1
|
Cottam C, White RT, Beck LC, Stewart CJ, Beatson SA, Lowe EC, Grinter R, Connolly JPR. Metabolism of L-arabinose converges with virulence regulation to promote enteric pathogen fitness. Nat Commun 2024; 15:4462. [PMID: 38796512 PMCID: PMC11127945 DOI: 10.1038/s41467-024-48933-7] [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: 12/18/2023] [Accepted: 05/16/2024] [Indexed: 05/28/2024] Open
Abstract
Virulence and metabolism are often interlinked to control the expression of essential colonisation factors in response to host-associated signals. Here, we identified an uncharacterised transporter of the dietary monosaccharide ʟ-arabinose that is widely encoded by the zoonotic pathogen enterohaemorrhagic Escherichia coli (EHEC), required for full competitive fitness in the mouse gut and highly expressed during human infection. Discovery of this transporter suggested that EHEC strains have an enhanced ability to scavenge ʟ-arabinose and therefore prompted us to investigate the impact of this nutrient on pathogenesis. Accordingly, we discovered that ʟ-arabinose enhances expression of the EHEC type 3 secretion system, increasing its ability to colonise host cells, and that the underlying mechanism is dependent on products of its catabolism rather than the sensing of ʟ-arabinose as a signal. Furthermore, using the murine pathogen Citrobacter rodentium, we show that ʟ-arabinose metabolism provides a fitness benefit during infection via virulence factor regulation, as opposed to supporting pathogen growth. Finally, we show that this mechanism is not restricted to ʟ-arabinose and extends to other pentose sugars with a similar metabolic fate. This work highlights the importance integrating central metabolism with virulence regulation in order to maximise competitive fitness of enteric pathogens within the host-niche.
Collapse
Affiliation(s)
- Curtis Cottam
- Newcastle University Biosciences Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Rhys T White
- Institute of Environmental Science and Research, Wellington, New Zealand
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Lauren C Beck
- Newcastle University Translation and Clinical Research Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Christopher J Stewart
- Newcastle University Translation and Clinical Research Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Scott A Beatson
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Elisabeth C Lowe
- Newcastle University Biosciences Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Rhys Grinter
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - James P R Connolly
- Newcastle University Biosciences Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK.
| |
Collapse
|
2
|
Phoka T, Techawiwattanaboon T, Sangjun N, Komanee P, Murray GL, Wongratanacheewin Sermswan R, Adler B, Patarakul K. Identification of in vivo expressed proteins in live attenuated lipopolysaccharide mutant that mediates heterologous protection against Leptospira spp. Vet Microbiol 2021; 262:109220. [PMID: 34509026 DOI: 10.1016/j.vetmic.2021.109220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/28/2021] [Indexed: 10/20/2022]
Abstract
Leptospirosis vaccines that elicit broad protection against a range of pathogenic Leptospira spp. would overcome a major drawback of currently licensed bacterin vaccines. Live attenuated vaccine produced from a lipopolysaccharide (LPS) mutant strain of L. interrogans serovar Manilae M1352 (Live M1352) stimulated better protective efficacy than heat killed M1352 (HK M1352) against a heterologous challenge with L. interrogans serovar Pomona. To identify antigens of Live M1352 potentially responsible for cross protection, in vivo-induced antigen technology (IVIAT), a powerful tool to identify in vivo-induced (ivi) genes expressed during infection, was employed in this study. Pooled sera from hamsters immunized with Live M1352 were sequentially adsorbed with various preparations of in vitro grown M1352. The pre-adsorbed sera were used to screen a genomic expression library of M1352. Nineteen strongly reactive clones were selected for DNA sequencing. These ivi genes are conserved in most Leptospira strains. Four selected genes including LIMLP_04965 (tolB), LIMLP_01535, LIMLP_06785 (fliI), and LIMLP_14930 were confirmed for their upregulated expression in kidneys of infected hamsters by RT-qPCR, suggesting their role in leptospiral infection. These ivi proteins represent potential targets for vaccine candidates that warrant further investigation for their protective efficacy.
Collapse
Affiliation(s)
- Theerapat Phoka
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Teerasit Techawiwattanaboon
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand; Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Noppadon Sangjun
- Armed Force Research Institute of Medical Sciences (AFRIMS), Bangkok, 10400, Thailand
| | - Pat Komanee
- Armed Force Research Institute of Medical Sciences (AFRIMS), Bangkok, 10400, Thailand
| | - Gerald L Murray
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia; The Royal Women's Hospital, Parkville, VIC, 3052, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Rasana Wongratanacheewin Sermswan
- Melioidosis Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand; Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Ben Adler
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Kanitha Patarakul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand; Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
3
|
Katani R, Kudva IT, Srinivasan S, Stasko JB, Schilling M, Li L, Cote R, DebRoy C, Arthur TM, Sokurenko EV, Kapur V. Strain and host-cell dependent role of type-1 fimbriae in the adherence phenotype of super-shed Escherichia coli O157:H7. Int J Med Microbiol 2021; 311:151511. [PMID: 33975122 PMCID: PMC8605689 DOI: 10.1016/j.ijmm.2021.151511] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/14/2021] [Accepted: 05/03/2021] [Indexed: 02/04/2023] Open
Abstract
Super-shed (SS) Escherichia coli O157 (E. coli O157) demonstrate a strong, aggregative, locus of enterocyte effacement (LEE)-independent adherence phenotype on bovine recto-anal junction squamous epithelial (RSE) cells, and harbor polymorphisms in non-LEE-adherence-related loci, including in the type 1 fimbriae operon. To elucidate the role of type 1 fimbriae in strain- and host-specific adherence, we evaluated the entire Fim operon (FimB-H) and its adhesion (FimH) deletion mutants in four E. coli O157 strains, SS17, SS52, SS77 and EDL933, and evaluated the adherence phenotype in bovine RSE and human HEp-2 adherence assays. Consistent with the prevailing dogma that fimH expression is genetically switched off in E. coli O157, the ΔfimHSS52, ΔfimB-HSS52, ΔfimB-HSS17, and ΔfimHSS77 mutants remained unchanged in adherence phenotype to RSE cells. In contrast, the ΔfimHSS17 and ΔfimB-HSS77 mutants changed from a wild-type strong and aggregative, to a moderate and diffuse adherence phenotype, while both ΔfimHEDL933 and ΔfimB-HEDL933 mutants demonstrated enhanced binding to RSE cells (p < 0.05). Additionally, both ΔfimHSS17 and ΔfimHEDL933 were non-adherent to HEp-2 cells (p < 0.05). Complementation of the mutant strains with their respective wild-type genes restored parental phenotypes. Microscopy revealed that the SS17 and EDL933 strains indeed carry type 1 fimbriae-like structures shorter than those seen in uropathogenic E. coli. Taken together, these results provide compelling evidence for a strain and host cell type-dependent role of fimH and the fim operon in E. coli O157 adherence that needs to be further evaluated.
Collapse
Affiliation(s)
- Robab Katani
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Indira T Kudva
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, USA.
| | - Sreenidhi Srinivasan
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Judith B Stasko
- Microscopy Services, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, USA
| | - Megan Schilling
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Lingling Li
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Rebecca Cote
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Chitrita DebRoy
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Terrance M Arthur
- Roman L. Hruska U.S. Meat Animal Research Center, Agricultural Research Service, U.S. Department of Agriculture, Clay Center, NE, USA
| | | | - Vivek Kapur
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA; Department of Animal Science, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
4
|
Naïli I, Gardette M, Garrivier A, Daniel J, Desvaux M, Pizza M, Gobert A, Marchal T, Loukiadis E, Jubelin G. Interplay between enterohaemorrhagic Escherichia coli and nitric oxide during the infectious process. Emerg Microbes Infect 2021; 9:1065-1076. [PMID: 32459575 PMCID: PMC7336997 DOI: 10.1080/22221751.2020.1768804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) are bacterial pathogens responsible for life-threatening diseases in humans such as bloody diarrhoea and the hemolytic and uremic syndrome. To date, no specific therapy is available and treatments remain essentially symptomatic. In recent years, we demonstrated in vitro that nitric oxide (NO), a major mediator of the intestinal immune response, strongly represses the synthesis of the two cardinal virulence factors in EHEC, namely Shiga toxins (Stx) and the type III secretion system, suggesting NO has a great potential to protect against EHEC infection. In this study, we investigated the interplay between NO and EHEC in vivo using mouse models of infection. Using a NO-sensing reporter strain, we determined that EHEC sense NO in the gut of infected mice. Treatment of infected mice with a specific NOS inhibitor increased EHEC adhesion to the colonic mucosa but unexpectedly decreased Stx activity in the gastrointestinal tract, protecting mice from renal failure. Taken together, our data indicate that NO can have both beneficial and detrimental consequences on the outcome of an EHEC infection, and underline the importance of in vivo studies to increase our knowledge in host–pathogen interactions.
Collapse
Affiliation(s)
- Ilham Naïli
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France.,GSK, Siena, Italy
| | - Marion Gardette
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France.,Université de Lyon, CNRS, INRAE, Université Claude Bernard Lyon 1, VetAgro Sup, Laboratoire d'Ecologie Microbienne, F-63280 Marcy l'Etoile, France
| | - Annie Garrivier
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| | - Julien Daniel
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| | - Mickaël Desvaux
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| | | | - Alain Gobert
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| | - Thierry Marchal
- VetAgro Sup, Laboratoire vétérinaire d'histopathologie, F-63280 Marcy-l'Etoile, France
| | - Estelle Loukiadis
- Université de Lyon, CNRS, INRAE, Université Claude Bernard Lyon 1, VetAgro Sup, Laboratoire d'Ecologie Microbienne, F-63280 Marcy l'Etoile, France.,VetAgro Sup, Laboratoire national de référence des E. coli, F-63280 Marcy-l'Etoile, France
| | - Grégory Jubelin
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| |
Collapse
|
5
|
Role of the Nitric Oxide Reductase NorVW in the Survival and Virulence of Enterohaemorrhagic Escherichia coli during Infection. Pathogens 2020; 9:pathogens9090683. [PMID: 32825770 PMCID: PMC7558590 DOI: 10.3390/pathogens9090683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 01/20/2023] Open
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) are bacterial pathogens responsible for life-threatening diseases in humans, such as hemolytic and uremic syndrome. It has been previously demonstrated that the interplay between EHEC and nitric oxide (NO), a mediator of the host immune innate response, is critical for infection outcome, since NO affects both Shiga toxin (Stx) production and adhesion to enterocytes. In this study, we investigated the role of the NO reductase NorVW in the virulence and fitness of two EHEC strains in a murine model of infection. We determined that the deletion of norVW in the strain O91:H21 B2F1 has no impact on its virulence, whereas it reduces the ability of the strain O157:H7 620 to persist in the mouse gut and to produce Stx. We also revealed that the fitness defect of strain 620 ΔnorVW is strongly attenuated when mice are treated with an NO synthase inhibitor. Altogether, these results demonstrate that the NO reductase NorVW participates in EHEC resistance against NO produced by the host and promotes virulence through the modulation of Stx synthesis. The contribution of NorVW in the EHEC infectious process is, however, strain-dependent and suggests that the EHEC response to nitrosative stress is complex and multifactorial.
