1
|
Cherny KE, Balaji A, Mukherjee J, Goo YA, Hauser AR, Ozer E, Satchell KJF, Bachta KER, Kochan TJ, Mitra SD, Kociolek LK. Identification of Clostridium innocuum hypothetical protein that is cross-reactive with C. difficile anti-toxin antibodies. Anaerobe 2022; 75:102555. [PMID: 35367613 PMCID: PMC9197939 DOI: 10.1016/j.anaerobe.2022.102555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/01/2022]
Abstract
OBJECTIVES Previously considered solely an opportunistic pathogen, Clostridium innocuum (CI) was recently reported in Taiwan to be an emerging cause of antibiotic-associated diarrhea and clinically indistinguishable from Clostridioides difficile (CD) infection. We previously identified CI culture supernatant being cross-reactive with commercial CD toxin enzyme immunoassays. We aimed to identify and characterize the cross-reacting protein and determine whether it functioned as a human toxin. METHODS We performed western blots using CI culture supernatants and CD anti-toxin antibodies and identified interacting bands. We identified protein(s) using tandem mass spectrometry and evaluated them by cytotoxicity assays. RESULTS CI, but not CD, was isolated from stool of 12 children and adults with diarrhea. Culture supernatant from 6/12 CI isolates, and an ATCC reference strain, tested positive for CD toxins (total 7/13 isolates) by commercial EIA. Using two of these isolates, we identified two ∼40 kDa hypothetical proteins, CI_01447 and CI_01448, and confirmed cross-reactivity with CD anti-toxin antibodies by enzyme immunoassay and Western blot. Whole-genome sequencing confirmed all 13 isolates contained both genes, which were highly conserved. We observed no cytopathic or cytotoxic effects to HeLa cells when treated with these proteins. We identified amino acid sequence similarity to the NlpC/P60 family of proteins. CONCLUSIONS Our findings do not suggest CI proteins CI_01448 and CI_01447, which cross-react with antibodies against CD toxins A and B, are toxic to HeLa cells. Further studies are needed to determine the function of these cross-reacting proteins and the potential virulence factors that could be responsible for CI diarrheal disease.
Collapse
Affiliation(s)
- K E Cherny
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - A Balaji
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - J Mukherjee
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Y A Goo
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - A R Hauser
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - E Ozer
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - K J F Satchell
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA; Center for Structural Genomics of Infectious Diseases, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - K E R Bachta
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - T J Kochan
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - S D Mitra
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - L K Kociolek
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Abstract
The NlpC/p60-family of peptidoglycan hydrolases are key enzymes that facilitate bacterial cell division and also modulate microbe-host interactions. These endopeptidases utilize conserved Cys-His residues in their active site and are expressed in most bacterial species as well as some eukaryotes. Here we describe methods for biochemical analysis of Enterococcus faecium SagA-NlpC/p60 peptidoglycan hydrolase activity (Kim et al., 2019; Rangan et al., 2016), which includes recombinant protein preparation and biochemical analysis using both gel-based and LC-MS profiling of peptidoglycan fragments. These protocols should also facilitate the biochemical analysis of other NlpC/p60 peptidoglycan hydrolases.
Collapse
|
3
|
Hou S, Chen G, Wang W, Xia L, Wang Z, Lu Y. Identification of a cell-wall peptidase (NlpC/P60) from Nocardia seriolae which induces apoptosis in fathead minnow cells. JOURNAL OF FISH DISEASES 2020; 43:571-581. [PMID: 32196698 DOI: 10.1111/jfd.13154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 06/10/2023]
Abstract
Nocardia seriolae, a Gram-positive bacterium, is the main pathogen of fish nocardiosis. Protein NlpC/P60 is a cell-wall peptidase and a potential virulence factor of N. seriolae. Subcellular localization research revealed that both NlpC/P60-GFP and NlpC/P60Δsig-GFP fusion proteins were evenly distributed in the whole cell of fathead minnow (FHM) cells. Furthermore, typical apoptotic features, such as nuclear pyrosis and apoptotic bodies, were observed in the transfected FHM cells and grouper spleen cells by the overexpression of protein NlpC/P60. Then, quantitative assays of mitochondrial membrane potential (ΔΨm) value, caspase-3 activity and apoptosis-related gene (Bax, BNIP3, TNF1 and TNF6) mRNA expression were conducted. The results showed that ΔΨm was decreased, caspase-3 was significantly activated, and the mRNA expression of pro-apoptotic genes (Bax and BNIP3) and tumour necrosis factors (TNF1 and TNF6) was up-regulated in NlpC/P60-overexpressed cells. Taken together, the results indicated that the protein NlpC/P60 of N. seriolae might involve in apoptosis regulation. This study may lay the foundation for further study on the function of N. seriolae NlpC/P60 and promote the understanding of the virulence factors and pathogenic mechanism of N. seriolae.
