1
|
Godoi APDS, Sobral GG, da Silva Vieira JC, Carneiro GF, Conceição FR, da Silva ER, Mendonça M. Phenotypical and molecular characterization of Rhodococcus equi isolated from foals in the Agreste region of Pernambuco - Brazil. Braz J Microbiol 2025; 56:1321-1331. [PMID: 40048142 PMCID: PMC12095708 DOI: 10.1007/s42770-025-01640-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/13/2025] [Indexed: 05/22/2025] Open
Abstract
Equine rhodococcosis is caused by Rhodococcus equi, an intracellular coccobacillus whose main virulence factor is a plasmid that harbors genes encoding proteins from the Vap family, with the vapA gene being the most important in equine isolates. Furthermore, other factors observed in R. equi strains, such as antimicrobial resistance and biofilm production, may represent significant challenges in the treatment of affected animals. The objective of this study was to characterize four isolates of R. equi from foals in the state of Pernambuco, Brazil. All isolates were identified as R. equi through biochemical tests, amplification of the choE gene, and sequencing of 16 S rRNA. PCR analysis revealed that three isolates were positive for the plasmid virulence genes (vapA, -C, -D, -E, -F, -H and traA), although vapD was absent in one of the three isolates. One isolate did not present any virulence genes, possibly due to the loss of the plasmid after repeated passages at 37ºC. In the antimicrobial susceptibility test, all isolates were susceptible to erythromycin, clarithromycin, azithromycin, rifampicin, gentamicin, and doxycycline. However, all isolates were capable of forming biofilms, with moderate biofilm formation in isolates Rhodo1 and Rhodo2, and weak biofilm formation in isolates Rhodo3 and Rhodo4, which may be associated with increased antimicrobial tolerance. This molecular characterization demonstrated, for the first time, the presence of the virulence plasmid in R. equi isolates from foals in Northeast Brazil, as well as their capacity for biofilm formation.
Collapse
Affiliation(s)
- Alysson Paulo Dos Santos Godoi
- Departamento de Medicina Veterinária, Universidade Federal Rural de Pernambuco, Rua Dom Manuel de Medeiros, Recife, Pernambuco, 52171-900, Brazil
- Programa de Pós-Graduação em Sanidade e Reprodução de Animais de Produção, Universidade Federal do Agreste de Pernambuco, Avenida Bom Pastor, Boa Vista, Garanhuns, 55292-270, Pernambuco, Brazil
| | - Gilvannya Gonçalves Sobral
- Departamento de Medicina Veterinária, Universidade Federal Rural de Pernambuco, Rua Dom Manuel de Medeiros, Recife, Pernambuco, 52171-900, Brazil
| | - Júlio César da Silva Vieira
- Programa de Pós-Graduação em Sanidade e Reprodução de Animais de Produção, Universidade Federal do Agreste de Pernambuco, Avenida Bom Pastor, Boa Vista, Garanhuns, 55292-270, Pernambuco, Brazil
| | - Gustavo Ferrer Carneiro
- Departamento de Medicina Veterinária, Universidade Federal Rural de Pernambuco, Rua Dom Manuel de Medeiros, Recife, Pernambuco, 52171-900, Brazil
| | - Fabricio Rochedo Conceição
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, Campus Capão do Leão, Pelotas, 96010-610, Rio Grande do Sul, Brazil
| | - Elizabete Rodrigues da Silva
- Programa de Pós-Graduação em Sanidade e Reprodução de Animais de Produção, Universidade Federal do Agreste de Pernambuco, Avenida Bom Pastor, Boa Vista, Garanhuns, 55292-270, Pernambuco, Brazil
| | - Marcelo Mendonça
- Programa de Pós-Graduação em Sanidade e Reprodução de Animais de Produção, Universidade Federal do Agreste de Pernambuco, Avenida Bom Pastor, Boa Vista, Garanhuns, 55292-270, Pernambuco, Brazil.
| |
Collapse
|
2
|
Vazquez-Boland JA, Val-Calvo J, Duquesne F, Decorosi F, Viti C, Petry S, Scortti M. Rhodococcus parequi sp. nov., a new species isolated from equine farm soil closely related to the pathogen Rhodococcus equi. Int J Syst Evol Microbiol 2025; 75:006679. [PMID: 40063668 PMCID: PMC11893733 DOI: 10.1099/ijsem.0.006679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/02/2025] [Indexed: 03/14/2025] Open
Abstract
We present the description of the new species, Rhodococcus parequi, found during phylogenomic investigations of a global collection of strains identified as Rhodococcus (Prescottella) equi. Strain PAM 2766T was isolated from horse-breeding farm soil in Normandy, France, and was indistinguishable from R. equi based on the usual identification tests. Whole-genome phylogenetic analyses located PAM 2766T in the same Rhodococcus sublineage as R. equi, together with Rhodococcus agglutinans, Rhodococcus defluvii, Rhodococcus soli, Rhodococcus subtropicus, Rhodococcus spongiicola and Rhodococcus xishaensis. PAM 2766T is most closely related to, but sufficiently distinct from, R. equi DSM 20307 T to be considered a separate species. The average nt identity (ANI) and average aa identity (AAI) values are 88.60% and 92.35, respectively, well below the species cutoff. The PAM 2766T draft genome is ~5.3 Mb in size with 68.98% G+C mol content. PAM 2766T is strictly aerobic and non-motile and produces smooth, creamy to buff-coloured colonies very similar to those of R. equi. It phenotypically differs from the latter by the ability to grow at 5 °C, a strongly positive urease test at 24 h and specificities in the carbon and nitrogen source utilization profile as determined by phenotype microarray screens. Our data indicate that PAM 2766T belongs to a novel species, for which the name Rhodococcus parequi sp. nov. is proposed. R. parequi was avirulent in macrophage infection assays and is assumed to be non-pathogenic. The type strain is PAM 2766T (=CETC 30995T=NCTC 14987T).