Collapse
|
6
|
Identification of Haemophilus parasuis genes uniquely expressed during infection using in vivo-induced antigen technology. Vet Microbiol 2020; 243:108650. [PMID: 32273024 DOI: 10.1016/j.vetmic.2020.108650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/04/2020] [Accepted: 03/17/2020] [Indexed: 01/12/2023]
Abstract
Haemophilus parasuis is the etiological agent of Glässer's disease which is characterized by fibrinous polyserositis, arthritis and meningitis. The pathogenesis of this bacterium remains largely unknown. Genes expressed in vivo may play an important role in the pathogenicity of H. parasuis. The development of in vivo-induced antigen technology (IVIAT) has provided a valuable tool for the identification of in vivo-induced genes during bacterial infection. In this study, IVIAT was applied to identify in vivo-induced antigens of H. parasuis. Pooled swine H. parasuis-positive sera, adsorbed against in vitro-grown cultures of H. parasuis SH0165 and Escherichia coli BL21 (DE3), were used to screen the inducible expression library of genomic proteins from whole genome sequenced H. parsuis SH0165. Finally, 24 unique genes expressed in vivo were successfully identified after secondary and tertiary screening with IVIAT. These genes were implicated in cell surface proteins, metabolism, stress response, regulation, transportation and other processes. Quantitative real-time PCR showed that the mRNA levels of 24 genes were all upregulated in vivo relative to in vitro, with 13 genes were detected significantly upregulated in H. parasuis infected pigs. Several potential virulence-associated genes were found to be uniquely expressed in vivo, including espP, lnt, hutZ, mreC, vtaA, pilB, tex, sunT and aidA. The results indicated that the proteins identified using IVIAT may play important roles in the pathogenesis of H. parasuis infection in vivo.
Collapse
|
7
|
Montero DA, Del Canto F, Salazar JC, Céspedes S, Cádiz L, Arenas-Salinas M, Reyes J, Oñate Á, Vidal RM. Immunization of mice with chimeric antigens displaying selected epitopes confers protection against intestinal colonization and renal damage caused by Shiga toxin-producing Escherichia coli. NPJ Vaccines 2020; 5:20. [PMID: 32194997 PMCID: PMC7067774 DOI: 10.1038/s41541-020-0168-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) cause diarrhea and dysentery, which may progress to hemolytic uremic syndrome (HUS). Vaccination has been proposed as a preventive approach against STEC infection; however, there is no vaccine for humans and those used in animals reduce but do not eliminate the intestinal colonization of STEC. The OmpT, Cah and Hes proteins are widely distributed among clinical STEC strains and are recognized by serum IgG and IgA in patients with HUS. Here, we develop a vaccine formulation based on two chimeric antigens containing epitopes of OmpT, Cah and Hes proteins against STEC strains. Intramuscular and intranasal immunization of mice with these chimeric antigens elicited systemic and local long-lasting humoral responses. However, the class of antibodies generated was dependent on the adjuvant and the route of administration. Moreover, while intramuscular immunization with the combination of the chimeric antigens conferred protection against colonization by STEC O157:H7, the intranasal conferred protection against renal damage caused by STEC O91:H21. This preclinical study supports the potential use of this formulation based on recombinant chimeric proteins as a preventive strategy against STEC infections.
Collapse
Affiliation(s)
- David A Montero
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,2Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe Del Canto
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan C Salazar
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sandra Céspedes
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Leandro Cádiz
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mauricio Arenas-Salinas
- 3Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, Talca, Chile
| | - José Reyes
- 4Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ángel Oñate
- 4Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roberto M Vidal
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,5Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
8
|
Gardette M, Le Hello S, Mariani-Kurkdjian P, Fabre L, Gravey F, Garrivier A, Loukiadis E, Jubelin G. Identification and prevalence of in vivo-induced genes in enterohaemorrhagic Escherichia coli. Virulence 2019; 10:180-193. [PMID: 30806162 PMCID: PMC6550539 DOI: 10.1080/21505594.2019.1582976] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/25/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) are food-borne pathogens responsible for bloody diarrhoea and renal failure in humans. While Shiga toxin (Stx) is the cardinal virulence factor of EHEC, its production by E. coli is not sufficient to cause disease and many Shiga-toxin producing E. coli (STEC) strains have never been implicated in human infection. So far, the pathophysiology of EHEC infection is not fully understood and more knowledge is needed to characterize the "auxiliary" factors that enable a STEC strain to cause disease in humans. In this study, we applied a recombinase-based in vivo expression technology (RIVET) to the EHEC reference strain EDL933 in order to identify genes specifically induced during the infectious process, using mouse as an infection model. We identified 31 in vivo-induced (ivi) genes having functions related to metabolism, stress adaptive response and bacterial virulence or fitness. Eight of the 31 ivi genes were found to be heterogeneously distributed in EHEC strains circulating in France these last years. In addition, they are more prevalent in strains from the TOP seven priority serotypes and particularly strains carrying significant virulence determinants such as Stx2 and intimin adhesin. This work sheds further light on bacterial determinants over-expressed in vivo during infection that may contribute to the potential of STEC strains to cause disease in humans.
Collapse
Affiliation(s)
- Marion Gardette
- UCA, INRA, UMR454 MEDIS, Clermont-Ferrand, France
- Laboratoire d’écologie microbienne de Lyon, Université de Lyon, CNRS, INRA, UCBL, VetAgro Sup, Marcy l’Etoile, France
| | - Simon Le Hello
- Centre de Référence National des Escherichia coli, Shigella et Salmonella, Institut Pasteur, Paris, France
- Université de Normandie, EA 2656 GRAM 2.0, UNICAEN, Caen, France
| | - Patricia Mariani-Kurkdjian
- Service de Microbiologie, Centre National de Référence associé Escherichia coli, Hôpital Robert-Debré, AP-HP, Paris, France
| | - Laetitia Fabre
- Centre de Référence National des Escherichia coli, Shigella et Salmonella, Institut Pasteur, Paris, France
| | - François Gravey
- Centre de Référence National des Escherichia coli, Shigella et Salmonella, Institut Pasteur, Paris, France
- Université de Normandie, EA 2656 GRAM 2.0, UNICAEN, Caen, France
| | | | - Estelle Loukiadis
- Laboratoire d’écologie microbienne de Lyon, Université de Lyon, CNRS, INRA, UCBL, VetAgro Sup, Marcy l’Etoile, France
- Laboratoire national de référence des E. coli, Université de Lyon, VetAgro Sup, Marcy l’Etoile, France
| | | |
Collapse
|
9
|
Galactose-1-phosphate uridyltransferase (GalT), an in vivo-induced antigen of Actinobacillus pleuropneumoniae serovar 5b strain L20, provided immunoprotection against serovar 1 strain MS71. PLoS One 2018; 13:e0198207. [PMID: 29856812 PMCID: PMC5983418 DOI: 10.1371/journal.pone.0198207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 05/15/2018] [Indexed: 11/19/2022] Open
Abstract
GALT is an important antigen of Actinobacillus pleuropneumoniae (APP), which was shown to provide partial protection against APP infection in a previous study in our lab. The main purpose of the present study is to investigate GALT induced cross-protection between different APP serotypes and elucidate key mechanisms of the immune response to GALT antigenic stimulation. Bioinformatic analysis demonstrated that galT is a highly conserved gene in APP, widely distributed across multiple pathogenic strains. Homologies between any two strains ranges from 78.9% to 100% regarding the galT locus. Indirect enzyme-linked immunosorbent assay (ELISA) confirmed that GALT specific antibodies could not be induced by inactivated APP L20 or MS71 whole cell bacterin preparations. A recombinant fusion GALT protein derived from APP L20, however has proven to be an effective cross-protective antigen against APP sevorar 1 MS71 (50%, 4/8) and APP sevorar 5b L20 (75%, 6/8). Histopathological examinations have confirmed that recombinant GALT vaccinated animals showed less severe pathological signs in lung tissues than negative controls after APP challenge. Immunohistochemical (IHC) analysis indicated that the infiltration of neutrophils in the negative group is significantly increased compared with that in the normal control (P<0.001) and that in surviving animals is decreased compared to the negative group. Anti-GALT antibodies were shown to mediate phagocytosis of neutrophils. After interaction with anti-GALT antibodies, survival rate of APP challenged vaccinated animals was significantly reduced (P<0.001). This study demonstrated that GALT is an effective cross-protective antigen, which could be used as a potential vaccine candidate against multiple APP serotypes.
Collapse
|
10
|
Zhang F, Cao S, Zhu Z, Yang Y, Wen X, Chang YF, Huang X, Wu R, Wen Y, Yan Q, Huang Y, Ma X, Zhao Q. Immunoprotective Efficacy of Six In vivo-Induced Antigens against Actinobacillus pleuropneumoniae as Potential Vaccine Candidates in Murine Model. Front Microbiol 2016; 7:1623. [PMID: 27818646 PMCID: PMC5073529 DOI: 10.3389/fmicb.2016.01623] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/29/2016] [Indexed: 11/21/2022] Open
Abstract
Six in vivo-induced (IVI) antigens—RnhB, GalU, GalT, Apl_1061, Apl_1166, and HflX were selected for a vaccine trial in a mouse model. The results showed that the IgG levels in each immune group was significantly higher than that of the negative control (P < 0.001). Except rRnhB group, proliferation of splenocytes was observed in all immunized groups and a relatively higher proliferation activity was observed in rGalU and rGalT groups (P < 0.05). In the rGalT vaccinated group, the proportion of CD4+ T cells in spleen was significant higher than that of negative control (P < 0.05). Moreover, proportions of CD4+ T cells in other vaccinated groups were all up-regulated to varying degrees. Up-regulation of both Th1 (IFN-γ, IL-2) and Th2 (IL-4) cytokines were detected. A survival rate of 87.5, 62.5, and 62.5% were obtained among rGalT, rAPL_1166, and rHflX group, respectively while the remaining three groups was only 25%. Histopathological analyses of lungs indicated that surviving animals from the vaccinated groups showed relatively normal pulmonary structure alveoli. These findings confirm that IVI antigens used as vaccine candidates provide partial protection against Actinobacillus pleuropneumoniae infection in a mouse model, which could be used as potential vaccine candidates in piglets.
