcription of the vcgAB operon gave rise to transcripts with a half-life similar to those determined for other virulence plasmid genes (1.8 min). Transcription started at a promoter similar to the vapA promoter, and proceeded through an inefficient terminator into the downstream vcgC gene. In addition, vcgC is also transcribed from a promoter downstream of vcgB. The vcgAB and vapA operons were coordinately regulated by temperature and pH in a synergistic manner. The latter parameter only affected transcription at higher growth temperatures, indicating that temperature is the dominant regulatory signal. Transcription of the vcgAB operon increased 10-fold during the late exponential and stationary growth phases. Transcription was also upregulated during the initial hours following phagocytosis by phagocytic cells. In contrast to vcgA and vcgC, the vcgB gene is conserved in the porcine VapB-encoding plasmid, as well as in pathogenic mycobacteria. The coordinated regulation of vcgB and vapA, transcription of vcgB following phagocytosis and conservation of vcgB in pathogenic mycobacteria indicate a role for vcgB and the vcg genes in the virulence of R. equi.


Subject(s)
Bacterial Proteins/biosynthesis , Gene Expression Regulation, Bacterial , Plasmids , Rhodococcus equi/pathogenicity , Virulence Factors/biosynthesis , Animals , Cell Line , Conserved Sequence , Genes, Bacterial , Genomic Islands , Hydrogen-Ion Concentration , Kinetics , Macrophages/immunology , Macrophages/microbiology , Mice , Mycobacterium/genetics , Operon , Phagocytosis , Promoter Regions, Genetic , RNA Stability , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Rhodococcus equi/genetics , Rhodococcus equi/growth & development , Temperature , Time Factors , Transcription Initiation Site , Transcription, Genetic , Virulence
16.
Infect Immun ; 79(5): 2098-111, 2011 May.
Article in English | MEDLINE | ID: mdl-21383050

ABSTRACT

Rhodococcus equi is an intracellular pathogen which causes pneumonia in young horses and in immunocompromised humans. R. equi arrests phagosome maturation in macrophages at a prephagolysosome stage and grows inside a privileged compartment. Here, we show that, in murine macrophages activated with gamma interferon and lipopolysaccharide, R. equi does not multiply but stays viable for at least 24 h. Whereas infection control of other intracellular pathogens by activated macrophages is executed by enhanced phagosome acidification or phagolysosome formation, by autophagy or by the interferon-inducible GTPase Irgm1, none of these mechanisms seems to control R. equi infection. Growth control by macrophage activation is fully mimicked by treatment of resting macrophages with nitric oxide donors, and inhibition of bacterial multiplication by either activation or nitric oxide donors is annihilated by cotreatment of infected macrophages with ferrous sulfate. Transcriptional analysis of the R. equi iron-regulated gene iupT demonstrates that intracellular R. equi encounters iron stress in activated, but not in resting, macrophages and that this stress is relieved by extracellular addition of ferrous sulfate. Our results suggest that nitric oxide is central to the restriction of bacterial access to iron in activated macrophages.


Subject(s)
Actinomycetales Infections/immunology , Iron/immunology , Macrophage Activation/immunology , Macrophages/microbiology , Nitric Oxide/immunology , Actinomycetales Infections/metabolism , Animals , Blotting, Western , Iron/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nitric Oxide/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhodococcus equi/growth & development , Rhodococcus equi/immunology , Rhodococcus equi/metabolism
17.
Microbes Infect ; 13(5): 438-46, 2011 May.
Article in English | MEDLINE | ID: mdl-21262372

ABSTRACT

Rhodococcus equi is an emerging opportunistic human pathogen associated with immunosuppressed people, especially those infected with the human immunodeficiency virus (HIV). This pathogen resides primarily within lung macrophages of infected patients, which may explain in part its ability to escape normal pulmonary defense mechanisms. Despite numerous studies as a pulmonary pathogen in foals, where a plasmid seems to play an important role in virulence, information on the pathogenesis of this pathogen in humans is still scarce. In this study, fluorescence microscopy and vancomycin protection assays were used to investigate the ability of R. equi human isolates to adhere to and to invade the human alveolar epithelial cell line A549. Our findings indicate that some R. equi clinical strains are capable of adhering, entering and surviving within the alveolar cell line, which may contribute to the pathogen persistence in lung tissues.


Subject(s)
Actinomycetales Infections/microbiology , Epithelial Cells/microbiology , Pulmonary Alveoli/microbiology , Rhodococcus equi/growth & development , Rhodococcus equi/pathogenicity , Bacterial Adhesion , Cell Line , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Pulmonary Alveoli/cytology , Pulmonary Alveoli/ultrastructure , Rhodococcus equi/genetics , Rhodococcus equi/isolation & purification , Virulence
18.
PLoS Genet ; 6(9): e1001145, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20941392