Collapse
Affiliation(s)
- Suying Hou
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Guoquan Chen
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Wenji Wang
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Liqun Xia
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning, China
| | - Zhiwen Wang
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Yishan Lu
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning, China
| |
Collapse
|
4
|
Kayode AJ, Igbinosa EO, Okoh AI. Overview of listeriosis in the Southern African Hemisphere—Review. J Food Saf 2019. [DOI: 10.1111/jfs.12732] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Adeoye J. Kayode
- Applied and Environmental Microbiology Research Group (AEMREG), Department of Biochemistry and MicrobiologyUniversity of Fort Hare Alice South Africa
- SAMRC Microbial Water Quality Monitoring CenterUniversity of Fort Hare Alice South Africa
| | - Etinosa O. Igbinosa
- Department of Microbiology, Faculty of Life SciencesPrivate Mail Bag 1154, University of Benin Benin City Nigeria
| | - Anthony I. Okoh
- Applied and Environmental Microbiology Research Group (AEMREG), Department of Biochemistry and MicrobiologyUniversity of Fort Hare Alice South Africa
- SAMRC Microbial Water Quality Monitoring CenterUniversity of Fort Hare Alice South Africa
| |
Collapse
|
5
|
Bäuerl C, Abitayeva G, Sosa-Carrillo S, Mencher-Beltrán A, Navarro-Lleó N, Coll-Marqués JM, Zúñiga-Cabrera M, Shaikhin S, Pérez-Martinez G. P40 and P75 Are Singular Functional Muramidases Present in the Lactobacillus casei /paracasei/rhamnosus Taxon. Front Microbiol 2019; 10:1420. [PMID: 31297099 PMCID: PMC6607858 DOI: 10.3389/fmicb.2019.01420] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/05/2019] [Indexed: 01/29/2023] Open
Abstract
Lactobacillus casei and Lactobacillus rhamnosus proteins P40 and P75 belong to a large family of secreted cell wall proteins that contain a carboxy(C)-terminal CHAP or NlpC/P60 superfamily domains. In addition to their peptidoglycan hydrolases activity, proteins in this family are specific antigens of pathogens, frequently responsible of interactions with the host. L. rhamnosus GG and L. casei BL23 purified P40 and P75 proteins have antiapoptotic activity by inducing the EGF/Akt pathway. The aim of this work was to study the genetics, phylogeny and dissemination of this family of proteins in the genus Lactobacillus as well as their characteristics and likely function. The scrutiny of their DNA encoding sequences revealed the presence of minisatellite DNA in the P75 encoding gene of L. casei/paracasei strains (cmuB) with intraspecific indels that gave raise to four different alleles (cmuB1-4), which are exclusive of this species. Phylogenic analyses suggest that both proteins are present mainly in the L. casei and Lactobacillus sakei phylogenomic groups. A P40 ancestral gene was possibly present in the common ancestor of Enterococcaceae, Lactobacillaceae and Streptococcaceae. P75 is also present in L. casei and L. sakei groups, but its evolution is difficult to explain only by vertical transmission. Antibodies raised against the N-terminal regions of P40 and P75 improved their immunological detection in culture supernatants as they recognized almost exclusively proteins of L. casei/paracasei/rhamnosus strains, highlighting their structural similarity, that allowed to detect them in different fermented dairy products that contained probiotic L. casei strains. Purified P40 and P75 proteins showed no evident lytic activity but they complemented L. casei BL23 cmuA and cmuB defective mutants, respectively, thus proving that they actively participate in cell division.