Collapse
Affiliation(s)
- Fei Zhang
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Sanjie Cao
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural UniversityChengdu, China; Sichuan Science-observation Experiment of Veterinary Drugs and Veterinary Biological Technology, Ministry of AgricultureChengdu, China
| | - Zhuang Zhu
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Yusheng Yang
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Xintian Wen
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca NY, USA
| | - Xiaobo Huang
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Rui Wu
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Yiping Wen
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Qigui Yan
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural UniversityChengdu, China; Sichuan Science-observation Experiment of Veterinary Drugs and Veterinary Biological Technology, Ministry of AgricultureChengdu, China
| | - Yong Huang
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Xiaoping Ma
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Qin Zhao
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| |
Collapse
|
11
|
Krueger J, Pohl S, Preusse M, Kordes A, Rugen N, Schniederjans M, Pich A, Häussler S. Unravelling post-transcriptional PrmC-dependent regulatory mechanisms in Pseudomonas aeruginosa. Environ Microbiol 2016; 18:3583-3592. [PMID: 27376486 DOI: 10.1111/1462-2920.13435] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/28/2016] [Indexed: 02/04/2023]
Abstract
Transcriptional regulation has a central role in cellular adaptation processes and is well investigated. In contrast, the importance of the post-transcriptional regulation on these processes is less well defined. The technological advancements have been critical to precisely quantify protein and mRNA level changes and hold promise to provide more insights into how post-transcriptional regulation determines phenotypes. In Pseudomonas aeruginosa the methyltransferase PrmC methylates peptide chain release factors to facilitate translation termination. Loss of PrmC activity abolishes anaerobic growth and leads to reduced production of quorum sensing-associated virulence factors. Here, by applying SILAC technology in combination with mRNA-sequencing, they provide evidence that the P. aeruginosa phenotype can be attributed to a change in protein to mRNA ratios of selected protein groups. The UAG-dependent translation termination was more dependent on PrmC activity than the UAA- and UGA-dependent translation termination. Additionally, a bias toward UAG stop codons in global transcriptional regulators was found. The finding that this bias in stop codon usage determines the P. aeruginosa phenotype is unexpected and adds complexity to regulatory circuits. Via modulation of PrmC activity the bacterial cell can cross-regulate targets independently of transcriptional signals, a process with an underestimated impact on the bacterial phenotype.
Collapse
Affiliation(s)
- Jonas Krueger
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Research Core Unit Proteomics and Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Sarah Pohl
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany
| | - Matthias Preusse
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany
| | - Adrian Kordes
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Nils Rugen
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Monika Schniederjans
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany
| | - Andreas Pich
- Research Core Unit Proteomics and Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Susanne Häussler
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany. .,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany.
| |
Collapse
|
12
|
O'Ryan M, Vidal R, del Canto F, Carlos Salazar J, Montero D. Vaccines for viral and bacterial pathogens causing acute gastroenteritis: Part II: Vaccines for Shigella, Salmonella, enterotoxigenic E. coli (ETEC) enterohemorragic E. coli (EHEC) and Campylobacter jejuni. Hum Vaccin Immunother 2015; 11:601-19. [PMID: 25715096 DOI: 10.1080/21645515.2015.1011578] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In Part II we discuss the following bacterial pathogens: Shigella, Salmonella (non-typhoidal), diarrheogenic E. coli (enterotoxigenic and enterohemorragic) and Campylobacter jejuni. In contrast to the enteric viruses and Vibrio cholerae discussed in Part I of this series, for the bacterial pathogens described here there is only one licensed vaccine, developed primarily for Vibrio cholerae and which provides moderate protection against enterotoxigenic E. coli (ETEC) (Dukoral(®)), as well as a few additional candidates in advanced stages of development for ETEC and one candidate for Shigella spp. Numerous vaccine candidates in earlier stages of development are discussed.
Collapse
Key Words
- CFU, colony-forming units
- CFs, colonization factors
- CT, cholera toxin
- CT-B cholera toxin B subunit
- Campylobacter
- CtdB, cytolethal distending toxin subunit B
- E. coli
- EHEC
- EPEC, enteropathogenic E. coli
- ETEC
- ETEC, enterotoxigenic E. coli
- GEMS, Global enterics multicenter study
- HUS, hemolytic uremic syndrome
- IM, intramuscular
- IgA, immunoglobulin A
- IgG, immunoglobulin G
- IgM, immunoglobulin M
- LEE, locus of enterocyte effacement
- LPS, lipopolysaccharide
- LT, heat labile toxin
- LT-B
- OMV, outer membrane vesicles
- ST, heat stable toxin
- STEC
- STEC, shigatoxin producing E. coli
- STh, human heat stable toxin
- STp, porcine heat stable toxin
- Salmonella
- Shigella
- Stx, shigatoxin
- TTSS, type III secretion system
- V. cholera
- WHO, World Health Organization
- acute diarrhea
- dmLT, double mutant heat labile toxin
- enteric pathogens
- enterohemorrhagic E. coli
- gastroenteritis
- heat labile toxin B subunit
- norovirus
- rEPA, recombinant exoprotein A of Pseudomonas aeruginosa
- rotavirus
- vaccines
Collapse
Affiliation(s)
- Miguel O'Ryan
- a Microbiology and Mycology Program; Institute of Biomedical Sciences; Faculty of Medicine; Universidad de Chile; Santiago, Chile
| | | | | | | | | |
Collapse
|
13
|
Lee SW, Shet UK, Park SW, Lim HP, Yun KD, Kang SS, Kim SE. Identification of Enterococcus faecalis antigens specifically expressed in vivo. Restor Dent Endod 2015; 40:306-11. [PMID: 26587417 PMCID: PMC4650527 DOI: 10.5395/rde.2015.40.4.306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/10/2015] [Indexed: 01/05/2023] Open
Abstract
Objectives Molecular mechanism of the pathogenicity of Enterococcus faecalis (E. faecalis), a suspected endodontic pathogen, has not yet been adequately elucidated due to limited information on its virulence factors. Here we report the identification of in vivo expressed antigens of E. faecalis by using a novel immunoscreening technique called change-mediated antigen technology (CMAT) and an experimental animal model of endodontic infection. Materials and Methods Among 4,500 E. coli recombinant clones screened, 19 positive clones reacted reproducibly with hyperimmune sera obtained from rabbits immunized with E. faecalis cells isolated from an experimental endodontic infection. DNA sequences from 16 of these in vivo-induced (IVI) genes were determined. Results Identified protein antigens of E. faecalis included enzymes involved in housekeeping functions, copper resistance protein, putative outer membrane proteins, and proteins of unknown function. Conclusions In vivo expressed antigens of E. faecalis could be identified by using a novel immune-screening technique CMAT and an experimental animal model of endodontic infection. Detailed analysis of these IVI genes will lead to a better understanding of the molecular mechanisms involved in the endodontic infection of E. faecalis.
Collapse
Affiliation(s)
- Seok-Woo Lee
- Department of Dental Education, Dental Science Research Institute and BK21 Project, School of Dentistry, Gwangju, Korea. ; Department of Periodontology, Dental Science Research Institute and BK21 Project, School of Dentistry, Gwangju, Korea
| | - Uttom K Shet
- Department of Maxillofacial Surgery, Dental Science Research Institute and BK21 Project, School of Dentistry, Gwangju, Korea
| | - Sang-Won Park
- Department of Prosthodontics, Dental Science Research Institute and BK21 Project, School of Dentistry, Gwangju, Korea
| | - Hyun-Pil Lim
- Department of Prosthodontics, Dental Science Research Institute and BK21 Project, School of Dentistry, Gwangju, Korea
| | - Kwi-Dug Yun
- Department of Prosthodontics, Dental Science Research Institute and BK21 Project, School of Dentistry, Gwangju, Korea
| | - Seong Soo Kang
- Department of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Se Eun Kim
- Department of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| |
Collapse
|
14
|
Baker CA, Rubinelli PM, Park SH, Ricke SC. Immuno-based detection of Shiga toxin-producing pathogenic Escherichia coli in food – A review on current approaches and potential strategies for optimization. Crit Rev Microbiol 2015; 42:656-75. [DOI: 10.3109/1040841x.2015.1009824] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Christopher A. Baker
- Department of Food Science, Center for Food Safety, University of Arkansas, Fayetteville, AR, USA
| | - Peter M. Rubinelli
- Department of Food Science, Center for Food Safety, University of Arkansas, Fayetteville, AR, USA
| | - Si Hong Park
- Department of Food Science, Center for Food Safety, University of Arkansas, Fayetteville, AR, USA
| | - Steven C. Ricke
- Department of Food Science, Center for Food Safety, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
15
|
Identification of in vivo-induced bacterial protein antigens during calf infection with Chlamydia psittaci. Int J Med Microbiol 2015; 305:310-21. [DOI: 10.1016/j.ijmm.2014.12.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 11/19/2014] [Accepted: 12/20/2014] [Indexed: 01/21/2023] Open
|
16
|
Geng S, Liu Z, Lin Z, Barrow P, Pan Z, Li Q, Jiao X. Identification of in vivo-induced genes during infection of chickens with Salmonella enterica serovar Enteritidis. Res Vet Sci 2015; 100:1-7. [PMID: 25843894 DOI: 10.1016/j.rvsc.2015.03.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 02/28/2015] [Accepted: 03/22/2015] [Indexed: 10/23/2022]
Abstract
Chickens are an important source of food worldwide and are often infected with food-poisoning serovars of Salmonella enterica, frequently Salmonella Enteritidis (SE), without exhibiting clinical signs of disease. Ivi (in vivo induced) genes identified using in vivo-induced antigen technology (IVIAT) are expressed only during bacterial infection and have the potential value of identifying epidemic strains and antigens which can form the basis for sub-unit vaccine development. We applied IVIAT to SE strain 50041 and identified 42 ivi genes. Eight representative ivi genes were further confirmed by qRT-PCR as being expressed only in vivo within 48 h of infection compared with that of in vitro-cultured. Although our results indicated that the identified ivi genes are expressed only in vivo, further research is needed to elucidate the exact roles of these genes during infection and pathogenesis.
Collapse
Affiliation(s)
- Shizhong Geng
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhicheng Liu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhijie Lin
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Paul Barrow
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire LE12 5RD, UK
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Qiuchun Li
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
17
|
Immunoproteomic analysis to identify Shiga toxin-producing Escherichia coli outer membrane proteins expressed during human infection. Infect Immun 2014; 82:4767-77. [PMID: 25156722 DOI: 10.1128/iai.02030-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Shiga-toxin producing Escherichia coli (STEC) is the etiologic agent of acute diarrhea, dysentery, and hemolytic-uremic syndrome (HUS). There is no approved vaccine for STEC infection in humans, and antibiotic use is contraindicated, as it promotes Shiga toxin production. In order to identify STEC-associated antigens and immunogenic proteins, outer membrane proteins (OMPs) were extracted from STEC O26:H11, O103, O113:H21, and O157:H7 strains, and commensal E. coli strain HS was used as a control. SDS-PAGE, two-dimensional-PAGE analysis, Western blot assays using sera from pediatric HUS patients and controls, and matrix-assisted laser desorption ionization-tandem time of flight analyses were used to identify 12 immunogenic OMPs, some of which were not reactive with control sera. Importantly, seven of these proteins have not been previously reported to be immunogenic in STEC strains. Among these seven proteins, OmpT and Cah displayed IgG and IgA reactivity with sera from HUS patients. Genes encoding these two proteins were present in a majority of STEC strains. Knowledge of the antigens produced during infection of the host and the immune response to those antigens will be important for future vaccine development.