Collapse
Affiliation(s)
- José A. Vazquez-Boland
- Microbial Pathogenomics Laboratory, Edinburgh Medical School (Biomedical Sciences), University of Edinburgh, Edinburgh, UK
| | - Jorge Val-Calvo
- Microbial Pathogenomics Laboratory, Edinburgh Medical School (Biomedical Sciences), University of Edinburgh, Edinburgh, UK
| | - Fabien Duquesne
- ANSES, Laboratory for Animal Health, Physiopathology and Epidemiology of Equine Diseases Unit, Goustranville, France
| | - Francesca Decorosi
- Department of Agriculture, Food, Environment and Forestry, University of Florence, Florence, Italy
| | - Carlo Viti
- Department of Agriculture, Food, Environment and Forestry, University of Florence, Florence, Italy
| | - Sandrine Petry
- ANSES, Laboratory for Animal Health, Physiopathology and Epidemiology of Equine Diseases Unit, Goustranville, France
| | - Mariela Scortti
- Microbial Pathogenomics Laboratory, Edinburgh Medical School (Biomedical Sciences), University of Edinburgh, Edinburgh, UK
| |
Collapse
|
3
|
Hu B, Gao S, Zhang H, Li Q, Li G, Zhang S, Xing Y, Huang Y, Han S, Tian Y, Zhang W, He H. Whole-genome sequencing and pathogenicity analysis of Rhodococcus equi isolated in horses. BMC Vet Res 2024; 20:362. [PMID: 39129003 PMCID: PMC11318318 DOI: 10.1186/s12917-024-04167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/01/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Rhodococcus equi (R. equi) is a Gram-positive zoonotic pathogen that frequently leads to illness and death in young horses (foals). This study presents the complete genome sequence of R. equi strain BJ13, which was isolated from a thoroughbred racehorse breeding farm in Beijing, China. RESULTS The BJ13 genome has a length of 5.30 Mb and consists of a complete chromosome and a plasmid measuring 5.22 Mb and 0.08 Mb, respectively. We predicted 4,929 coding gene open reading frames, along with 52 tRNAs and 12 rRNAs. Through analysis of mobile genetic elements, we identified 6 gene islands and 1 prophage gene. Pathogenic system analysis predicted the presence of 418 virulence factors and 225 drug resistance genes. Secretion system analysis revealed the prediction of 297 secreted proteins and 1,106 transmembrane proteins. BJ13 exhibits genomic features, virulence-associated genes, potential drug resistance, and a virulence plasmid structure that may contribute to the evolution of its pathogenicity. Lastly, the pathogenicity of the isolated strain was assessed through animal experiments, which resulted in inflammatory reactions or damage in the lungs, liver, and spleen of mice. Moreover, by the 7th day post-infection, the mortality rate of the mice reached 50.0%, indicating complex immune regulatory mechanisms, including overexpression of IL-10 and increased production of pro-inflammatory cytokines like TNF-α. These findings validate the strong pathogenicity of the isolated strain and provide insights for studying the pathogenic mechanisms of Rhodococcus equi infection. CONCLUSIONS The complete genome sequence of R. equi strain BJ13 provides valuable insights into its genomic characteristics, virulence potential, drug resistance, and secretion systems. The strong pathogenicity observed in animal experiments underscores the need for further investigation into the pathogenic mechanisms of R. equi infection.
Collapse
Affiliation(s)
- Bin Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sichao Gao
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Anhui University of Science and Technology, Huainan, China
| | - Hao Zhang
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiaoqiao Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Anhui University of Science and Technology, Huainan, China
| | - Gaojian Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuairan Zhang
- College of Shenyang Institute of Technology, Shenyang, Liaoning, China
| | - Yanan Xing
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanyi Huang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuyi Han
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying Tian
- Beijing Wildlife Rescue and Rehabilitation Center, Beijing, China
| | - Wei Zhang
- Beijing Wildlife Rescue and Rehabilitation Center, Beijing, China
| | - Hongxuan He
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.
| |
Collapse
|
4
|
Miranda-CasoLuengo R, Yerlikaya Z, Luo H, Cheng C, Blanco A, Haas A, Meijer WG. The N-terminal domain is required for cell surface localisation of VapA, a member of the Vap family of Rhodococcus equi virulence proteins. PLoS One 2024; 19:e0298900. [PMID: 38421980 PMCID: PMC10903876 DOI: 10.1371/journal.pone.0298900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Rhodococcus equi pneumonia is an important cause of mortality in foals worldwide. Virulent equine isolates harbour an 80-85kb virulence plasmid encoding six virulence-associated proteins (Vaps). VapA, the main virulence factor of this intracellular pathogen, is known to be a cell surface protein that creates an intracellular niche for R. equi growth. In contrast, VapC, VapD and VapE are secreted into the intracellular milieu. Although these Vaps share very high degree of sequence identity in the C-terminal domain, the N-terminal domain (N-domain) of VapA is distinct. It has been proposed that this domain plays a role in VapA surface localization but no direct experimental data provides support to such hypothesis. In this work, we employed R. equi 103S harbouring an unmarked deletion of vapA (R. equi ΔvapA) as the genetic background to express C-terminal Strep-tagged Vap-derivatives integrated in the chromosome. The surface localization of these proteins was assessed by flow cytometry using the THE2122;-NWSHPQFEK Tag FITC-antibody. We show that VapA is the only cell surface Vap encoded in the virulence plasmid. We present compelling evidence for the role of the N-terminal domain of VapA on cell surface localization using fusion proteins in which the N-domain of VapD was exchanged with the N-terminus of VapA. Lastly, using an N-terminally Strep-tagged VapA, we found that the N-terminus of VapA is exposed to the extracellular environment. Given the lack of a lipobox in VapA and the exposure of the N-terminal Strep-tag, it is possible that VapA localization on the cell surface is mediated by interactions between the N-domain and components of the cell surface. We discuss the implications of this work on the light of the recent discovery that soluble recombinant VapA added to the extracellular medium functionally complement the loss of VapA.
Collapse
Affiliation(s)
- Raúl Miranda-CasoLuengo
- UCD School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Zeynep Yerlikaya
- UCD School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
- Department of Microbiology, School of Veterinary Medicine, Firat University, Elazığ, Türkiye
| | - Haixia Luo
- UCD School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Cheng Cheng
- UCD School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Alfonso Blanco
- Flow Cytometry Core Technology, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Albert Haas
- Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Wim G. Meijer
- UCD School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Haubenthal T, Hansen P, Krämer I, Gindt M, Jünger-Leif A, Utermöhlen O, Haas A. Specific preadaptations of Rhodococcus equi cooperate with its Virulence-associated protein A during macrophage infection. Mol Microbiol 2023; 119:285-301. [PMID: 36627747 DOI: 10.1111/mmi.15026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023]
Abstract
Gram-positive Rhodococcus equi (Prescotella equi) is a lung pathogen of foals and immunocompromised humans. Intra-macrophage multiplication requires production of the bacterial Virulence-associated protein A (VapA) which is released into the phagosome lumen. VapA pH-neutralizes intracellular compartments allowing R. equi to multiply in an atypical macrophage phagolysosome. Here, we show that VapA does not support intra-macrophage growth of several other bacterial species demonstrating that only few bacteria have the specific preadaptations needed to profit from VapA. We show that the closest relative of R. equi, environmental Rhodococcus defluvii (Prescotella defluvii), does not multiply in macrophages at 37°C even when VapA is present because of its thermosensitivity but it does so once the infection temperature is lowered providing rare experimental evidence for 'thermal restriction'. Using growth experiments with isolated macrophage lysosomes and modified infection schemes we provide evidence that R. equi resists the attack by phagolysosome contents at low pH for several hours. During this time, R. equi produces and secretes VapA which enables it to grow at the expense of lysosome constituents. We present arguments that, under natural infection conditions, R. equi is VapA-less during the initial encounter with the host. This has important implications for vaccine development.