Collapse
Affiliation(s)
- Christine Bäuerl
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - Gulyaim Abitayeva
- Department of Microbiology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan.,Laboratory of Genetics and Biochemistry of Microorganisms, Republican Collection of Microorganisms at Science Committee of Ministry of Education and Science RK, Astana, Kazakhstan
| | - Sebastián Sosa-Carrillo
- Computational Biology Department, Inria, Institut Pasteur and Université Paris Diderot, Paris, France
| | | | - Noemí Navarro-Lleó
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - José M Coll-Marqués
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - Manuel Zúñiga-Cabrera
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - Serik Shaikhin
- Department of Microbiology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan.,Laboratory of Genetics and Biochemistry of Microorganisms, Republican Collection of Microorganisms at Science Committee of Ministry of Education and Science RK, Astana, Kazakhstan
| | - Gaspar Pérez-Martinez
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| |
Collapse
|
6
|
|
7
|
Parthasarathy G, Lun S, Guo H, Ammerman NC, Geiman DE, Bishai WR. Rv2190c, an NlpC/P60 family protein, is required for full virulence of Mycobacterium tuberculosis. PLoS One 2012; 7:e43429. [PMID: 22952680 PMCID: PMC3432046 DOI: 10.1371/journal.pone.0043429] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 07/20/2012] [Indexed: 11/18/2022] Open
Abstract
Mycobacterium tuberculosis, the etiologic agent of tuberculosis (TB) possesses at least five genes predicted to encode proteins with NlpC/P60 hydrolase domains, including the relatively uncharacterized Rv2190c. As NlpC/P60 domain-containing proteins are associated with diverse roles in bacterial physiology, our objective was to characterize Rv2190c in M. tuberculosis growth and virulence. Our data indicate that lack of Rv2190c is associated with impaired growth, both in vitro and during an in vivo mouse model of TB. These growth defects are associated with altered colony morphology and phthiocerol dimycocerosate levels, indicating that Rv2190c is involved in cell wall maintenance and composition. In addition, we have demonstrated that Rv2190c is expressed during active growth phase and that its protein product is immunogenic during infection. Our findings have significant implications, both for better understanding the role of Rv2190c in M. tuberculosis biology and also for translational developments.
Collapse
Affiliation(s)
- Geetha Parthasarathy
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Shichun Lun
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Haidan Guo
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Nicole C. Ammerman
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Deborah E. Geiman
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William R. Bishai
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
- * E-mail:
| |
Collapse
|
8
|
Milillo SR, Friedly EC, Saldivar JC, Muthaiyan A, O'bryan C, Crandall PG, Johnson MG, Ricke SC. A Review of the Ecology, Genomics, and Stress Response ofListeria innocuaandListeria monocytogenes. Crit Rev Food Sci Nutr 2012; 52:712-25. [DOI: 10.1080/10408398.2010.507909] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
9
|
Actin polymerization drives septation of Listeria monocytogenes namA hydrolase mutants, demonstrating host correction of a bacterial defect. Infect Immun 2011; 79:1458-70. [PMID: 21263016 DOI: 10.1128/iai.01140-10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The Gram-positive bacterial cell wall presents a structural barrier that requires modification for protein secretion and large-molecule transport as well as for bacterial growth and cell division. The Gram-positive bacterium Listeria monocytogenes adjusts cell wall architecture to promote its survival in diverse environments that include soil and the cytosol of mammalian cells. Here we provide evidence for the enzymatic flexibility of the murein hydrolase NamA and demonstrate that bacterial septation defects associated with a loss of NamA are functionally complemented by physical forces associated with actin polymerization within the host cell cytosol. L. monocytogenes ΔnamA mutants formed long bacterial chains during exponential growth in broth culture; however, normal septation could be restored if mutant cells were cocultured with wild-type L. monocytogenes bacteria or by the addition of exogenous NamA. Surprisingly, ΔnamA mutants were not significantly attenuated for virulence in mice despite the pronounced exponential growth septation defect. The physical force of L. monocytogenes-mediated actin polymerization within the cytosol was sufficient to sever ΔnamA mutant intracellular chains and thereby enable the process of bacterial cell-to-cell spread so critical for L. monocytogenes virulence. The inhibition of actin polymerization by cytochalasin D resulted in extended intracellular bacterial chains for which septation was restored following drug removal. Thus, despite the requirement for NamA for the normal septation of exponentially growing L. monocytogenes cells, the hydrolase is essentially dispensable once L. monocytogenes gains access to the host cell cytosol. This phenomenon represents a notable example of eukaryotic host cell complementation of a bacterial defect.