Collapse
|
18
|
Landstorfer R, Simon S, Schober S, Keim D, Scherer S, Neuhaus K. Comparison of strand-specific transcriptomes of enterohemorrhagic Escherichia coli O157:H7 EDL933 (EHEC) under eleven different environmental conditions including radish sprouts and cattle feces. BMC Genomics 2014; 15:353. [PMID: 24885796 PMCID: PMC4048457 DOI: 10.1186/1471-2164-15-353] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 03/31/2014] [Indexed: 12/26/2022] Open
Abstract
Background Multiple infection sources for enterohemorrhagic Escherichia coli O157:H7 (EHEC) are known, including animal products, fruit and vegetables. The ecology of this pathogen outside its human host is largely unknown and one third of its annotated genes are still hypothetical. To identify genetic determinants expressed under a variety of environmental factors, we applied strand-specific RNA-sequencing, comparing the SOLiD and Illumina systems. Results Transcriptomes of EHEC were sequenced under 11 different biotic and abiotic conditions: LB medium at pH4, pH7, pH9, or at 15°C; LB with nitrite or trimethoprim-sulfamethoxazole; LB-agar surface, M9 minimal medium, spinach leaf juice, surface of living radish sprouts, and cattle feces. Of 5379 annotated genes in strain EDL933 (genome and plasmid), a surprising minority of only 144 had null sequencing reads under all conditions. We therefore developed a statistical method to distinguish weakly transcribed genes from background transcription. We find that 96% of all genes and 91.5% of the hypothetical genes exhibit a significant transcriptional signal under at least one condition. Comparing SOLiD and Illumina systems, we find a high correlation between both approaches for fold-changes of the induced or repressed genes. The pathogenicity island LEE showed highest transcriptional activity in LB medium, minimal medium, and after treatment with antibiotics. Unique sets of genes, including many hypothetical genes, are highly up-regulated on radish sprouts, cattle feces, or in the presence of antibiotics. Furthermore, we observed induction of the shiga-toxin carrying phages by antibiotics and confirmed active biofilm related genes on radish sprouts, in cattle feces, and on agar plates. Conclusions Since only a minority of genes (2.7%) were not active under any condition tested (null reads), we suggest that the assumption of significant genome over-annotations is wrong. Environmental transcriptomics uncovered hitherto unknown gene functions and unique regulatory patterns in EHEC. For instance, the environmental function of azoR had been elusive, but this gene is highly active on radish sprouts. Thus, NGS-transcriptomics is an appropriate technique to propose new roles of hypothetical genes and to guide future research. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-353) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Klaus Neuhaus
- Lehrstuhl für Mikrobielle Ökologie, Wissenschaftszentrum Weihenstephan, Technische Universität München, Weihenstephaner Berg 3, D-85350 Freising, Germany.
| |
Collapse
|
19
|
Tao Q, Xiao J, Wang Y, Fang K, Li N, Hu M, Zhou Y, Zhao J. Identification of genes expressed during Toxoplasma gondii infection by in vivo-induced antigen technology (IVIAT) with positive porcine sera. J Parasitol 2014; 100:470-9. [PMID: 24646180 DOI: 10.1645/13-240.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Infection of pigs with Toxoplasma gondii is a common source of human toxoplasmosis and causes serious economic losses. In vivo-induced antigen technology (IVIAT) is an effective immunological technique to identify the antigens that a pathogen specifically expressed during infection. To discover the genes that are important in T. gondii infection of pigs, we employed IVIAT using sera from infected pigs. Fourteen antigens were identified including microneme protein 11 (MIC11), dense granule protein 5 (GRA5), 18 kDa cyclophilin (C-18), serine proteinase inhibitor (PI), calmodulin (CaM), leucine-rich repeat protein ( LRRP), D-3-phosphoglycerate dehydrogenase (D3PD), elongation factor 1-gamma (EF1), and 6 hypothetical proteins. The increased transcription levels of 5 (MIC11, GRA5, C-18, PI, and CaM) of the 14 molecules identified by IVIAT were confirmed by real-time PCR. The full length or partial proteins encoded by these 5 genes were expressed in Escherichia coli , and their immunogenicity was confirmed by Western blot analysis with positive porcine sera. Further functional studies were conducted with CaM. Suppression of CaM expression by RNA interference decreased T. gondii tachyzoites cell attachment, invasion, and egress but did not influence their replication. The proteins identified in this study are predicted to be involved in cell invasion, ion-protein binding, protein folding, biosynthesis, and metabolism. The results of the functional analysis support the hypothesis that CaM contributes to parasite pathogenesis during infection. These results may have significant implications for the discovery of candidate molecules for the development of potential therapies and preventive measures against toxoplasmosis in pigs.
Collapse
Affiliation(s)
- Qing Tao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
LaRocque RC, Harris JB, Ryan ET, Qadri F, Calderwood SB. Postgenomic approaches to cholera vaccine development. Expert Rev Vaccines 2014; 5:337-46. [PMID: 16827618 DOI: 10.1586/14760584.5.3.337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cholera remains an important public health threat. A cholera vaccine that provides durable protection at the mucosal surface, especially among children in endemic settings, is urgently needed. The availability of the complete genome sequence of a clinical isolate of Vibrio cholerae O1 El Tor has allowed for comparative and functional genomic approaches in the study of cholera. This work holds promise for the identification of bacterial targets of protective human immune responses and may contribute to the development of a new generation of cholera vaccines.
Collapse
Affiliation(s)
- Regina C LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, GRJ 504, 55 Fruit Street, Boston, MA 02114, USA.
| | | | | | | | | |
Collapse
|
21
|
Amerizadeh A, Khoo BY, Teh AY, Golkar M, Abdul Karim IZ, Osman S, Yunus MH, Noordin R. Identification and real-time expression analysis of selected Toxoplasma gondii in-vivo induced antigens recognized by IgG and IgM in sera of acute toxoplasmosis patients. BMC Infect Dis 2013; 13:287. [PMID: 23800344 PMCID: PMC3695809 DOI: 10.1186/1471-2334-13-287] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 06/13/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is an obligate intracellular zoonotic parasite of the phylum Apicomplexa which infects a wide range of warm-blooded animals, including humans. In this study in-vivo induced antigens of this parasite was investigated using in-vivo induced antigen technology (IVIAT) and pooled sera from patients with serological evidence of acute infection. METHODS The pooled sera was first pre-absorbed against three different preparations of antigens from in-vitro-grown cells of each T. gondii and E. coli XL1-Blue MRF', subsequently it was used to screen T. gondii cDNA phage expression library. Positive clones from each group were subjected to quantitative real-time PCR expression analysis on mRNA of in-vivo and in-vitro grown parasites. RESULTS A total of 29 reactive clones from each IgM and IgG immunoscreenings were found to have high homology to T. gondii genes. Quantitative real-time PCR expression analysis showed that 20 IgM-detected genes and 11 IgG-detected genes were up-regulated in-vivo relative to their expression levels in-vitro. These included genes encoding micronemes, sterol-regulatory element binding protein site, SRS34A, MIC2-associated protein M2AP, nucleoredoxin, protein phosphatase 2C and several hypothetical proteins. A hypothetical protein (GenBank accession no. 7899266) detected by IgG had the highest in-vivo over in-vitro fold change of 499.86; while another up-regulated hypothetical protein (GenBank accession no. 7898829) recognized by IgM showed high sensitivity (90%) and moderate specificity (70%) in detecting T. gondii antibodies when tested with 20 individual serum samples. CONCLUSION The highly up-regulated genes and the corresponding proteins, in particular the hypothetical proteins, may be useful in further studies on understanding the disease pathogenesis and as potential vaccine candidates.
Collapse
Affiliation(s)
- Atefeh Amerizadeh
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Identification of Salmonella enterica serovar Pullorum antigenic determinants expressed in vivo. Infect Immun 2013; 81:3119-27. [PMID: 23774596 DOI: 10.1128/iai.00145-13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Salmonella enterica serovar Pullorum affecting poultry causes pullorum disease and results in severe economic loss in the poultry industry. Currently, it remains a major threat in countries with poor poultry surveillance and no efficient control measures. As S. Pullorum could induce strong humoral immune responses, we applied an immunoscreening technique, the in vivo-induced antigen technology (IVIAT), to identify immunogenic bacterial proteins expressed or upregulated during S. Pullorum infection. Convalescent-phase sera from chickens infected with S. Pullorum were pooled, adsorbed against antigens expressed in vitro, and used to screen an S. Pullorum genomic expression library. Forty-five proteins were screened out, and their functions were implicated in molecular biosynthesis and degradation, transport, metabolism, regulation, cell wall synthesis and antibiotic resistance, environmental adaptation, or putative functions. In addition, 11 of these 45 genes were assessed for their differential expression by quantitative real-time reverse transcription-PCR (RT-PCR), revealing that 9 of 11 genes were upregulated to different degrees under in vivo conditions, especially the regulator of virulence determinants, phoQ. Then, four in vivo-induced proteins (ShdA, PhoQ, Cse3, and PbpC) were tested for their immunoreactivity in 28 clinical serum samples from chickens infected with S. Pullorum. The rate of detection of antibodies against ShdA reached 82% and was the highest among these proteins. ShdA is a host colonization factor known to be upregulated in vivo and related to the persistence of S. Typhimurium in the intestine. Furthermore, these antigens identified by IVIAT warrant further evaluation for their contributions to pathogenesis, and more potential roles, such as diagnostic, therapeutic, and preventive uses, need to be developed in future studies.
Collapse
|
23
|
Pustelny C, Brouwer S, Müsken M, Bielecka A, Dötsch A, Nimtz M, Häussler S. The peptide chain release factor methyltransferase PrmC is essential for pathogenicity and environmental adaptation of Pseudomonas aeruginosa PA14. Environ Microbiol 2012; 15:597-609. [PMID: 23278968 DOI: 10.1111/1462-2920.12040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 11/02/2012] [Indexed: 01/30/2023]
Abstract
Pseudomonas aeruginosa pathogenicity and its capability to adapt to multiple environments are dependent on the production of diverse virulence factors, controlled by the sophisticated quorum sensing (QS) network of P. aeruginosa. To better understand the molecular mechanisms that underlie this adaptation we searched for novel key regulators of virulence factor production by screening a PA14 transposon mutant library for potential candidates acting downstream of the unique 2-alkyl-4-quinolone (AQ) QS system of P. aeruginosa. We focused the work on a protein named HemK with high homology to PrmC of Escherichia coli displaying a similar enzymatic activity (therefore also referred to as PrmC). In this study, we demonstrate that PrmC is an S-adenosyl-l-methionine (AdoMet)-dependent methyltransferase of peptide chain release factors (RFs) essential for the expression of several virulence factors, such as pyocyanin, rhamnolipids and the type III-secreted toxin ExoT. Furthermore, the PA14_prmC mutant strain is unable to grow under anoxic conditions and has a significantly reduced pathogenicity in the infection model Galleria mellonella. Along with transcriptomic and proteomic analyses, the presented data indicate that the methylation of RFs in P. aeruginosa seems to have a global effect on cellular processes related to the virulence of this nosocomial pathogen.