Collapse
Affiliation(s)
| | - Philipp Hansen
- Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Ina Krämer
- Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Mélanie Gindt
- Institute for Cell Biology, University of Bonn, Bonn, Germany
| | | | - Olaf Utermöhlen
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Germany
| | - Albert Haas
- Institute for Cell Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
6
|
GAPDH, rhbC, and vapA gene expression in Rhodococcus equi cultured under different iron concentrations. Microb Pathog 2019; 139:103885. [PMID: 31790793 DOI: 10.1016/j.micpath.2019.103885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 11/07/2019] [Accepted: 11/21/2019] [Indexed: 11/22/2022]
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.
Collapse
|
7
|
Clonal Confinement of a Highly Mobile Resistance Element Driven by Combination Therapy in Rhodococcus equi. mBio 2019; 10:mBio.02260-19. [PMID: 31615959 PMCID: PMC6794481 DOI: 10.1128/mbio.02260-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
MDR clades arise upon acquisition of resistance traits, but the determinants of their clonal expansion remain largely undefined. Taking advantage of the unique features of Rhodococcus equi infection control in equine farms, involving the same dual antibiotic treatment since the 1980s (a macrolide and rifampin), this study sheds light into the determinants of multiresistance clonality and the importance of combination therapy in limiting the dissemination of mobile resistance elements. Clinically effective therapeutic alternatives against R. equi foal pneumonia are currently lacking, and the identified macrolide-rifampin MDR clone 2287 has serious implications. Still at early stages of evolution and local spread, R. equi 2287 may disseminate globally, posing a significant threat to the equine industry and, also, public health due to the risk of zoonotic transmission. The characterization of the 2287 clone and its resistance determinants will enable targeted surveillance and control interventions to tackle the emergence of MDR R. equi. Antibiotic use has been linked to changes in the population structure of human pathogens and the clonal expansion of multidrug-resistant (MDR) strains among healthcare- and community-acquired infections. Here we present a compelling example in a veterinary pathogen, Rhodococcus equi, the causative agent of a severe pulmonary infection affecting foals worldwide. We show that the erm(46) gene responsible for emerging macrolide resistance among equine R. equi isolates in the United States is part of a 6.9-kb transposable element, TnRErm46, actively mobilized by an IS481 family transposase. TnRErm46 is carried on an 87-kb conjugative plasmid, pRErm46, transferable between R. equi strains at frequencies up to 10−3. The erm(46) gene becomes stabilized in R. equi by pRErm46’s apparent fitness neutrality and wholesale TnRErm46 transposition onto the host genome. This includes the conjugally exchangeable pVAPA virulence plasmid, enabling the possibility of cotransfer of two essential traits for survival in macrolide-treated foals in a single mating event. Despite its high horizontal transfer potential, phylogenomic analyses show that erm(46) is paradoxically confined to a specific R. equi clone, 2287. R. equi 2287 also carries a unique rpoBS531F mutation conferring high-level resistance to rifampin, systematically administered together with macrolides against rhodococcal pneumonia on equine farms. Our data illustrate that under sustained combination therapy, several independent “founder” genetic events are concurrently required for resistance, limiting not only its emergence but also, crucially, horizontal spread, ultimately determining multiresistance clonality.
Collapse
|
8
|
Vázquez‐Boland JA, Meijer WG. The pathogenic actinobacterium Rhodococcus equi: what's in a name? Mol Microbiol 2019; 112:1-15. [PMID: 31099908 PMCID: PMC6852188 DOI: 10.1111/mmi.14267] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2019] [Indexed: 12/17/2022]
Abstract
Rhodococcus equi is the only recognized animal pathogenic species within an extended genus of metabolically versatile Actinobacteria of considerable biotechnological interest. Best known as a horse pathogen, R. equi is commonly isolated from other animal species, particularly pigs and ruminants, and causes severe opportunistic infections in people. As typical in the rhodococci, R. equi niche specialization is extrachromosomally determined, via a conjugative virulence plasmid that promotes intramacrophage survival. Progress in the molecular understanding of R. equi and its recent rise as a novel paradigm of multihost adaptation has been accompanied by an unusual nomenclatural instability, with a confusing succession of names: "Prescottia equi", "Prescotella equi", Corynebacterium hoagii and Rhodococcus hoagii. This article reviews current advances in the genomics, biology and virulence of this pathogenic actinobacterium with a unique mechanism of plasmid-transferable animal host tropism. It also discusses the taxonomic and nomenclatural issues around R. equi in the light of recent phylogenomic evidence that confirms its membership as a bona fide Rhodococcus.
Collapse
Affiliation(s)
- José A. Vázquez‐Boland
- Microbial Pathogenesis Group, Edinburgh Medical School (Biomedical Sciences – Infection Medicine)University of EdinburghChancellor's Building, Little France campusEdinburghEH16 4SBUK
| | - Wim G. Meijer
- UCD School of Biomolecular and Biomedical ScienceUniversity College DublinDublin 4Ireland
| |
Collapse
|
9
|
Willingham-Lane JM, Coulson GB, Hondalus MK. Identification of a VapA virulence factor functional homolog in Rhodococcus equi isolates housing the pVAPB plasmid. PLoS One 2018; 13:e0204475. [PMID: 30286098 PMCID: PMC6171844 DOI: 10.1371/journal.pone.0204475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/07/2018] [Indexed: 11/30/2022] Open
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.
Collapse
Affiliation(s)
| | - Garry B. Coulson
- Department of Infectious Disease, University of Georgia, Athens, Georgia, United States of America
| | - Mary K. Hondalus
- Department of Infectious Disease, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
10
|
Kalinowski M, Grądzki Z, Jarosz Ł, Adaszek Ł. Molecular analysis of the chromosomal 16S rRNA gene and vapA plasmid gene of Polish field strains of R. equi. PLoS One 2018; 13:e0204024. [PMID: 30252885 PMCID: PMC6155501 DOI: 10.1371/journal.pone.0204024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 08/31/2018] [Indexed: 12/29/2022] Open
Abstract
Rhodococcus equi (R. hoagii) is an opportunistic pathogen commonly found in foals up to 6 months old and animal environment. The R. equi genome contains genetically stable chromosomal DNA and an 80–90 kb plasmid containing vapA gene, responsible for virulence. Most reports from around the world focus on the determination of R. equi plasmid profiles. Few studies have attempted to determine differences in nucleotide sequences between virulent strains of R. equi isolated from foals and breeding environment. The aim of the study was to perform a molecular analysis of a fragment of the chromosomal gene encoding the 16S rRNA subunit and the vapA plasmid gene of virulent R. equi strains isolated on Polish studs from foals and from the breeding environment of horses. The sequencing method was used to compare the primary structure of fragments of the chromosomal and plasmid DNA of the virulent R. equi strains. The sequences of 22 clinical and 18 environmental R. equi isolates were compared with the sequences of the gene fragments of reference strains available in the NCBI GenBank database. All sequenced 16S rRNA amplicons of Polish field strains were identical and showed 99.5% similarity to the four randomly selected sequences of this gene fragment in the GenBank database. The results confirm that fragments of the 16S rRNA gene of R. equi strains are highly conserved and do not undergo variation in field conditions. Analysis of the sequencing results for the vapA gene fragment of the strains used in our study revealed two polymorphic variants and clear differences between the sequences of strains isolated from foals and from soil samples. Presumably, R. equi strains present in the breeding environment are more exposed than clinical strains to adverse external factors. This may result in changes in the DNA sequence due to natural selection.