Collapse
|
10
|
Yeoman CJ, Yildirim S, Thomas SM, Durkin AS, Torralba M, Sutton G, Buhay CJ, Ding Y, Dugan-Rocha SP, Muzny DM, Qin X, Gibbs RA, Leigh SR, Stumpf R, White BA, Highlander SK, Nelson KE, Wilson BA. Comparative genomics of Gardnerella vaginalis strains reveals substantial differences in metabolic and virulence potential. PLoS One 2010; 5:e12411. [PMID: 20865041 PMCID: PMC2928729 DOI: 10.1371/journal.pone.0012411] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2010] [Accepted: 07/22/2010] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Gardnerella vaginalis is described as a common vaginal bacterial species whose presence correlates strongly with bacterial vaginosis (BV). Here we report the genome sequencing and comparative analyses of three strains of G. vaginalis. Strains 317 (ATCC 14019) and 594 (ATCC 14018) were isolated from the vaginal tracts of women with symptomatic BV, while Strain 409-05 was isolated from a healthy, asymptomatic individual with a Nugent score of 9. PRINCIPAL FINDINGS Substantial genomic rearrangement and heterogeneity were observed that appeared to have resulted from both mobile elements and substantial lateral gene transfer. These genomic differences translated to differences in metabolic potential. All strains are equipped with significant virulence potential, including genes encoding the previously described vaginolysin, pili for cytoadhesion, EPS biosynthetic genes for biofilm formation, and antimicrobial resistance systems, We also observed systems promoting multi-drug and lantibiotic extrusion. All G. vaginalis strains possess a large number of genes that may enhance their ability to compete with and exclude other vaginal colonists. These include up to six toxin-antitoxin systems and up to nine additional antitoxins lacking cognate toxins, several of which are clustered within each genome. All strains encode bacteriocidal toxins, including two lysozyme-like toxins produced uniquely by strain 409-05. Interestingly, the BV isolates encode numerous proteins not found in strain 409-05 that likely increase their pathogenic potential. These include enzymes enabling mucin degradation, a trait previously described to strongly correlate with BV, although commonly attributed to non-G. vaginalis species. CONCLUSIONS Collectively, our results indicate that all three strains are able to thrive in vaginal environments, and therein the BV isolates are capable of occupying a niche that is unique from 409-05. Each strain has significant virulence potential, although genomic and metabolic differences, such as the ability to degrade mucin, indicate that the detection of G. vaginalis in the vaginal tract provides only partial information on the physiological potential of the organism.
Collapse
Affiliation(s)
- Carl J. Yeoman
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Suleyman Yildirim
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Susan M. Thomas
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
| | - A. Scott Durkin
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Manolito Torralba
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Granger Sutton
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Christian J. Buhay
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Yan Ding
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shannon P. Dugan-Rocha
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Donna M. Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xiang Qin
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Steven R. Leigh
- Department of Anthropology, University of Illinois, Urbana, Illinois, United States of America
| | - Rebecca Stumpf
- Department of Anthropology, University of Illinois, Urbana, Illinois, United States of America
| | - Bryan A. White
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Sarah K. Highlander
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Karen E. Nelson
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Brenda A. Wilson
- Institute for Genomic Biology, University of Illinois, Urbana, Illinois, United States of America
- Department of Microbiology, University of Illinois, Urbana, Illinois, United States of America
| |
Collapse
|
11
|
Myers GSA, Parker D, Al-Hasani K, Kennan RM, Seemann T, Ren Q, Badger JH, Selengut JD, Deboy RT, Tettelin H, Boyce JD, McCarl VP, Han X, Nelson WC, Madupu R, Mohamoud Y, Holley T, Fedorova N, Khouri H, Bottomley SP, Whittington RJ, Adler B, Songer JG, Rood JI, Paulsen IT. Genome sequence and identification of candidate vaccine antigens from the animal pathogen Dichelobacter nodosus. Nat Biotechnol 2007; 25:569-75. [PMID: 17468768 DOI: 10.1038/nbt1302] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Accepted: 04/04/2007] [Indexed: 11/09/2022]
Abstract
Dichelobacter nodosus causes ovine footrot, a disease that leads to severe economic losses in the wool and meat industries. We sequenced its 1.4-Mb genome, the smallest known genome of an anaerobe. It differs markedly from small genomes of intracellular bacteria, retaining greater biosynthetic capabilities and lacking any evidence of extensive ongoing genome reduction. Comparative genomic microarray studies and bioinformatic analysis suggested that, despite its small size, almost 20% of the genome is derived from lateral gene transfer. Most of these regions seem to be associated with virulence. Metabolic reconstruction indicated unsuspected capabilities, including carbohydrate utilization, electron transfer and several aerobic pathways. Global transcriptional profiling and bioinformatic analysis enabled the prediction of virulence factors and cell surface proteins. Screening of these proteins against ovine antisera identified eight immunogenic proteins that are candidate antigens for a cross-protective vaccine.