ABSTRACT

We report the genome of the facultative intracellular parasite Rhodococcus equi, the only animal pathogen within the biotechnologically important actinobacterial genus Rhodococcus. The 5.0-Mb R. equi 103S genome is significantly smaller than those of environmental rhodococci. This is due to genome expansion in nonpathogenic species, via a linear gain of paralogous genes and an accelerated genetic flux, rather than reductive evolution in R. equi. The 103S genome lacks the extensive catabolic and secondary metabolic complement of environmental rhodococci, and it displays unique adaptations for host colonization and competition in the short-chain fatty acid-rich intestine and manure of herbivores--two main R. equi reservoirs. Except for a few horizontally acquired (HGT) pathogenicity loci, including a cytoadhesive pilus determinant (rpl) and the virulence plasmid vap pathogenicity island (PAI) required for intramacrophage survival, most of the potential virulence-associated genes identified in R. equi are conserved in environmental rhodococci or have homologs in nonpathogenic Actinobacteria. This suggests a mechanism of virulence evolution based on the cooption of existing core actinobacterial traits, triggered by key host niche-adaptive HGT events. We tested this hypothesis by investigating R. equi virulence plasmid-chromosome crosstalk, by global transcription profiling and expression network analysis. Two chromosomal genes conserved in environmental rhodococci, encoding putative chorismate mutase and anthranilate synthase enzymes involved in aromatic amino acid biosynthesis, were strongly coregulated with vap PAI virulence genes and required for optimal proliferation in macrophages. The regulatory integration of chromosomal metabolic genes under the control of the HGT-acquired plasmid PAI is thus an important element in the cooptive virulence of R. equi.


Subject(s)
Evolution, Molecular , Genes, Bacterial/genetics , Rhodococcus equi/pathogenicity , Adaptation, Physiological/genetics , Animals , Chromosomes, Bacterial/genetics , Gene Duplication/genetics , Gene Regulatory Networks/genetics , Gene Transfer, Horizontal/genetics , Genetic Loci/genetics , Genomics , Intracellular Space/microbiology , Kinetics , Macrophages/cytology , Macrophages/microbiology , Mice , Mutation/genetics , Phylogeny , Plasmids/genetics , Rhodococcus equi/genetics , Rhodococcus equi/growth & development , Rhodococcus equi/ultrastructure , Virulence/genetics
19.
Vet Immunol Immunopathol ; 130(1-2): 102-6, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19211153

ABSTRACT

Defensins are small effector molecules of the innate immune system, synthesised by various organisms including plants and animals. The peptides act as endogenous antibiotics with an antimicrobial activity against a broad spectrum of microbes including bacteria, fungi and viruses. alpha-Defensins are a subgroup of the defensin family, their synthesis is limited to some tissues and furthermore to some mammalian species including the horse. Equine DEFA1 is an enteric alpha-defensin exclusively produced in Paneth cells. The peptide showed an activity against a broad spectrum of microbes, but typical pathogens of the horse were not included in the previous antimicrobial studies. Here, we report the antibacterial properties of DEFA1 against clinical isolates of typical horse pathogens including Rhodococcus equi, various streptococci strains, Salmonella choleraesuis, and Pasteurella multocida. The recombinantly expressed DEFA1 peptide exerted potent activity against these pathogenic bacteria. The highest susceptibility showed R. equi. Three genetically different strains of R. equi were killed at low micromolar concentrations, comparable with conventionally used antibiotics.


Subject(s)
Actinomycetales Infections/veterinary , Anti-Infective Agents/pharmacology , Horse Diseases/microbiology , Rhodococcus equi/drug effects , alpha-Defensins/pharmacology , Actinomycetales Infections/drug therapy , Actinomycetales Infections/microbiology , Amino Acid Sequence , Animals , Horse Diseases/drug therapy , Horses , Microbial Sensitivity Tests/veterinary , Molecular Sequence Data , Pasteurella multocida/drug effects , Pasteurella multocida/growth & development , Recombinant Proteins/pharmacology , Rhodococcus equi/growth & development , Rhodococcus equi/isolation & purification , Salmonella/drug effects , Salmonella/growth & development , Streptococcus/drug effects , Streptococcus/growth & development
20.
Vet Microbiol ; 135(3-4): 334-45, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19019579

ABSTRACT

Rhodococcus equi is one of the most important causes of mortality in foals between 1 and 6 months of age. Although rare, infection also occurs in a variety of other mammals including humans, often following immunosuppression of various causes. Secreted proteins are known to mediate important pathogen-host interactions and consequently are favored candidates for vaccine development as they are the most easily accessible microbial antigens to the immune system. Here, we describe the results of a proteomic analysis based on SDS-PAGE, immunoblot and mass spectrometry, which was carried out aiming the identification of secreted proteins that are differently expressed at 30 degrees C versus 37 degrees C and at mid-exponential versus early-stationary growth phase and antigenic proteins from R. equi ATCC 33701. A total of 48 proteins was identified regardless of growth conditions. The cholesterol oxidase ChoE appears to be the major secretory protein. Moreover, four proteins revealed high homologies with the mycolyl transferases of the Ag85 complex from Mycobacterium tuberculosis. The sequence analysis predicted that 24 proteins are transported by a signal peptide-dependent pathway. Moreover, five antigenic proteins of R. equi were identified by immunoblot, including a novel strongly immunoreactive protein of unknown function. In conclusion, the elucidation of the secretome of R. equi identified several proteins with different biological functions and a new candidate for developing vaccines against R. equi infection in horse.


Subject(s)
Actinomycetales Infections/veterinary , Bacterial Proteins/genetics , Horse Diseases/microbiology , Proteome , Rhodococcus equi/genetics , Amino Acid Sequence , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Bacterial Proteins/immunology , Bacterial Proteins/isolation & purification , DNA Primers , Electrophoresis, Polyacrylamide Gel , Enzymes/chemistry , Horses , Immunoblotting , Mass Spectrometry , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Rhodococcus equi/growth & development , Rhodococcus equi/immunology , Sequence Homology, Amino Acid , Temperature , Trypsin
SELECTION OF CITATIONS
SEARCH DETAIL
...