Collapse
Affiliation(s)
- Marcin Kalinowski
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
- * E-mail:
| | - Zbigniew Grądzki
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Łukasz Jarosz
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Łukasz Adaszek
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| |
Collapse
|
11
|
Wright LM, Carpinone EM, Bennett TL, Hondalus MK, Starai VJ. VapA of Rhodococcus equi binds phosphatidic acid. Mol Microbiol 2017; 107:428-444. [PMID: 29205554 DOI: 10.1111/mmi.13892] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 12/30/2022]
Abstract
Rhodococcus equi is a multihost, facultative intracellular bacterial pathogen that primarily causes pneumonia in foals less than six months in age and immunocompromised people. Previous studies determined that the major virulence determinant of R. equi is the surface bound virulence associated protein A (VapA). The presence of VapA inhibits the maturation of R. equi-containing phagosomes and promotes intracellular bacterial survival, as determined by the inability of vapA deletion mutants to replicate in host macrophages. While the mechanism of action of VapA remains elusive, we show that soluble recombinant VapA32-189 both rescues the intramacrophage replication defect of a wild type R. equi strain lacking the vapA gene and enhances the persistence of nonpathogenic Escherichia coli in macrophages. During macrophage infection, VapA was observed at both the bacterial surface and at the membrane of the host-derived R. equi containing vacuole, thus providing an opportunity for VapA to interact with host constituents and promote alterations in phagolysosomal function. In support of the observed host membrane binding activity of VapA, we also found that rVapA32-189 interacted specifically with liposomes containing phosphatidic acid in vitro. Collectively, these data demonstrate a lipid binding property of VapA, which may be required for its function during intracellular infection.
Collapse
Affiliation(s)
- Lindsay M Wright
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Emily M Carpinone
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Terry L Bennett
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Mary K Hondalus
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Vincent J Starai
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.,Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
12
|
Berghaus LJ, Giguère S, Bordin AI, Cohen ND. Effects of priming with cytokines on intracellular survival and replication of Rhodococcus equi in equine macrophages. Cytokine 2017; 102:7-11. [PMID: 29245049 DOI: 10.1016/j.cyto.2017.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 11/24/2022]
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)-1β, 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-1β 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-1β or IL-10 were detrimental.
Collapse
Affiliation(s)
- Londa J Berghaus
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Steeve Giguère
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| | - Angela I Bordin
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Noah D Cohen
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
13
|
MacArthur I, Anastasi E, Alvarez S, Scortti M, Vázquez-Boland JA. Comparative Genomics of Rhodococcus equi Virulence Plasmids Indicates Host-Driven Evolution of the vap Pathogenicity Island. Genome Biol Evol 2017; 9:1241-1247. [PMID: 28369330 PMCID: PMC5434932 DOI: 10.1093/gbe/evx057] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2017] [Indexed: 01/16/2023] Open
Abstract
The conjugative virulence plasmid is a key component of the Rhodococcus equi accessory genome essential for pathogenesis. Three host-associated virulence plasmid types have been identified: the equine pVAPA and porcine pVAPB circular variants, and the linear pVAPN found in bovine (ruminant) isolates. We recently characterized the R. equi pangenome (Anastasi E, et al. 2016. Pangenome and phylogenomic analysis of the pathogenic actinobacterium Rhodococcus equi. Genome Biol Evol. 8:3140–3148.) and we report here the comparative analysis of the virulence plasmid genomes. Plasmids within each host-associated type were highly similar despite their diverse origins. Variation was accounted for by scattered single nucleotide polymorphisms and short nucleotide indels, while larger indels—mostly in the plasticity region near the vap pathogencity island (PAI)—defined plasmid genomic subtypes. Only one of the plasmids analyzed, of pVAPN type, was exceptionally divergent due to accumulation of indels in the housekeeping backbone. Each host-associated plasmid type carried a unique PAI differing in vap gene complement, suggesting animal host-specific evolution of the vap multigene family. Complete conservation of the vap PAI was observed within each host-associated plasmid type. Both diversity of host-associated plasmid types and clonality of specific chromosomal-plasmid genomic type combinations were observed within the same R. equi phylogenomic subclade. Our data indicate that the overall strong conservation of the R. equi host-associated virulence plasmids is the combined result of host-driven selection, lateral transfer between strains, and geographical spread due to international livestock exchanges.
Collapse
Affiliation(s)
- Iain MacArthur
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Elisa Anastasi
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Sonsiray Alvarez
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Mariela Scortti
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Medical School (Biomedical Sciences), University of Edinburgh, Edinburgh, United Kingdom
| | - José A Vázquez-Boland
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Medical School (Biomedical Sciences), University of Edinburgh, Edinburgh, United Kingdom.,Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
14
|
Sangkanjanavanich N, Kawai M, Kakuda T, Takai S. Rescue of an intracellular avirulent Rhodococcus equi replication defect by the extracellular addition of virulence-associated protein A. J Vet Med Sci 2017; 79:1323-1326. [PMID: 28690290 PMCID: PMC5573816 DOI: 10.1292/jvms.17-0350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
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.
Collapse
Affiliation(s)
- Nuttapone Sangkanjanavanich
- Laboratory of Animal Hygiene, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori 034-8628, Japan
| | - Masanori Kawai
- Laboratory of Animal Hygiene, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori 034-8628, Japan
| | - Tsutomu Kakuda
- Laboratory of Animal Hygiene, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori 034-8628, Japan
| | - Shinji Takai
- Laboratory of Animal Hygiene, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori 034-8628, Japan
| |
Collapse
|
15
|
Trevisani MM, Hanna ES, Oliveira AF, Cardoso SA, Roque-Barreira MC, Soares SG. Vaccination of Mice with Virulence-Associated Protein G (VapG) Antigen Confers Partial Protection against Rhodococcus equi Infection through Induced Humoral Immunity. Front Microbiol 2017; 8:857. [PMID: 28553279 PMCID: PMC5425581 DOI: 10.3389/fmicb.2017.00857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 04/27/2017] [Indexed: 11/13/2022] Open
Abstract
Rhodococcus equi is a facultative intracellular bacterium causing severe pyogranulomatous pneumonia, ulcerative enterocolitis, and mesenteric lymphadenopathy in foals aged less than 6 months. Less frequently, this pathogen affects various other species, such as pigs, cattle, cats, and even humans. Although rhodococcosis is treated with a combination of antimicrobial agents, resistance is developed in some cases, and thus, antimicrobial susceptibility must be monitored and managed. Considering these limitations of the current therapy and unavailability of a vaccine to prevent the disease, research is particularly focused on the development of an effective vaccine against rhodococcosis. Most vaccines undergoing development utilize the virulence-associated protein (Vap) A antigen, which was identified previously as a key virulence factor of R. equi. Nevertheless, other proteins, such as VapG, present in most virulent R. equi strains, are also encoded by vap genes located on the R. equi bacterial virulence plasmid. In the present study, we evaluated the effect of VapG immunization on the survival of R. equi-challenged mice. We used attenuated Salmonella as a carrier for VapG (Salmonella-vapG+), a procedure previously adopted to develop a VapA-based vaccine. We observed that vaccination with Salmonella-vapG+ induced both an increased IFN-γ, IL-12, and TNF-α production, and a decreased bacterial burden in organs of the R. equi-challenged mice. Nevertheless, Salmonella-vapG+ vaccination protected only 50% of the mice challenged with a lethal dose of R. equi. Interestingly, we observed an increased frequency of B cells in the spleen of Salmonella-vapG+-vaccinated mice and showed that Salmonella-vapG+-vaccinated R. equi-challenged B-cell-knockout mice did not reduce the bacterial burden. Given these results, we discussed the potential role of the humoral immune response induced by Salmonella-vapG+ vaccination in conferring protection against R. equi infection, as well as the employment of VapG antigen for obtaining hyperimmune plasma to prevent rhodoccocosis in young foals.