Collapse
Affiliation(s)
- Christian Pustelny
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.
| | | | | | | | | | | | | |
Collapse
|
24
|
Identification of Toxoplasma gondii in-vivo induced antigens by cDNA library immunoscreening with chronic toxoplasmosis sera. Microb Pathog 2012; 54:60-6. [PMID: 23044055 DOI: 10.1016/j.micpath.2012.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/01/2012] [Accepted: 09/16/2012] [Indexed: 11/22/2022]
Abstract
Toxoplasmosis is an infection caused by the parasite Toxoplasma gondii. Chronically-infected individuals with a compromised immune system are at risk for reactivation of the disease. In-vivo induced antigen technology (IVIAT) is a promising method for the identification of antigens expressed in-vivo. The aim of the present study was to apply IVIAT to identify antigens which are expressed in-vivo during T. gondii infection using sera from individuals with chronic toxoplasmosis. Forty serum samples were pooled, pre-adsorped against three different preparations of antigens, from each in-vitro grown T. gondii and Escherichia coli XLBlue MRF', and then used to screen a T. gondii cDNA expression library. Sequencing of DNA inserts from positive clones showed eight open reading frames with high homology to T. gondii genes. Expression analysis using quantitative real-time PCR showed that SAG1-related sequence 3 (SRS3) and two hypothetical genes were up-regulated in-vivo relative to their expression levels in-vitro. These three proteins also showed high sensitivity and specificity when tested with individual serum samples. Five other proteins namely M16 domain peptidase, microneme protein, elongation factor 1-alpha, pre-mRNA-splicing factor and small nuclear ribonucleoprotein F had lower RNA expression in-vivo as compared to in-vitro. SRS3 and the two hypothetical proteins warrant further investigation into their roles in the pathogenesis of toxoplasmosis.
Collapse
|
25
|
Yan X, Fratamico PM, Needleman DS, Bayles DO. DNA sequence and analysis of a 90.1-kb plasmid in Shiga toxin-producing Escherichia coli (STEC) O145:NM 83-75. Plasmid 2012; 68:25-32. [DOI: 10.1016/j.plasmid.2012.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 02/09/2012] [Accepted: 02/12/2012] [Indexed: 10/28/2022]
|
26
|
Kudva IT, Griffin RW, Krastins B, Sarracino DA, Calderwood SB, John M. Proteins other than the locus of enterocyte effacement-encoded proteins contribute to Escherichia coli O157:H7 adherence to bovine rectoanal junction stratified squamous epithelial cells. BMC Microbiol 2012; 12:103. [PMID: 22691138 PMCID: PMC3420319 DOI: 10.1186/1471-2180-12-103] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 06/12/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In this study, we present evidence that proteins encoded by the Locus of Enterocyte Effacement (LEE), considered critical for Escherichia coli O157 (O157) adherence to follicle-associated epithelial (FAE) cells at the bovine recto-anal junction (RAJ), do not appear to contribute to O157 adherence to squamous epithelial (RSE) cells also constituting this primary site of O157 colonization in cattle. RESULTS Antisera targeting intimin-γ, the primary O157 adhesin, and other essential LEE proteins failed to block O157 adherence to RSE cells, when this pathogen was grown in DMEM, a culture medium that enhances expression of LEE proteins. In addition, RSE adherence of a DMEM-grown-O157 mutant lacking the intimin protein was comparable to that seen with its wild-type parent O157 strain grown in the same media. These adherence patterns were in complete contrast to that observed with HEp-2 cells (the adherence to which is mediated by intimin-γ), assayed under same conditions. This suggested that proteins other than intimin-γ that contribute to adherence to RSE cells are expressed by this pathogen during growth in DMEM. To identify such proteins, we defined the proteome of DMEM-grown-O157 (DMEM-proteome). GeLC-MS/MS revealed that the O157 DMEM-proteome comprised 684 proteins including several components of the cattle and human O157 immunome, orthologs of adhesins, hypothetical secreted and outer membrane proteins, in addition to the known virulence and LEE proteins. Bioinformatics-based analysis of the components of the O157 DMEM proteome revealed several new O157-specific proteins with adhesin potential. CONCLUSION Proteins other than LEE and intimin-γ proteins are involved in O157 adherence to RSE cells at the bovine RAJ. Such proteins, with adhesin potential, are expressed by this human pathogen during growth in DMEM. Ongoing experiments to evaluate their role in RSE adherence should provide both valuable insights into the O157-RSE interactions and new targets for more efficacious anti-adhesion O157 vaccines.
Collapse
Affiliation(s)
- Indira T Kudva
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, Iowa, 50010, USA
| | - Robert W Griffin
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, 02114, USA
| | - Bryan Krastins
- Harvard Partners Center for Genetics and Genomics, 65 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
- Present Address: Thermo-Fisher Scientific, Cambridge, Massachusetts, 02139, USA
| | - David A Sarracino
- Harvard Partners Center for Genetics and Genomics, 65 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
- Present Address: Thermo-Fisher Scientific, Cambridge, Massachusetts, 02139, USA
| | - Stephen B Calderwood
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, 02114, USA
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Manohar John
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, 02114, USA
- Present Address: Pathovacs Inc., Ames, Iowa, 50010, USA
| |
Collapse
|
27
|
Crépin S, Chekabab SM, Le Bihan G, Bertrand N, Dozois CM, Harel J. The Pho regulon and the pathogenesis of Escherichia coli. Vet Microbiol 2011; 153:82-8. [PMID: 21700403 DOI: 10.1016/j.vetmic.2011.05.043] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 05/18/2011] [Accepted: 05/25/2011] [Indexed: 11/18/2022]
Abstract
During the course of infection, bacteria must coordinately regulate gene expression in response to environmental stimuli. The phosphate (Pho) regulon is controlled by the two component-regulatory system PhoBR. PhoBR is activated during starvation and regulates genes involved in phosphate homeostasis. Several studies have highlighted the importance of the Pho regulon in bacterial pathogenesis, showing how induction of PhoBR, in addition to regulating genes participating in phosphate metabolism, leads to modulation of many cellular processes. The pleiotropic effects of Pho regulon activation include attenuated virulence and alteration of many virulence traits, including adhesion to host cells and resistance to cationic antimicrobial peptides, acidity and oxidative stresses. This review provides an overview of the relationship between the Pho regulon and virulence in Escherichia coli and illustrates that, in addition to regulating phosphate homeostasis, the Pho regulon plays a key role in regulating stress responses and virulence.
Collapse
Affiliation(s)
- Sébastien Crépin
- INRS-Institut Armand-Frappier, 531 Boul. des Prairies, Laval, Québec, Canada H7V 1B7
| | | | | | | | | | | |
Collapse
|
28
|
Li B, Tan Y, Guo J, Cui B, Wang Z, Wang H, Zhou L, Guo Z, Zhu Z, Du Z, Yang R. Use of protein microarray to identify gene expression changes ofYersinia pestisat different temperatures. Can J Microbiol 2011; 57:287-94. [DOI: 10.1139/w11-007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Yersinia pestis is a bacterium that is transmitted between fleas, which have a body temperature of 26 °C, and mammalian hosts, which have a body temperature of 37 °C. To adapt to the temperature shift, phenotype variations, including virulence, occur. In this study, an antigen microarray including 218 proteins of Y. pestis was used to evaluate antibody responses in a pooled plague serum that was unadsorbed, adsorbed by Y. pestis cultivated at 26 °C, or adsorbed by Y. pestis cultivated at 26 and 37 °C to identify protein expression changes during the temperature shift. We identified 12 proteins as being expressed at 37 °C but not at 26 °C, or expressed at significantly higher levels at 37 °C than at 26 °C. The antibodies against 7 proteins in the serum adsorbed by Y. pestis cultivated at 26 and 37 °C remained positive, suggesting that they were not expressed on the surface of Y. pestis in LB broth in vitro or specifically expressed in vivo. This study proved that protein microarray and antibody profiling comprise a promising technique for monitoring gene expression at the protein level and for better understanding pathogenicity, to find new vaccine targets against plague.
Collapse
Affiliation(s)
- Bei Li
- Taihe Hospital, Hubei Medical University, Shiyan, Hubei Province 442000, China
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yafang Tan
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Jingyu Guo
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Baizhong Cui
- Qinghai Institute for Endemic Diseases Prevention and Control, Xining, Qinghai Province 811602, China
| | - Zuyun Wang
- Qinghai Institute for Endemic Diseases Prevention and Control, Xining, Qinghai Province 811602, China
| | - Hu Wang
- Qinghai Institute for Endemic Diseases Prevention and Control, Xining, Qinghai Province 811602, China
| | - Lei Zhou
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Zhaobiao Guo
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ziwen Zhu
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Zongmin Du
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ruifu Yang
- Laboratory of Analytical Microbiology, State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| |
Collapse
|
29
|
Identification of in vivo-induced antigens including an RTX family exoprotein required for uropathogenic Escherichia coli virulence. Infect Immun 2011; 79:2335-44. [PMID: 21422188 DOI: 10.1128/iai.00110-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Uncomplicated urinary tract infections (UTI) are caused most commonly by uropathogenic Escherichia coli (UPEC). Whole-genome screening approaches, including transcriptomic, proteomic, and signature-tagged mutagenesis, have shown that UPEC highly expresses or requires genes for translational machinery, capsule, lipopolysaccharide, type 1 fimbriae, and iron acquisition systems during UTI. To identify additional genes expressed by UPEC during UTI, an immunoscreening approach termed in vivo-induced antigen technology (IVIAT) was employed to identify antigens produced during experimental infection that are not produced during in vitro culture. An inducible protein expression library, constructed from genomic DNA isolated from UPEC strain CFT073, was screened using exhaustively adsorbed pooled sera from 20 chronically infected female CBA/J mice. Using this approach, we identified 93 antigens induced by UPEC in vivo. A representative subset of these genes was tested by quantitative PCR for expression by CFT073 in vivo and during growth in human urine or LB medium in vitro; proWX, narJI, lolA, lolD, tosA (upxA), c2432, katG, ydhX, kpsS, and yddQ were poorly expressed in vitro but highly expressed in vivo. Of these, tosA, a gene encoding a predicted repeat-in-toxin family member, was expressed exclusively during UTI. Deletion of tosA in UPEC strain CFT073 resulted in significant attenuation in bladder and kidney infections during ascending UTI. By screening for in vivo-induced antigens, we identified a novel UPEC virulence factor and additional proteins that could be useful as potential vaccine targets.