Collapse
Affiliation(s)
- Garry S A Myers
- The Institute for Genomic Research, 9712 Medical Center Drive, Rockville, Maryland 20850, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Wang L, Lin M. Identification of IspC, an 86-kilodalton protein target of humoral immune response to infection with Listeria monocytogenes serotype 4b, as a novel surface autolysin. J Bacteriol 2006; 189:2046-54. [PMID: 17172332 PMCID: PMC1855743 DOI: 10.1128/jb.01375-06] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We identified and biochemically characterized a novel surface-localized autolysin from Listeria monocytogenes serotype 4b, an 86-kDa protein consisting of 774 amino acids and known from our previous studies as the target (designated IspC) of the humoral immune response to listerial infection. Recombinant IspC, expressed in Escherichia coli, was purified and used to raise specific rabbit polyclonal antibodies for protein characterization. The native IspC was detected in all growth phases at a relatively stable low level during a 22-h in vitro culture, although its gene was transiently transcribed only in the early exponential growth phase. This and our previous findings suggest that IspC is upregulated in vivo during infection. The protein was unevenly distributed in clusters on the cell surface, as shown by immunofluorescence and immunogold electron microscopy. The recombinant IspC was capable of hydrolyzing not only the cell walls of the gram-positive bacterium Micrococcus lysodeikticus and the gram-negative bacterium E. coli but also that of the IspC-producing strain of L. monocytogenes serotype 4b, indicating that it was an autolysin. The IspC autolysin exhibited peptidoglycan hydrolase activity over a broad pH range of between 3 and 9, with a pH optimum of 7.5 to 9. Analysis of various truncated forms of IspC for cell wall-hydrolyzing or -binding activity has defined two separate functional domains: the N-terminal catalytic domain (amino acids [aa] 1 to 197) responsible for the hydrolytic activity and the C-terminal domain (aa 198 to 774) made up of seven GW modules responsible for anchoring the protein to the cell wall. In contrast to the full-length IspC, the N-terminal catalytic domain showed hydrolytic activity at acidic pHs, with a pH optimum of between 4 and 6 and negligible activity at alkaline pHs. This suggests that the cell wall binding domain may be of importance in modulating the activity of the N-terminal hydrolase domain. Elucidation of the biochemical properties of IspC may have provided new insights into its biological function(s) and its role in pathogenesis.
Collapse
Affiliation(s)
- Linru Wang
- Canadian Food Inspection Agency, Animal Diseases Research Institute, Ottawa, Ontario, Canada K2H 8P9
| | | |
Collapse
|
13
|
M.D. EA, . KDM, . AZ. Listeriosis in a Premature Neonate with Meconium Staining. JOURNAL OF MEDICAL SCIENCES 2006. [DOI: 10.3923/jms.2006.505.506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
14
|
Krawczyk-Balska A, Markiewicz Z, Bielecki J. Listeria monocytogenes protein p60 affects hemolytic activity and uptake of bacteria by macrophages. Folia Microbiol (Praha) 2005; 50:515-8. [PMID: 16681150 DOI: 10.1007/bf02931440] [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: 10/21/2022]
Abstract
Bacillus subtilis strains expressing listeriolysin O (LLO) and simultaneously LLO and p60 protein were constructed. The effect of p60 protein on hemolytic activity and on the invasion of professional phagocytes was demonstrated in the absence of other virulence factors of L. monocytogenes. The hemolytic activity of LLO in the presence of p60 protein decreased which indicates that p60 promoted adhesion and subsequent invasion of professional phagocytes.