Collapse
Affiliation(s)
- Marcel M Trevisani
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São PauloSão Paulo, Brazil
| | - Ebert S Hanna
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São PauloSão Paulo, Brazil
| | - Aline F Oliveira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São PauloSão Paulo, Brazil
| | - Silvia A Cardoso
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São PauloSão Paulo, Brazil
| | - Maria C Roque-Barreira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São PauloSão Paulo, Brazil
| | - Sandro G Soares
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São PauloSão Paulo, Brazil
| |
Collapse
|
16
|
Influence of Plasmid Type on the Replication of Rhodococcus equi in Host Macrophages. mSphere 2016; 1:mSphere00186-16. [PMID: 27747295 PMCID: PMC5061997 DOI: 10.1128/msphere.00186-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/31/2016] [Indexed: 11/20/2022] Open
Abstract
The soil-dwelling, saprophytic actinomycete Rhodococcus equi is a multihost, facultative intracellular pathogen of macrophages. When inhaled by susceptible foals, it causes severe bronchopneumonia. It is also a pathogen of pigs, which may develop submaxillary lymphadenitis upon exposure. R. equi isolates obtained from foals and pigs possess conjugative plasmids housing a pathogenicity island (PAI) containing a novel family of genes of unknown function called the virulence-associated protein or vap family. The PAI regions of the equine and swine plasmids differ in vap gene composition, with equine isolates possessing six vap genes, including the major virulence determinant vapA, while the PAIs of swine isolates house vapB and five other unique vap genes. Possession of the pVAPA-type virulence plasmid by equine isolates bestows the capacity for intramacrophage replication essential for disease development in vivo. Swine isolates of R. equi are largely unstudied. Here, we show that R. equi isolates from pigs, carrying pVAPB-type plasmids, are able to replicate in a plasmid-dependent manner in macrophages obtained from a variety of species (murine, swine, and equine) and anatomical locations. Similarly, equine isolates carrying pVAPA-type plasmids are capable of replication in swine macrophages. Plasmid swapping between equine and swine strains through conjugation did not alter the intracellular replication capacity of the parental strain, indicating that coevolution of the plasmid and chromosome is not crucial for this attribute. These results demonstrate that while distinct plasmid types exist among R. equi isolates obtained from equine and swine sources, this tropism is not determined by host species-specific intramacrophage replication capabilities. IMPORTANCE This work greatly advances our understanding of the opportunistic pathogen Rhodococcus equi, a disease agent of animals and immunocompromised people. Clinical isolates from diseased foals carry a conjugative virulence plasmid, pVAPA1037, that expresses Vap proteins, including VapA, essential for intramacrophage replication and virulence in vivo. The understudied R. equi isolates from pigs carry a related but different plasmid, pVAPB, expressing distinct Vap proteins, including VapB. In this work, we document for the first time that R. equi isolates carrying pVAPB-type plasmids are capable of intramacrophage replication. Moreover, we show that R. equi isolates carrying either plasmid type can replicate in both equine and swine macrophages, indicating that host species tropism is not due to species-specific intramacrophage replication capabilities defined by plasmid type. Furthermore, plasmid swapping between equine and swine strains did not alter intracellular replication capacity, indicating that coevolution of the plasmid and chromosome is not essential for intracellular growth.
Collapse
|
17
|
Witkowski L, Rzewuska M, Takai S, Kizerwetter-Świda M, Kita J. Molecular epidemiology of Rhodococcus equi in slaughtered swine, cattle and horses in Poland. BMC Microbiol 2016; 16:98. [PMID: 27234339 PMCID: PMC4882809 DOI: 10.1186/s12866-016-0712-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 05/19/2016] [Indexed: 12/17/2022] Open
Abstract
Background Rhodococcus equi is an emerging zoonotic presumably foodborne pathogen. Since the data on the worldwide prevalence of R. equi in meat animals are scarce, the present study aimed to investigate the molecular epidemiology of R. equi in swine, cattle and horse carcasses intended for human consumption in Poland. Results Totally 1028 lymph node samples were examined. R. equi was isolated from 26.6 % (105/395) swine and 1.3 % (3/234) bovine healthy submaxillary lymph nodes. In horses, R. equi was isolated only from 0.5 % (1/198) samples of middle tracheo-branchiales lymph node while no lymphocentrum retropharyngeum sample was positive (0/198). The purulent lesions were observed only in 0.8 % swine submaxillary lymph nodes samples (3/398) and in two of them R. equi was detected. All bovine and most of swine isolates (98.1 %) were vapB-positive. 87.9 % of swine isolates carried 95-kb type 5 plasmid, 3.7 % type 1 and plasmid types: 4, 7, 10, 11, 21, 31 were carried by a single isolate (0.9 %). All bovine isolates carried VAPB type 26. Single horse isolate was vapA-positive and carried plasmid VAPA 85-kb type I. Conclusions The prevalence of vapB-positive R. equi in investigated healthy swine intended for human consumption was very high. Not only swine, but also even apparently healthy cattle or horse carcasses should be considered as a potential source of R. equi for humans, especially in countries where undercooked or raw beef or horsemeat is traditionally consumed.