Collapse
|
30
|
Le Maréchal C, Jardin J, Jan G, Even S, Pulido C, Guibert JM, Hernandez D, François P, Schrenzel J, Demon D, Meyer E, Berkova N, Thiéry R, Vautor E, Le Loir Y. Staphylococcus aureus seroproteomes discriminate ruminant isolates causing mild or severe mastitis. Vet Res 2011; 42:35. [PMID: 21324116 PMCID: PMC3052181 DOI: 10.1186/1297-9716-42-35] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 02/15/2011] [Indexed: 11/24/2022] Open
Abstract
Staphylococcus aureus is a major cause of mastitis in ruminants. In ewe mastitis, symptoms range from subclinical to gangrenous mastitis. S. aureus factors or host-factors contributing to the different outcomes are not completely elucidated. In this study, experimental mastitis was induced on primiparous ewes using two S. aureus strains, isolated from gangrenous (strain O11) or subclinical (strain O46) mastitis. Strains induced drastically distinct clinical symptoms when tested in ewe and mice experimental mastitis. Notably, they reproduced mild (O46) or severe (O11) mastitis in ewes. Ewe sera were used to identify staphylococcal immunoreactive proteins commonly or differentially produced during infections of variable severity and to define core and accessory seroproteomes. Such SERological Proteome Analysis (SERPA) allowed the identification of 89 immunoreactive proteins, of which only 52 (58.4%) were previously identified as immunogenic proteins in other staphylococcal infections. Among the 89 proteins identified, 74 appear to constitute the core seroproteome. Among the 15 remaining proteins defining the accessory seroproteome, 12 were specific for strain O11, 3 were specific for O46. Distribution of one protein specific for each mastitis severity was investigated in ten other strains isolated from subclinical or clinical mastitis. We report here for the first time the identification of staphylococcal immunogenic proteins common or specific to S. aureus strains responsible for mild or severe mastitis. These findings open avenues in S. aureus mastitis studies as some of these proteins, expressed in vivo, are likely to account for the success of S. aureus as a pathogen of the ruminant mammary gland.
Collapse
Affiliation(s)
- Caroline Le Maréchal
- INRA, UMR1253 Science et Technologie du Lait et de l'Œuf, F-35042 Rennes, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lee HR, Rhyu IC, Kim HD, Jun HK, Min BM, Lee SH, Choi BK. In-vivo-induced antigenic determinants of Fusobacterium nucleatum subsp. nucleatum. Mol Oral Microbiol 2011; 26:164-72. [DOI: 10.1111/j.2041-1014.2010.00594.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Kima PE, Bonilla JA, Cho E, Ndjamen B, Canton J, Leal N, Handfield M. Identification of Leishmania proteins preferentially released in infected cells using change mediated antigen technology (CMAT). PLoS Negl Trop Dis 2010; 4. [PMID: 20957202 PMCID: PMC2950143 DOI: 10.1371/journal.pntd.0000842] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 09/08/2010] [Indexed: 01/01/2023] Open
Abstract
Although Leishmania parasites have been shown to modulate their host cell's responses to multiple stimuli, there is limited evidence that parasite molecules are released into infected cells. In this study, we present an implementation of the change mediated antigen technology (CMAT) to identify parasite molecules that are preferentially expressed in infected cells. Sera from mice immunized with cell lysates prepared from L. donovani or L. pifanoi-infected macrophages were adsorbed with lysates of axenically grown amastigotes of L. donovani or L. pifanoi, respectively, as well as uninfected macrophages. The sera were then used to screen inducible parasite expression libraries constructed with genomic DNA. Eleven clones from the L. pifanoi and the L. donovani screen were selected to evaluate the characteristics of the molecules identified by this approach. The CMAT screen identified genes whose homologs encode molecules with unknown function as well as genes that had previously been shown to be preferentially expressed in the amastigote form of the parasite. In addition a variant of Tryparedoxin peroxidase that is preferentially expressed within infected cells was identified. Antisera that were then raised to recombinant products of the clones were used to validate that the endogenous molecules are preferentially expressed in infected cells. Evaluation of the distribution of the endogenous molecules in infected cells showed that some of these molecules are secreted into parasitophorous vacuoles (PVs) and that they then traffic out of PVs in vesicles with distinct morphologies. This study is a proof of concept study that the CMAT approach can be applied to identify putative Leishmania parasite effectors molecules that are preferentially expressed in infected cells. In addition we provide evidence that Leishmania molecules traffic out of the PV into the host cell cytosol and nucleus. Leishmania are intracellular parasites that reside within parasitophorous vacuoles (PV) in phagocytes. From within these compartments parasites control the host cell's responses to multiple stimuli. There is limited knowledge of the molecules that Leishmania parasites elaborate in the host cell to target processes therein. Furthermore, the mechanism by which such molecules would access their targets beyond the PV is not known. In the study presented here, we implemented the change mediated antigen technology (CMAT) to identify parasite molecules that are preferentially expressed inside infected cells. The approach was based on the reasoning that parasites express ‘new’ or antigenically modified molecules in the intracellular environment; therefore antiserum that is reactive to infected cells would contain immunoglobulins that are specific to these ‘new’ molecules. After adsorption of the antiserum with axenically cultured parasites, the antiserum was used to screen a parasite genomic expression library to identify genes encoding the molecules that are preferentially expressed in infected cells. We present for the first time evidence that some of these CMAT molecules accumulate in the PV and then traffic into the host cell in vesicles of distinct morphologies. Furthermore, several of these parasite molecules become localized in discrete compartments within the host cell.
Collapse
Affiliation(s)
- Peter E Kima
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America.
| | | | | | | | | | | | | |
Collapse
|
33
|
Wallet SM, Chung J, Handfield M. Use of in vivo-induced antigen technology (IVIAT) to identify virulence factors of Porphyromonas gingivalis. Methods Mol Biol 2010; 666:181-195. [PMID: 20717786 DOI: 10.1007/978-1-60761-820-1_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Porphyromonas gingivalis is a Gram-negative anaerobic bacterium associated with the initiation and progression of adult periodontal disease. The pathogenicity of P. gingivalis is multifaceted and the infection process is influenced by both microbial and host factors. It is generally accepted that genes of a pathogen that are specifically expressed during infection are likely to be important for pathogenicity. Numerous technologies have been developed to identify these genes. A novel strategy known as in vivo-induced antigen technology (IVIAT) avoids the use of animal models and utilizes serum from patients who have experienced disease caused by the pathogen of interest. While a number of putative virulence factors have been described for P. gingivalis, the identity, relevance, and mechanisms of action of virulence factors that actually provide a selective advantage to the organism in the oral cavity of diseased patients is still unclear. Here we describe the IVIAT protocol for identification of in vivo-induced genes of P. gingivalis, which can be adapted with few modifications to any microbial pathogen.
Collapse
Affiliation(s)
- Shannon M Wallet
- Department of Periodontology, University of Florida College of Dentistry, Gainesville, FL, USA
| | | | | |
Collapse
|
34
|
Gu H, Zhu H, Lu C. Use of in vivo-induced antigen technology (IVIAT) for the identification of Streptococcus suis serotype 2 in vivo-induced bacterial protein antigens. BMC Microbiol 2009; 9:201. [PMID: 19765272 PMCID: PMC2758882 DOI: 10.1186/1471-2180-9-201] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2009] [Accepted: 09/18/2009] [Indexed: 11/10/2022] Open
Abstract
Background Streptococcus suis serotype 2 (SS2) is a zoonotic agent that causes death and disease in both humans and swine. A better understanding of SS2-host molecular interactions is crucial for understanding SS2 pathogenesis and immunology. Conventional genetic and biochemical approaches used to study SS2 virulence factors are unable to take into account the complex and dynamic environmental stimuli associated with the infection process. In this study, in vivo-induced antigen technology (IVIAT), an immunoscreening technique, was used to identify the immunogenic bacterial proteins that are induced or upregulated in vivo during SS2 infection. Results Convalescent-phase sera from pigs infected with SS2 were pooled, adsorbed against in vitro antigens, and used to screen SS2 genomic expression libraries. Upon analysis of the identified proteins, we were able to assign a putative function to 40 of the 48 proteins. These included proteins implicated in cell envelope structure, regulation, molecule synthesis, substance and energy metabolism, transport, translation, and those with unknown functions. The in vivo-induced changes in the expression of 10 of these 40 genes were measured using real-time reverse transcription (RT)-PCR, revealing that the expression of 6 of the 10 genes was upregulated in the in vivo condition. The strain distribution of these 10 genes was analyzed by PCR, and they were found in the most virulent SS2 strains. In addition, protein sequence alignments of the newly identified proteins demonstrate that three are putative virulence-associated proteins. Conclusion Collectively, our results suggest that these in vivo-induced or upregulated genes may contribute to SS2 disease development. We hypothesize that the identification of factors specifically induced or upregulated during SS2 infection will aid in our understanding of SS2 pathogenesis and may contribute to the control SS2 outbreaks. In addition, the proteins identified using IVIAT may be useful potential vaccine candidates or virulence markers.
Collapse
Affiliation(s)
- Hongwei Gu
- Key Lab Animal Disease Diagnostic & Immunology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, PR China.
| | | | | |
Collapse
|
35
|
Chaudhuri RR, Peters SE, Pleasance SJ, Northen H, Willers C, Paterson GK, Cone DB, Allen AG, Owen PJ, Shalom G, Stekel DJ, Charles IG, Maskell DJ. Comprehensive identification of Salmonella enterica serovar typhimurium genes required for infection of BALB/c mice. PLoS Pathog 2009; 5:e1000529. [PMID: 19649318 PMCID: PMC2712085 DOI: 10.1371/journal.ppat.1000529] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 07/06/2009] [Indexed: 01/13/2023] Open
Abstract
Genes required for infection of mice by Salmonella Typhimurium can be identified by the interrogation of random transposon mutant libraries for mutants that cannot survive in vivo. Inactivation of such genes produces attenuated S. Typhimurium strains that have potential for use as live attenuated vaccines. A quantitative screen, Transposon Mediated Differential Hybridisation (TMDH), has been developed that identifies those members of a large library of transposon mutants that are attenuated. TMDH employs custom transposons with outward-facing T7 and SP6 promoters. Fluorescently-labelled transcripts from the promoters are hybridised to whole-genome tiling microarrays, to allow the position of the transposon insertions to be determined. Comparison of microarray data from the mutant library grown in vitro (input) with equivalent data produced after passage of the library through mice (output) enables an attenuation score to be determined for each transposon mutant. These scores are significantly correlated with bacterial counts obtained during infection of mice using mutants with individual defined deletions of the same genes. Defined deletion mutants of several novel targets identified in the TMDH screen are effective live vaccines.