Collapse
Affiliation(s)
- A Krawczyk-Balska
- Department of General Microbiology, Faculty of Biology, University of Warsaw, Poland.
| | | | | |
Collapse
|
15
|
Schmid MW, Ng EYW, Lampidis R, Emmerth M, Walcher M, Kreft J, Goebel W, Wagner M, Schleifer KH. Evolutionary history of the genus Listeria and its virulence genes. Syst Appl Microbiol 2005; 28:1-18. [PMID: 15709360 DOI: 10.1016/j.syapm.2004.09.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The genus Listeria contains the two pathogenic species Listeria monocytogenes and Listeria ivanovii and the four apparently apathogenic species Listeria innocua, Listeria seeligeri, Listeria welshimeri, and Listeria grayi. Pathogenicity of the former two species is enabled by an approximately 9 kb virulence gene cluster which is also present in a modified form in L. seeligeri. For all Listeria species, the sequence of the virulence gene cluster locus and its flanking regions was either determined in this study or assembled from public databases. Furthermore, some virulence-associated internalin loci were compared among the six species. Phylogenetic analyses were performed on a data set containing the sequences of prs, ldh, vclA, and vclB (all directly flanking the virulence gene cluster), as well as the iap gene and the 16S and 23S-rRNA coding genes which are located at different sites in the listerial chromosomes. L. grayi represents the deepest branch within the genus. The remaining five species form two groupings which have a high bootstrap support and which are consistently found by using different treeing methods. One lineage represents L. monocytogenes and L. innocua, while the other contains L. welshimeri, L. ivanovii and L. seeligeri, with L. welshimeri forming the deepest branch. Based on this perception, we tried to reconstruct the evolution of the virulence gene cluster. Since no traces of lateral gene transfer events could be detected the most parsimonious scenario is that the virulence gene cluster was present in the common ancestor of L. monocytogenes, L. innocua, L. ivanovii, L. seeligeri and L. welshimeri and that the pathogenic capability has been lost in two separate events represented by L. innocua and L. welshimeri. This hypothesis is also supported by the location of the putative deletion breakpoints of the virulence gene cluster within L. innocua and L. welshimeri.
Collapse
MESH Headings
- Bacterial Proteins/genetics
- DNA, Bacterial/chemistry
- DNA, Ribosomal/chemistry
- DNA, Ribosomal/genetics
- Evolution, Molecular
- Gene Deletion
- Genes, Bacterial/genetics
- Genes, rRNA
- Lipoproteins/genetics
- Listeria/genetics
- Listeria/pathogenicity
- Molecular Sequence Data
- Multigene Family
- Phylogeny
- RNA, Ribosomal, 16S/genetics
- RNA, Ribosomal, 23S/genetics
- Sequence Analysis, DNA
- Virulence/genetics
- Virulence Factors/genetics
Collapse
Affiliation(s)
- Michael W Schmid
- Lehrstuhl für Mikrobiologie, Technische Universität München, Am Hochanger 4, D-85350 Freising, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Teng F, Kawalec M, Weinstock GM, Hryniewicz W, Murray BE. An Enterococcus faecium secreted antigen, SagA, exhibits broad-spectrum binding to extracellular matrix proteins and appears essential for E. faecium growth. Infect Immun 2003; 71:5033-41. [PMID: 12933846 PMCID: PMC187350 DOI: 10.1128/iai.71.9.5033-5041.2003] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2003] [Revised: 05/28/2003] [Accepted: 06/17/2003] [Indexed: 11/20/2022] Open
Abstract
A gene encoding a major secreted antigen, SagA, was identified in Enterococcus faecium by screening an E. faecium genomic expression library with sera from patients with E. faecium-associated endocarditis. Recombinant SagA protein showed broad-spectrum binding to extracellular matrix (ECM) proteins, including fibrinogen, collagen type I, collagen type IV, fibronectin, and laminin. A fibrinogen-binding protein, purified from culture supernatants of an E. faecium clinical isolate, was found to match the N-terminal sequence of the predicted SagA protein and to react with the anti-SagA antibody, confirming that it was the SagA protein; this protein appeared as an 80- to 90-kDa smear on a Western blot that was sensitive to proteinase K and resistant to periodate treatment and glycoprotein staining. When overexpressed in E. faecium and Escherichia coli, the native and recombinant SagA proteins formed stable oligomers, apparently via their C-terminal domains. The SagA protein is composed of three domains: (i) a putative coiled-coil N-terminal domain that shows homology to the N-terminal domain of Streptococcus mutans SagA protein (42% similarity), previously shown to be involved in cell wall integrity and cell shape maintenance, and to the P45 protein of Listeria monocytogenes (41% similarity); (ii) a central domain containing direct repeats; and (iii) a C-terminal domain that is similar to that found in various proteins, including P45 (50% similarity) and P60 (52% similarity) of L. monocytogenes. The P45 and P60 proteins both have cell wall hydrolase activity, and the latter has also been shown to be involved in virulence, whereas cell wall hydrolase activity was not detected for SagA protein. The E. faecium sagA gene, like the S. mutans homologue, is located in a cluster of genes encoding proteins that appear to be involved in cell wall metabolism and could not be disrupted unless it was first transcomplemented, suggesting that the sagA gene is essential for E. faecium growth and may be involved in cell wall metabolism. In conclusion, the extracelluar E. faecium SagA protein is apparently essential for growth, shows broad-spectrum binding to ECM proteins, forms oligomers, and is antigenic during infection.