Collapse
Affiliation(s)
- Lucjan Witkowski
- Laboratory of Veterinary Epidemiology and Economics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776, Warsaw, Poland.
| | - Magdalena Rzewuska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Shinji Takai
- Department of Animal Hygiene, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan
| | - Magdalena Kizerwetter-Świda
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Jerzy Kita
- Laboratory of Veterinary Epidemiology and Economics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776, Warsaw, Poland
| |
Collapse
|
18
|
Giguère S, Berghaus LJ, Lee EA. Activity of 10 antimicrobial agents against intracellular Rhodococcus equi. Vet Microbiol 2015; 178:275-8. [PMID: 26051479 DOI: 10.1016/j.vetmic.2015.05.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 05/20/2015] [Accepted: 05/22/2015] [Indexed: 11/17/2022]
Abstract
Studies with facultative intracellular bacterial pathogens have shown that evaluation of the bactericidal activity of antimicrobial agents against intracellular bacteria is more closely associated with in vivo efficacy than traditional in vitro susceptibility testing. The objective of this study was to determine the relative activity of 10 antimicrobial agents against intracellular Rhodococcus equi. Equine monocyte-derived macrophages were infected with virulent R. equi and exposed to erythromycin, clarithromycin, azithromycin, rifampin, ceftiofur, gentamicin, enrofloxacin, vancomycin, imipenem, or doxycycline at concentrations achievable in plasma at clinically recommended dosages in foals. The number of intracellular R. equi was determined 48h after infection by counting colony forming units (CFUs). The number of R. equi CFUs in untreated control wells were significantly higher than those of monolayers treated with antimicrobial agents. Numbers of R. equi were significantly lower in monolayers treated with enrofloxacin followed by those treated with gentamicin, and vancomycin, when compared to monolayers treated with other antimicrobial agents. Numbers of R. equi in monolayers treated with doxycycline were significantly higher than those of monolayers treated with other antimicrobial agents. Differences in R. equi CFUs between monolayers treated with other antimicrobial agents were not statistically significant. Enrofloxacin, gentamicin, and vancomycin are the most active drugs in equine monocyte-derived macrophages infected with R. equi. Additional studies will be needed to determine if these findings correlate with in vivo efficacy.
Collapse
Affiliation(s)
- Steeve Giguère
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States.
| | - Londa J Berghaus
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States
| | - Elise A Lee
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville FL 32610, United States
| |
Collapse
|
19
|
Transcriptome reprogramming by plasmid-encoded transcriptional regulators is required for host niche adaption of a macrophage pathogen. Infect Immun 2015; 83:3137-45. [PMID: 26015480 DOI: 10.1128/iai.00230-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/18/2015] [Indexed: 11/20/2022] Open
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.
Collapse
|
20
|
An Invertron-Like Linear Plasmid Mediates Intracellular Survival and Virulence in Bovine Isolates of Rhodococcus equi. Infect Immun 2015; 83:2725-37. [PMID: 25895973 DOI: 10.1128/iai.00376-15] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/16/2015] [Indexed: 12/16/2022] Open
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.
Collapse
|
21
|
Okoko T, Blagova EV, Whittingham JL, Dover LG, Wilkinson AJ. Structural characterisation of the virulence-associated protein VapG from the horse pathogen Rhodococcus equi. Vet Microbiol 2015; 179:42-52. [PMID: 25746683 PMCID: PMC4518536 DOI: 10.1016/j.vetmic.2015.01.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 01/30/2015] [Accepted: 01/31/2015] [Indexed: 01/20/2023]
Abstract
The 3-dimensional structure of a Rhodococcus equi virulence protein was determined. VapG comprises a closed beta barrel domain preceded by a natively disordered region. The structures of VapB, VapD and VapG are closely superimposable. The VAP structures lack recognisable ligand or protein binding sites. Phagosome-induced conformational changes may be required for virulence.
Virulence and host range in Rhodococcus equi depends on the variable pathogenicity island of their virulence plasmids. Notable gene products are a family of small secreted virulence-associated proteins (Vaps) that are critical to intramacrophagic proliferation. Equine-adapted strains, which cause severe pyogranulomatous pneumonia in foals, produce a cell-associated VapA that is necessary for virulence, alongside five other secreted homologues. In the absence of biochemical insight, attention has turned to the structures of these proteins to develop a functional hypothesis. Recent studies have described crystal structures for VapD and a truncate of the VapA orthologue of porcine-adapted strains, VapB. Here, we crystallised the full-length VapG and determined its structure by molecular replacement. Electron density corresponding to the N-terminal domain was not visible suggesting that it is disordered. The protein core adopted a compact elliptical, anti-parallel β-barrel fold with β1–β2–β3–β8–β5–β6–β7–β4 topology decorated by a single peripheral α-helix unique to this family. The high glycine content of the protein allows close packing of secondary structural elements. Topologically, the surface has no indentations that indicate a nexus for molecular interactions. The distribution of polar and apolar groups on the surface of VapG is markedly uneven. One-third of the surface is dominated by exposed apolar side-chains, with no ionisable and only four polar side-chains exposed, giving rise to an expansive flat hydrophobic surface. Other surface regions are more polar, especially on or near the α-helix and a belt around the centre of the β-barrel. Possible functional significance of these recent structures is discussed.
Collapse
Affiliation(s)
- Tebekeme Okoko
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Elena V Blagova
- Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York YO10 5DD, UK
| | - Jean L Whittingham
- Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York YO10 5DD, UK.
| | - Lynn G Dover
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Anthony J Wilkinson
- Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York YO10 5DD, UK
| |
Collapse
|
22
|
Burton AJ, Giguère S, Berghaus LJ, Hondalus MK, Arnold RD. Efficacy of liposomal gentamicin against Rhodococcus equi in a mouse infection model and colocalization with R. equi in equine alveolar macrophages. Vet Microbiol 2015; 176:292-300. [PMID: 25666452 DOI: 10.1016/j.vetmic.2015.01.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/16/2015] [Accepted: 01/19/2015] [Indexed: 11/24/2022]
Abstract
Rhodococcus equi, a facultative intracellular pathogen and an important cause of pneumonia in foals, is highly susceptible to killing by gentamicin in vitro. However, gentamicin is not effective in vivo, due to its poor cellular penetration. Encapsulation of drugs in liposomes enhances cellular uptake. The objectives of this study were to compare liposomal gentamicin and free gentamicin with respect to their uptake by equine macrophages and intracellular colocalization with R. equi and to compare the efficacies of liposomal gentamicin, free gentamicin and clarithromycin with rifampin for the reduction of R. equi CFU in a mouse model of infection. After ex vivo exposure, a significantly higher mean (±SD) percentage of equine alveolar macrophages contained liposomal gentamicin (91.9±7.6%) as opposed to free gentamicin (16.8±12.5%). Intracellular colocalization of drug and R. equi, as assessed by confocal microscopy, occurred in a significantly higher proportion of cells exposed to liposomal gentamicin (81.2±17.8%) compared to those exposed to free gentamicin (10.4±8.7%). The number of R. equi CFU in the spleen was significantly lower in mice treated with liposomal gentamicin compared to that of mice treated with free gentamicin or to untreated control mice. Treatment with liposomal gentamicin also resulted in a significantly greater reduction in the number of R. equi CFU in the liver compared to treatment with clarithromycin in combination with rifampin. These results support further investigation of liposomal gentamicin as a new treatment for infections caused by R. equi.