Collapse
Affiliation(s)
- Roy R. Chaudhuri
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sarah E. Peters
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J. Pleasance
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Helen Northen
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Chrissie Willers
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Gavin K. Paterson
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Danielle B. Cone
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Gil Shalom
- Arrow Therapeutics Ltd., London, United Kingdom
| | - Dov J. Stekel
- Centre for Systems Biology, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Ian G. Charles
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | - Duncan J. Maskell
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Identification and characterization of antigenic proteins potentially expressed during the infectious process of Paracoccidioides brasiliensis. Microbes Infect 2009; 11:895-903. [PMID: 19500685 DOI: 10.1016/j.micinf.2009.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 05/05/2009] [Accepted: 05/24/2009] [Indexed: 11/23/2022]
Abstract
Paracoccidioides brasiliensis causes paracoccidioidomycosis (PCM), a systemic mycosis presenting clinical manifestations ranging from mild to severe forms. A P. brasiliensis cDNA expression library was produced and screened with pooled sera from PCM patients adsorbed against antigens derived from in vitro-grown P. brasiliensis yeast cells. Sequencing DNA inserts from clones reactive with PCM patients sera indicated 35 open reading frames presenting homology to genes involved in metabolic pathways, transport, among other predicted functions. The complete cDNAs encoding aromatic-l-amino-acid decarboxylase (Pbddc), lumazine synthase (Pbls) and a homologue of the high affinity copper transporter (Pbctr3) were obtained. Recombinant proteins PbDDC and PbLS were obtained; a peptide was synthesized for PbCTR3. The proteins and the synthetic peptide were recognized by sera of patients with confirmed PCM and not by sera of healthy patients. Using the in vivo-induced antigen technology (IVIAT), we identified immunogenic proteins expressed at high levels during infection. Quantitative real time RT-PCR demonstrated high transcript levels of Pbddc, Pbls and Pbctr3 in yeast cells infecting macrophages. Transcripts in yeast cells derived from spleen and liver of infected mice were also measured by qRT-PCR. Our results suggest a putative role for the immunogenic proteins in the infectious process of P. brasiliensis.
Collapse
|
37
|
Su YC, Wan KL, Mohamed R, Nathan S. A genome level survey of Burkholderia pseudomallei immunome expressed during human infection. Microbes Infect 2008; 10:1335-45. [PMID: 18761419 DOI: 10.1016/j.micinf.2008.07.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 07/22/2008] [Accepted: 07/30/2008] [Indexed: 11/26/2022]
Abstract
Burkholderia pseudomallei is the etiological agent of melioidosis, a severe infectious disease of humans and animals. The role of the bacterium's proteins expressed in vivo during human melioidosis continues to remain an enigma. This study's aim was to identify B. pseudomallei target proteins that elicit the humoral immune response in infected humans. A small insert genomic expression library was constructed and immunoscreened to identify peptides that reacted exclusively with melioidosis patients' sera. Sero-positive clones expressing immunogenic peptides were sequenced and annotated, and shown to represent 109 proteins involved in bacterial cell envelope biogenesis, cell motility and secretion, transcription, amino acid, ion and protein metabolism, energy production, DNA repair and unknown hypothetical proteins. Western blot analysis of three randomly selected full-length immunogenic polypeptides with patients' sera verified the findings of the immunome screening. The patients' humoral immune response to the 109 proteins suggests the induction or significant upregulation of these proteins in vivo during human infection and thus may play a role in the pathogenesis of B. pseudomallei. Identification of B. pseudomallei immunogens has shed new light on the elucidation of the bacterium's pathogenesis mechanism and disease severity. These immunogens can be further evaluated as prophylactic and serodiagnostic candidates as well as drug targets.
Collapse
Affiliation(s)
- Yu-Ching Su
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor Darul Ehsan, Malaysia
| | | | | | | |
Collapse
|
38
|
Kudva IT, Calderwood SB, John M. Proteomics-based expression library screening - a platform technology for rapid discovery of pathogen-specific markers of infection. ACTA ACUST UNITED AC 2008; 2:979-89. [PMID: 23495870 DOI: 10.1517/17530059.2.8.979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The development of new management modalities is imperative for reducing the global burden of infectious diseases. OBJECTIVE To develop a platform technology for rapid definition of comprehensive pathogen immunoproteomes (the repertoire of microbial proteins that elicit and interact with host immune responses). METHODS Standard molecular biology techniques were coupled with cutting-edge proteomics to develop proteomics-based expression library screening (PELS). RESULTS Proof of principle of PELS was demonstrated by defining a comprehensive immunoproteome of the human gastrointestinal pathogen, Escherichia coli O157:H7, in bovine reservoirs in just 3 weeks. CONCLUSIONS PELS, an immunoproteomics-based platform technology, is a rapid and inexpensive alternative to protein/antigen arrays/chips. It is applicable to any parasitic, fungal, viral or bacterial pathogen (or commensal) that: has a sequenced genome; can be cultured in the laboratory; and elicits host humoral immune responses during the process of infection/disease.
Collapse
Affiliation(s)
- Indira T Kudva
- Massachusetts General Hospital, Division of Infectious Diseases, Boston, MA 02114, USA +1 781 244 4505 ;
| | | | | |
Collapse
|
39
|
Rollins SM, Peppercorn A, Young JS, Drysdale M, Baresch A, Bikowski MV, Ashford DA, Quinn CP, Handfield M, Hillman JD, Lyons CR, Koehler TM, Calderwood SB, Ryan ET. Application of in vivo induced antigen technology (IVIAT) to Bacillus anthracis. PLoS One 2008; 3:e1824. [PMID: 18350160 PMCID: PMC2265799 DOI: 10.1371/journal.pone.0001824] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 02/05/2008] [Indexed: 01/23/2023] Open
Abstract
In vivo induced antigen technology (IVIAT) is an immuno-screening technique that identifies bacterial antigens expressed during infection and not during standard in vitro culturing conditions. We applied IVIAT to Bacillus anthracis and identified PagA, seven members of a N-acetylmuramoyl-L-alanine amidase autolysin family, three P60 family lipoproteins, two transporters, spore cortex lytic protein SleB, a penicillin binding protein, a putative prophage holin, respiratory nitrate reductase NarG, and three proteins of unknown function. Using quantitative real-time PCR comparing RNA isolated from in vitro cultured B. anthracis to RNA isolated from BALB/c mice infected with virulent Ames strain B. anthracis, we confirmed induced expression in vivo for a subset of B. anthracis genes identified by IVIAT, including L-alanine amidases BA3767, BA4073, and amiA (pXO2-42); the bacteriophage holin gene BA4074; and pagA (pXO1-110). The exogenous addition of two purified putative autolysins identified by IVIAT, N-acetylmuramoyl-L-alanine amidases BA0485 and BA2446, to vegetative B. anthracis cell suspensions induced a species-specific change in bacterial morphology and reduction in viable bacterial cells. Many of the proteins identified in our screen are predicted to affect peptidoglycan re-modeling, and our results support significant cell wall structural remodeling activity during B. anthracis infection. Identification of L-alanine amidases with B. anthracis specificity may suggest new potential therapeutic targets.
Collapse
Affiliation(s)
- Sean M Rollins
- Massachusetts General Hospital, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Huang Y, Leming CL, Suyemoto M, Altier C. Genome-wide screen of Salmonella genes expressed during infection in pigs, using in vivo expression technology. Appl Environ Microbiol 2007; 73:7522-30. [PMID: 17921269 PMCID: PMC2168049 DOI: 10.1128/aem.01481-07] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Accepted: 09/26/2007] [Indexed: 11/20/2022] Open
Abstract
Pigs are a food-producing species that readily carry Salmonella but, in the great majority of cases, do not show clinical signs of disease. Little is known about the functions required by Salmonella to be maintained in pigs. We have devised a recombinase-based promoter-trapping strategy to identify genes with elevated expression during pig infection with Salmonella enterica serovar Typhimurium. A total of 55 clones with in vivo-induced promoters were selected from a genomic library of approximately 10,000 random Salmonella DNA fragments fused to the recombinase cre, and the cloned DNA fragments were analyzed by sequencing. Thirty-one genes encoding proteins involved in bacterial adhesion and colonization (including bcfA, hscA, rffG, and yciR), virulence (metL), heat shock (hscA), and a sensor of a two-component regulator (hydH) were identified. Among the 55 clones, 19 were isolated from both the tonsils and the intestine, while 23 were identified only in the intestine and 13 only in tonsils. High temperature and increased osmolarity were identified as environmental signals that induced in vivo-expressed genes, suggesting possible signals for expression.
Collapse
Affiliation(s)
- Yanyan Huang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|
41
|
Garbom S, Olofsson M, Björnfot AC, Srivastava MK, Robinson VL, Oyston PCF, Titball RW, Wolf-Watz H. Phenotypic characterization of a virulence-associated protein, VagH, of Yersinia pseudotuberculosis reveals a tight link between VagH and the type III secretion system. MICROBIOLOGY-SGM 2007; 153:1464-1473. [PMID: 17464060 DOI: 10.1099/mic.0.2006/000323-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recently, a number of attenuated mutants of Yersinia pseudotuberculosis have been identified using a bioinformatics approach. One of the target genes identified in that study was vagH, which the authors now characterized further. VagH shows homology to HemK of Escherichia coli, possessing methyltransferase activity similar to that of HemK, and targeting release factors 1 and 2. Microarray studies comparing the wild-type and the vagH mutant revealed that the mRNA levels of only a few genes were altered in the mutant. By proteome analysis, expression of the virulence determinant YopD was found to be increased, indicating a possible connection between VagH and the virulence plasmid-encoded type III secretion system (T3SS). Further analysis showed that Yop expression and secretion were repressed in a vagH mutant. This phenotype could be suppressed by trans-complementation with the wild-type vagH gene or by deletion of the negative regulator yopD. Also, in a similar manner to a T3SS-negative mutant, the avirulent vagH mutant was rapidly cleared from Peyer's patches and could not reach the spleen after oral infection of mice. In a manner analogous to that of T3SS mutants, the vagH mutant could not block phagocytosis by macrophages. However, a vagH mutant showed no defects in the T3SS-independent ability to proliferate intracellularly and replicated to levels similar to those of the wild-type in macrophages. In conclusion, the vagH mutant exhibits a virulence phenotype similar to that of a T3SS-negative mutant, indicating a tight link between VagH and type III secretion in Y. pseudotuberculosis.
Collapse
Affiliation(s)
- Sara Garbom
- Department of Molecular Biology, Umeå University, S-90187 Umeå, Sweden
| | - Martina Olofsson
- Department of Molecular Biology, Umeå University, S-90187 Umeå, Sweden
| | | | - Manoj Kumar Srivastava
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, S-90183 Umeå, Sweden
| | | | - Petra C F Oyston
- Biomedical Sciences, Dstl Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Richard W Titball
- Biomedical Sciences, Dstl Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Hans Wolf-Watz
- Department of Molecular Biology, Umeå University, S-90187 Umeå, Sweden
| |
Collapse
|
42
|
Yoo JY, Kim HC, Zhu W, Kim SM, Sabet M, Handfield M, Hillman J, Progulske-Fox A, Lee SW. Identification of Tannerella forsythia antigens specifically expressed in patients with periodontal disease. FEMS Microbiol Lett 2007; 275:344-52. [PMID: 17868359 DOI: 10.1111/j.1574-6968.2007.00906.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Molecular pathogenesis of Tannerella forsythia, a putative periodontal pathogen, has not yet been adequately elucidated due to limited information on its virulence factors. Here, identification of in vivo expressed antigens of T. forsythia is reported using in vivo-induced antigen technology (IVIAT). Among 13 000 recombinant clones screened, 16 positive clones were identified that reacted reproducibly with sera obtained from patients with periodontal disease. DNA sequences from 12 of these in vivo-induced genes were determined. IVIAT-identified protein antigens of T. forsythia include: BspA, a well-defined virulence factor of T. forsythia; enzymes involved in housekeeping functions (tRNA synthetases, glycine hydroxymethyltransferase, and glucoside glucohydrolase); enzymes implicated in tissue destruction (dipeptidyl peptidase IV); a DNA mismatch repair protein; and putative outer membrane proteins of unknown function. The in vivo gene expression of these IVIAT-identified antigens was confirmed by a quantitative real-time PCR analysis. This is, to the best of the authors' knowledge, the first report using IVIAT in T. forsythia. It is anticipated that detailed analysis of the in vivo-induced genes identified by IVIAT in this study will lead to a better understanding of the molecular mechanisms mediating periodontal infection by T. forsythia.