Collapse
Affiliation(s)
- Fang Teng
- Division of Infectious Disease, Department of Internal Medicine and Center for the Study of Emerging and Reemerging Pathogens, University of Texas Houston Medical School, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
17
|
Hahn HP, von Specht BU. Secretory delivery of recombinant proteins in attenuated Salmonella strains: potential and limitations of Type I protein transporters. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2003; 37:87-98. [PMID: 12832111 DOI: 10.1016/s0928-8244(03)00092-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Live attenuated Salmonella strains have been extensively explored as oral delivery systems for recombinant vaccine antigens and effector proteins with immunoadjuvant and immunomodulatory potential. The feasibility of this approach was demonstrated in human vaccination trials for various antigens. However, immunization efficiencies with live vaccines are generally significantly lower compared to those monitored in parenteral immunizations with the same vaccine antigen. This is, at least partly, due to the lack of secretory expression systems, enabling large-scale extracellular delivery of vaccine and effector proteins by these strains. Because of their low complexity and the terminal location of the secretion signal in the secreted protein, Type I (ATP-binding cassette) secretion systems appear to be particularly suited for development of such recombinant extracellular expression systems. So far, the Escherichia coli hemolysin system is the only Type I secretion system, which has been adapted to recombinant protein secretion in Salmonella. However, this system has a number of disadvantages, including low secretion capacity, complex genetic regulation, and structural restriction to the secreted protein, which eventually hinder high-level in vivo delivery of recombinant vaccines and effector proteins. Thus, the development of more efficient recombinant protein secretion systems, based on Type I exporters can help to improve efficacies of live recombinant Salmonella vaccines. Type I secretion systems, mediating secretion of bacterial surface layer proteins, such as RsaA in Caulobacter crescentus, are discussed as promising candidates for improved secretory delivery systems.
Collapse
Affiliation(s)
- Heinz P Hahn
- Chirurgische Universitätsklinik, Chirurgische Forschung, i. Br., Freiburg, Germany.
| | | |
Collapse
|
18
|
Pilgrim S, Kolb-Mäurer A, Gentschev I, Goebel W, Kuhn M. Deletion of the gene encoding p60 in Listeria monocytogenes leads to abnormal cell division and loss of actin-based motility. Infect Immun 2003; 71:3473-84. [PMID: 12761132 PMCID: PMC155713 DOI: 10.1128/iai.71.6.3473-3484.2003] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2002] [Revised: 09/24/2002] [Accepted: 02/24/2003] [Indexed: 11/20/2022] Open
Abstract
Protein p60 encoded by the iap gene is regarded as an essential gene product of Listeria monocytogenes. Here we report, however, the successful construction of a viable iap deletion mutant of L. monocytogenes EGD. The mutant, which produces no p60, shows abnormal septum formation and tends to form short filaments and hooked forms during logarithmic growth. These abnormal bacterial cells break into almost normal sized single bacteria in the late-stationary-growth phase. The iap mutant is strongly attenuated in a mouse model after intravenous injection, demonstrating the importance of p60 during infection, and the invasiveness of the Deltaiap mutant for 3T6 fibroblasts and Caco-2 epithelial cells is slightly reduced. Upon uptake by epithelial cells and macrophages, the iap mutant escapes from the phagosome into the cytosol with the same efficiency as the wild-type strain, and the mutant bacteria also grow intracellularly at a rate similar to that of the wild-type strain. Intracellular movement and cell-to-cell spread are drastically reduced in various cell lines, since the iap-negative bacteria fail to induce the formation of actin tails. However, the bacteria are covered with actin filaments. Most intracellular bacteria show a nonpolar and uneven distribution of ActA around the cell, in contrast to that for the wild-type strain, where ActA is concentrated at the old pole. In an iap(+) revertant strain that produces wild-type levels of p60, intracellular movement, cell-to-cell spread, and polar distribution of ActA are fully restored. In vitro analysis of ActA distribution on the filaments of the Deltaiap strain shows that the loss of bacterial septum formation leads to ActA accumulation at the presumed division sites. In the light of data presented here and elswhere, we propose to rename iap (invasion-associated protein) cwhA (cell wall hydrolase A).