Collapse
Affiliation(s)
- Alexandra J Burton
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Steeve Giguère
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.
| | - Londa J Berghaus
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Mary K Hondalus
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Robert D Arnold
- Department of Drug Discovery & Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| |
Collapse
|
23
|
Giles C, Vanniasinkam T, Ndi S, Barton MD. Rhodococcus equi (Prescottella equi)vaccines; the future of vaccine development. Equine Vet J 2014; 47:510-8. [DOI: 10.1111/evj.12310] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 06/12/2014] [Indexed: 12/29/2022]
Affiliation(s)
- C. Giles
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide Australia
| | - T. Vanniasinkam
- School of Biomedical Sciences; Charles Sturt University; Wagga Wagga New South Wales Australia
| | - S. Ndi
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide Australia
| | - M. D. Barton
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide Australia
| |
Collapse
|
24
|
Characterization of Rhodococcus equi isolates from submaxillary lymph nodes of wild boars (Sus scrofa), red deer (Cervus elaphus) and roe deer (Capreolus capreolus). Vet Microbiol 2014; 172:272-8. [PMID: 24878324 DOI: 10.1016/j.vetmic.2014.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/24/2014] [Accepted: 04/26/2014] [Indexed: 12/24/2022]
Abstract
Rhodococcus equi is a soil saprophyte and an opportunistic pathogen causing infections in animals, and rarely in humans. The presence of R. equi in tissues and faeces of some wild animal species was demonstrated previously. In this study we characterized R. equi isolates from submaxillary lymph nodes of free-living wild boars (n=23), red deer (n=2) and roe deer (n=2). This is the first description of R. equi strains isolated from tissues of the Cervidae. All isolates were initially recognized as R. equi based on the phenotypic properties. Their identification was confirmed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, detection of the choE gene and by sequence analysis of the 16S rRNA and rpoB genes. The presence of three plasmidic genes (traA, vapA and vapB) associated with R. equi virulence was investigated by PCR. In 16 wild boar isolates the traA and vapB genes were detected and they were located on virulence plasmids type 5, 7 or 11. The isolates from cervids and the remaining wild boar isolates were classified as avirulent based on a genotype traA(-)/vapA(-)B(-). In summary, these results confirm that wild boars can be a source of intermediately virulent R. equi strains, and indicate that red deer and roe deer can be a reservoir of avirulent R. equi strains.
Collapse
|
25
|
Berghaus LJ, Giguère S, Sturgill TL. Effects of age and macrophage lineage on intracellular survival and cytokine induction after infection with Rhodococcus equi. Vet Immunol Immunopathol 2014; 160:41-50. [PMID: 24736188 DOI: 10.1016/j.vetimm.2014.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 02/20/2014] [Accepted: 03/23/2014] [Indexed: 11/25/2022]
Abstract
Rhodococcus equi, a facultative intracellular pathogen of macrophages, causes life-threatening pneumonia in foals and in people with underlying immune deficiencies. As a basis for this study, we hypothesized that macrophage lineage and age would affect intracellular survival of R. equi and cytokine induction after infection. Monocyte-derived and bronchoalveolar macrophages from 10 adult horses and from 10 foals (sampled at 1-3 days, 2 weeks, 1 month, 3 months, and 5 months of age) were infected ex vivo with virulent R. equi. Intracellular R. equi were quantified and mRNA expression of IL-1β, IL-4, IL-6, IL-8, IL-10, IL-12 p40, IL-18, IFN-γ, and TNF-α was measured. Intracellular replication of R. equi was significantly (P<0.001) greater in bronchoalveolar than in monocyte-derived macrophages, regardless of age. Regardless of the macrophage lineage, replication of R. equi was significantly (P=0.002) higher in 3-month-old foals than in 3-day old foals, 2-week-old foals, 1-month-old foals, and adult horses. Expression of IL-4 mRNA was significantly higher in monocyte-derived macrophages whereas expression of IL-6, IL-18, and TNF-α was significantly higher in bronchoalveolar macrophages. Induction of IL-1β, IL-10, IL-12 p40, and IL-8 mRNA in bronchoalveolar macrophages of 1-3-day old foals was significantly higher than in older foals or adult horses. Preferential intracellular survival of R. equi in bronchoalveolar macrophages of juvenile horses may play a role in the pulmonary tropism of the pathogen and in the window of age susceptibility to infection.
Collapse
Affiliation(s)
- Londa J Berghaus
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States
| | - Steeve Giguère
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States.
| | - Tracy L Sturgill
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States
| |
Collapse
|
26
|
IcgA is a virulence factor of Rhodococcus equi that modulates intracellular growth. Infect Immun 2014; 82:1793-800. [PMID: 24549327 DOI: 10.1128/iai.01670-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Virulence of the intracellular pathogen Rhodococcus equi depends on a 21.3-kb pathogenicity island located on a conjugative plasmid. To date, the only nonregulatory pathogenicity island-encoded virulence factor identified is the cell envelope-associated VapA protein. Although the pathogenicity islands from porcine and equine R. equi isolates have undergone major rearrangements, the virR operon (virR-icgA-vapH-orf7-virS) is highly conserved in both, suggesting these genes play an important role in pathogenicity. VirR and VirS are transcriptional regulators controlling expression of pathogenicity island genes, including vapA. Here, we show that while vapH and orf7 are dispensable for intracellular growth of R. equi, deletion of icgA, formerly known as orf5, encoding a major facilitator superfamily transport protein, elicited an enhanced growth phenotype in macrophages and a significant reduction in macrophage viability, while extracellular growth in broth remained unaffected. Transcription of virS, located downstream of icgA, and vapA was not affected by the icgA deletion during growth in broth or in macrophages, showing that the enhanced growth phenotype caused by deletion of icgA was not mediated through abnormal transcription of these genes. Transcription of icgA increased 6-fold within 2 h following infection of macrophages and remained significantly higher 48 h postinfection compared to levels at the start of the infection. The major facilitator superfamily transport protein IcgA is the first factor identified in R. equi that negatively affects intracellular replication. Aside from VapA, it is only the second pathogenicity island-encoded structural protein shown to play a direct role in intracellular growth of this pathogenic actinomycete.
Collapse
|
27
|
Vázquez-Boland JA, Giguère S, Hapeshi A, MacArthur I, Anastasi E, Valero-Rello A. Rhodococcus equi: the many facets of a pathogenic actinomycete. Vet Microbiol 2013; 167:9-33. [PMID: 23993705 DOI: 10.1016/j.vetmic.2013.06.016] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 06/27/2013] [Indexed: 12/20/2022]
Abstract
Rhodococcus equi is a soil-dwelling pathogenic actinomycete that causes pulmonary and extrapulmonary pyogranulomatous infections in a variety of animal species and people. Young foals are particularly susceptible and develop a life-threatening pneumonic disease that is endemic at many horse-breeding farms worldwide. R. equi is a facultative intracellular parasite of macrophages that replicates within a modified phagocytic vacuole. Its pathogenicity depends on a virulence plasmid that promotes intracellular survival by preventing phagosome-lysosome fusion. Species-specific tropism of R. equi for horses, pigs and cattle appears to be determined by host-adapted virulence plasmid types. Molecular epidemiological studies of these plasmids suggest that human R. equi infection is zoonotic. Analysis of the recently determined R. equi genome sequence has identified additional virulence determinants on the bacterial chromosome. This review summarizes our current understanding of the clinical aspects, biology, pathogenesis and immunity of this fascinating microbe with plasmid-governed infectivity.