Collapse
Affiliation(s)
- Ji Yeon Yoo
- Division of Periodontics, College of Dental Medicine, Columbia University, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yu WL, Dan H, Lin M. Novel protein targets of the humoral immune response to Listeria monocytogenes infection in rabbits. J Med Microbiol 2007; 56:888-895. [PMID: 17577052 DOI: 10.1099/jmm.0.46977-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The role of the humoral immune response in protective immunity against listerial infection has been overlooked and is essentially unknown. This study aimed to discover the protein targets of Listeria monocytogenes that elicit an antibody response following infection in a rabbit model. A genomic expression library for L. monocytogenes was constructed and differentially screened to identify genes encoding proteins that reacted with antiserum from rabbits infected with live L. monocytogenes serotype 4b (RalphaL), but not with that from animals immunized with heat-killed bacteria (RalphaK). Thirty-one clones expressing proteins that reacted exclusively with RalphaL were identified and sequenced. Sequence analysis, together with Western blot analysis of the proteins expressed from positive clones, led to the identification of eight L. monocytogenes proteins as targets of humoral immune responses during listerial infection: three internalin members (InlA, InlD and InlC2) and five novel proteins of unknown function (designated IspA, IspB, IspC, IspD and IspE, respectively). Exhibition of humoral immune responses to these proteins in actively infected rabbits but not in animals receiving heat-killed L. monocytogenes suggested that they were induced or significantly upregulated in vivo during infection and thus are important in Listeria pathogenesis. With the exception of antibodies to InlA, this is the first demonstration of antibodies to the other seven proteins in infected hosts. These immunogenic proteins may be useful candidates for elucidation of the role of antibodies in protective immunity in the context of listerial infection, as well as potential targets for serodiagnostic reagents and vaccine and drug development.
Collapse
Affiliation(s)
- Wei Ling Yu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Animal Diseases Research Institute, Ottawa, ON K2H 8P9, Canada
| | - Hanhong Dan
- Animal Diseases Research Institute, Ottawa, ON K2H 8P9, Canada
| | - Min Lin
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Animal Diseases Research Institute, Ottawa, ON K2H 8P9, Canada
| |
Collapse
|
44
|
Dziva F, Mahajan A, Cameron P, Currie C, McKendrick IJ, Wallis TS, Smith DGE, Stevens MP. EspP, a Type V-secreted serine protease of enterohaemorrhagic Escherichia coli O157:H7, influences intestinal colonization of calves and adherence to bovine primary intestinal epithelial cells. FEMS Microbiol Lett 2007; 271:258-64. [PMID: 17451446 DOI: 10.1111/j.1574-6968.2007.00724.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) comprise a group of zoonotic diarrhoeal pathogens of worldwide importance. Cattle are a key reservoir; however the molecular mechanisms that promote persistent colonization of the bovine intestines by EHEC are ill-defined. The large plasmid of EHEC O157:H7 encodes several putative virulence factors. Here, it is reported that the pO157-encoded Type V-secreted serine protease EspP influences the intestinal colonization of calves. To dissect the basis of attenuation, a bovine primary rectal epithelial cell line was developed. Adherence of E. coli O157:H7 to such cells was significantly impaired by espP mutation but restored upon addition of highly purified exogenous EspP. Data of this study add to the growing body of evidence that cytotoxins facilitate intestinal colonization by EHEC.
Collapse
Affiliation(s)
- Francis Dziva
- Division of Microbiology, Institute for Animal Health, Compton, Newbury, Berkshire, UK.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Sharp FC, Sperandio V. QseA directly activates transcription of LEE1 in enterohemorrhagic Escherichia coli. Infect Immun 2007; 75:2432-40. [PMID: 17339361 PMCID: PMC1865749 DOI: 10.1128/iai.02003-06] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Quorum sensing (QS) in enterohemorrhagic Escherichia coli (EHEC) regulates the expression of the locus of enterocyte effacement (LEE). The LEE contains five major operons named LEE1 through LEE5. QseA was previously shown to be activated through QS and to activate the transcription of LEE1. The LEE1 operon encodes Ler, the transcription activator of all other LEE genes, and has two promoters: a distal promoter (P1) and a proximal promoter (P2). We have previously reported that QseA acts on P1 and not P2. To identify the minimal region of LEE1 that is necessary for QseA-mediated activation, a series of nested-deletion constructs of the LEE1 promoter fused to a lacZ reporter were constructed in both the EHEC and E. coli K-12 backgrounds. In an EHEC background, QseA-dependent activation of LEE1 can be observed for the entire regulatory region (beginning at nucleotide -393 and ending at nucleotide -123). In contrast to what occurred in EHEC, in K-12 there was no QseA-dependent activation of LEE1 transcription between base pairs -393 and -343. These data indicate that a QseA-dependent EHEC-specific regulator is required for the activation of transcription in this region. We also observed QseA-dependent LEE1 activation from nucleotides -218 to -123 in K-12, similar to results of the nested-deletion analysis performed with EHEC. Electrophoretic mobility shift assays established that QseA directly binds to the region of LEE1 from bp -173 to -42 and not to the region from bp -393 to -343. These studies suggest that QseA activates the transcription of LEE1 by directly binding upstream of its P1 promoter region.
Collapse
Affiliation(s)
- Faith C Sharp
- Department of Microbiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9048, USA
| | | |
Collapse
|
46
|
Hancock V, Klemm P. Global gene expression profiling of asymptomatic bacteriuria Escherichia coli during biofilm growth in human urine. Infect Immun 2007; 75:966-76. [PMID: 17145952 PMCID: PMC1828481 DOI: 10.1128/iai.01748-06] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Accepted: 11/17/2006] [Indexed: 11/20/2022] Open
Abstract
Urinary tract infection (UTI) is an important health problem worldwide, with many millions of cases each year, and Escherichia coli is the most common organism causing UTI in humans. Also, E. coli is responsible for most infections in patients with chronic indwelling bladder catheter. The two asymptomatic bacteriuria (ABU) E. coli strains 83972 and VR50 are significantly better biofilm formers in their natural growth medium, human urine, than the two uropathogenic E. coli isolates CFT073 and 536. We used DNA microarrays to monitor the expression profile during biofilm growth in urine of the two ABU strains 83972 and VR50. Significant differences in expression levels were seen between the biofilm expression profiles of the two strains with the corresponding planktonic expression profiles in morpholinepropanesulfonic acid minimal laboratory medium and human urine; 417 and 355 genes were up- and down-regulated, respectively, during biofilm growth in urine of 83972 and VR50. Many genes involved in transcription and stress were up-regulated in biofilm-grown cells. The role in biofilm formation of four of the up-regulated genes, i.e., yceP, yqgA, ygiD, and aaeX, was investigated by creating single-knockout mutant versions of 83972 and VR50; all mutants showed reduced biofilm formation in urine by 18 to 43% compared with the wild type (P < 0.05). Also, the expression profile of strain 83972 in the human urinary tract partially overlaps with the biofilm expression profile.
Collapse
Affiliation(s)
- Viktoria Hancock
- Microbial Adhesion Group, Center for Biomedical Microbiology, BioCentrum-DTU, Building 301, Technical University of Denmark, DK-2800 Lyngby, Denmark
| | | |
Collapse
|
47
|
Marra A. Targeting virulence for antibacterial chemotherapy: identifying and characterising virulence factors for lead discovery. Drugs R D 2006; 7:1-16. [PMID: 16620133 DOI: 10.2165/00126839-200607010-00001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The antibacterial drug discovery industry is fast losing participants; at the same time it is facing the challenge of developing new antibiotics that are effective against frequently occurring and multiply resistant organisms. One intriguing approach is to target bacterial virulence, and the last decade or so has seen a focus on bacterial pathogenesis along with the development of reagents and strategies that could make this possible. Several processes utilised by a range of bacteria to cause infection may be conserved enough to make attractive targets; indeed it is known that mammalian cells can affect bacterial gene expression and vice versa. Interesting targets involving virulence include type III secretion systems, two-component signal transduction systems, quorum sensing, and biofilm formation. In order to better understand these systems and strategies, investigators have developed novel strategies of their own, involving negative selections, surrogate models of infection, and screens for gene induction and antigenicity. Inhibitors of such targets would be unlikely to adversely affect patients, be cross-resistant to existing therapies, or cause resistance themselves. It might be the case that virulence target-based therapies would not be powerful enough to clear an existing infection alone, but if they are instead considered as adjunct therapy to existing antibiotics, or potentiators of the host immune response, they may show efficacy in a non-traditional way.
Collapse
|
48
|
Kudva IT, Krastins B, Sheng H, Griffin RW, Sarracino DA, Tarr PI, Hovde CJ, Calderwood SB, John M. Proteomics-based expression library screening (PELS): a novel method for rapidly defining microbial immunoproteomes. Mol Cell Proteomics 2006; 5:1514-9. [PMID: 16737953 PMCID: PMC2754196 DOI: 10.1074/mcp.t600013-mcp200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Current methodologies for global identification of microbial proteins that elicit host humoral immune responses have several limitations and are not ideally suited for use in the postgenomic era. Here we describe a novel application of proteomics, proteomics-based expression library screening, to rapidly define microbial immunoproteomes. Proteomics-based expression library screening is broadly applicable to any cultivable, sequenced pathogen eliciting host antibody responses and hence is ideal for rapidly mining microbial proteomes for targets with diagnostic, prophylactic, and therapeutic potential. In this report, we demonstrate "proof-of-principle" by identifying 207 proteins of the Escherichia coli O157:H7 immunome in bovine reservoirs in only 3 weeks.
Collapse
Affiliation(s)
- Indira T. Kudva
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
| | - Bryan Krastins
- Harvard Partners Center For Genetics and Genomics, Cambridge, Massachusetts 02139
| | - Haiqing Sheng
- Department of Microbiology, Molecular Biology, and Biochemistry, University of Idaho, Moscow, Idaho 83844
| | - Robert W. Griffin
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - David A. Sarracino
- Harvard Partners Center For Genetics and Genomics, Cambridge, Massachusetts 02139
| | - Phillip I. Tarr
- Departments of Pediatrics and Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Carolyn J. Hovde
- Department of Microbiology, Molecular Biology, and Biochemistry, University of Idaho, Moscow, Idaho 83844
| | - Stephen B. Calderwood
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts 02114
| | - Manohar John
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- To whom correspondence should be addressed. Tel.: 617−724−7528; Fax: 617−726−7416; E-mail:
| |
Collapse
|