Collapse
Affiliation(s)
- José A Vázquez-Boland
- Microbial Pathogenesis Unit, School of Biomedical Sciences and Edinburgh Infectious Diseases, University of Edinburgh, Edinburgh EH9 3JT, UK; Grupo de Patogenómica Bacteriana, Facultad de Veterinaria, Universidad de León, 24071 León, Spain.
| | | | | | | | | | | |
Collapse
|
28
|
Conjugal transfer of a virulence plasmid in the opportunistic intracellular actinomycete Rhodococcus equi. J Bacteriol 2012; 194:6790-801. [PMID: 23042997 DOI: 10.1128/jb.01210-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rhodococcus equi is a facultative intracellular, Gram-positive, soilborne actinomycete which can cause severe pyogranulomatous pneumonia with abscessation in young horses (foals) and in immunocompromised people, such as persons with AIDS. All strains of R. equi isolated from foals and approximately a third isolated from humans contain a large, ~81-kb plasmid which is essential for the intramacrophage growth of the organism and for virulence in foals and murine in vivo model systems. We found that the entire virulence plasmid could be transferred from plasmid-containing strains of R. equi (donor) to plasmid-free R. equi strains (recipient) at a high frequency and that plasmid transmission reestablished the capacity for intracellular growth in macrophages. Plasmid transfer required living cells and cell-to-cell contact and was unaffected by the presence of DNase, factors pointing to conjugation as the major means of genetic transfer. Deletion of a putative relaxase-encoding gene, traA, located in the proposed conjugative region of the plasmid, abolished plasmid transfer. Reversion of the traA mutation restored plasmid transmissibility. Finally, plasmid transmission to other Rhodococcus species and some additional related organisms was demonstrated. This is the first study showing a virulence plasmid transfer in R. equi, and it establishes a mechanism by which the virulence plasmid can move among bacteria in the soil.
Collapse
|
29
|
Giguère S, Cohen N, Keith Chaffin M, Hines S, Hondalus M, Prescott J, Slovis N. Rhodococcus equi: Clinical Manifestations, Virulence, and Immunity. J Vet Intern Med 2011; 25:1221-30. [DOI: 10.1111/j.1939-1676.2011.00804.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/01/2011] [Accepted: 08/15/2011] [Indexed: 11/27/2022] Open
Affiliation(s)
- S. Giguère
- Department of Large Animal Medicine; University of Georgia; Athens; GA
| | - N.D. Cohen
- Department of Large Animal Clinical Sciences; College of Veterinary Medicine; Texas A&M University; College Station; TX
| | - M. Keith Chaffin
- Department of Large Animal Clinical Sciences; College of Veterinary Medicine; Texas A&M University; College Station; TX
| | - S.A. Hines
- Department of Veterinary Microbiology and Pathology; Washington State University; Pullman; WA
| | - M.K. Hondalus
- Department of Infectious Diseases; University of Georgia; Athens; GA
| | - J.F. Prescott
- Department of Pathobiology; University of Guelph; Guelph; ON; Canada
| | - N.M. Slovis
- Hagyard Equine Medical Institute; Lexington; KY
| |
Collapse
|
30
|
The steroid catabolic pathway of the intracellular pathogen Rhodococcus equi is important for pathogenesis and a target for vaccine development. PLoS Pathog 2011; 7:e1002181. [PMID: 21901092 PMCID: PMC3161971 DOI: 10.1371/journal.ppat.1002181] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 06/12/2011] [Indexed: 01/06/2023] Open
Abstract
Rhodococcus equi causes fatal pyogranulomatous pneumonia in foals and immunocompromised animals and humans. Despite its importance, there is currently no effective vaccine against the disease. The actinobacteria R. equi and the human pathogen Mycobacterium tuberculosis are related, and both cause pulmonary diseases. Recently, we have shown that essential steps in the cholesterol catabolic pathway are involved in the pathogenicity of M. tuberculosis. Bioinformatic analysis revealed the presence of a similar cholesterol catabolic gene cluster in R. equi. Orthologs of predicted M. tuberculosis virulence genes located within this cluster, i.e. ipdA (rv3551), ipdB (rv3552), fadA6 and fadE30, were identified in R. equi RE1 and inactivated. The ipdA and ipdB genes of R. equi RE1 appear to constitute the α-subunit and β-subunit, respectively, of a heterodimeric coenzyme A transferase. Mutant strains RE1ΔipdAB and RE1ΔfadE30, but not RE1ΔfadA6, were impaired in growth on the steroid catabolic pathway intermediates 4-androstene-3,17-dione (AD) and 3aα-H-4α(3′-propionic acid)-5α-hydroxy-7aβ-methylhexahydro-1-indanone (5α-hydroxy-methylhexahydro-1-indanone propionate; 5OH-HIP). Interestingly, RE1ΔipdAB and RE1ΔfadE30, but not RE1ΔfadA6, also displayed an attenuated phenotype in a macrophage infection assay. Gene products important for growth on 5OH-HIP, as part of the steroid catabolic pathway, thus appear to act as factors involved in the pathogenicity of R. equi. Challenge experiments showed that RE1ΔipdAB could be safely administered intratracheally to 2 to 5 week-old foals and oral immunization of foals even elicited a substantial protective immunity against a virulent R. equi strain. Our data show that genes involved in steroid catabolism are promising targets for the development of a live-attenuated vaccine against R. equi infections. Rhodococcus equi causes fatal pyogranulomatous bronchopneumonia in young foals and is an emerging opportunistic pathogen of immunocompromised humans. Despite its importance, there is currently no safe and effective vaccine against R. equi infections. Like Mycobacterium tuberculosis, the causative agent of human tuberculosis, R. equi is able to infect, survive and multiply inside alveolar macrophages. Recently we have shown that essential steps in the cholesterol catabolic pathway (encoded by the rv3551, rv3552, fadE30 genes) are involved in the pathogenicity of M. tuberculosis. We hypothesized that the orthologous genes in the cholesterol catabolic gene cluster of R. equi also are essential for its virulence mechanism. Analysis of the respective R. equi strain RE1 mutants revealed that they were impaired in growth on intermediates of the steroid catabolic pathway and had attenuated phenotypes in a macrophage infection assay. Mutant RE1ΔipdAB, carrying a deletion of the orthologs of rv3551 and rv3552, could be safely administered to 2–5 week-old foals intratracheally and oral immunization provided a substantial protection against infection by a virulent R. equi strain. Our data show that genes important for methylhexahydroindanone propionate degradation, part of the steroid catabolic pathway, are promising targets for the development of a live-attenuated vaccine against R. equi infections.
Collapse
|
31
|
BERGHAUS LJ, GIGUÈRE S, STURGILL TL, BADE D, MALINSKI TJ, HUANG R. Plasma pharmacokinetics, pulmonary distribution, and in vitro activity of gamithromycin in foals. J Vet Pharmacol Ther 2011; 35:59-66. [DOI: 10.1111/j.1365-2885.2011.01292.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|