Collapse
Affiliation(s)
- Sabine Pilgrim
- Lehrstuhl für Mikrobiologie der Universität Würzburg, Theodor-Boveri-Institut für Biowissenschaften, Am Hubland, Germany
| | | | | | | | | |
Collapse
|
19
|
Dietrich G, Viret JF, Gentschev I. Haemolysin A and listeriolysin--two vaccine delivery tools for the induction of cell-mediated immunity. Int J Parasitol 2003; 33:495-505. [PMID: 12782050 DOI: 10.1016/s0020-7519(03)00058-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Haemolysin A of Escherichia coli and listeriolysin of Listeria monocytogenes represent important bacterial virulence factors. While such cytolysins are usually the reason for morbidity and even mortality, vaccine researchers have turned haemolysin A and listeriolysin into tools for vaccine delivery. Both cytolysins have found widespread application in vaccine research and are highly suitable for the elicitation of cell-mediated immunity. In this paper, we will review vaccine delivery mediated by the haemolysin A secretion system and listeriolysin and will highlight their use in vaccination approaches against protozoan parasites.
Collapse
Affiliation(s)
- Guido Dietrich
- Vaccine Research, Berna Biotech AG, Rehhagstr. 79, CH-3018, Bern, Switzerland.
| | | | | |
Collapse
|
20
|
Lenz LL, Portnoy DA. Identification of a second Listeria secA gene associated with protein secretion and the rough phenotype. Mol Microbiol 2002; 45:1043-56. [PMID: 12180923 DOI: 10.1046/j.1365-2958.2002.03072.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We describe the identification and characterization of a second secA gene in Listeria monocytogenes. This gene, termed secA2, is involved in smooth-rough phenotypic variation and secA2 expression contributes to bacterial virulence. Spontaneous rough (R-) variants of L. monocytogenes grow in chains and form rough colonies on solid media. A subset of R-variants, classified here as type I, also shows reduced secretion of an autolysin, p60. We find that disruptions and in frame deletions in secA2 confer phenotypes identical to those of spontaneous type I R-variants. Additionally, the secA2 genes from two spontaneous type I R-variants encoded truncated SecA2 proteins. Mutations were not found in the secA2 genes from the remaining five independent R-variants, four of which showed a distinct (type II) rough morphology and secreted wild-type levels of p60. Expression of an epitope-tagged SecA2 in the DeltasecA2 strain and a spontaneous R-variant restored normal cell septation and smooth colony morphology. These data suggest that mutations in both secA2 and other genes contribute to smooth-rough phase variation in L. monocytogenes. Expression of the full-length SecA2 also promotes secretion of p60 and a set of additional L. monocytogenes proteins. We hypothesize that SecA2-dependent protein secretion plays a role in the colonization of environmental and host surfaces.
Collapse
Affiliation(s)
- Laurel L Lenz
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
| | | |
Collapse
|
21
|
Herler M, Bubert A, Goetz M, Vega Y, Vazquez-Boland JA, Goebel W. Positive selection of mutations leading to loss or reduction of transcriptional activity of PrfA, the central regulator of Listeria monocytogenes virulence. J Bacteriol 2001; 183:5562-70. [PMID: 11544218 PMCID: PMC95447 DOI: 10.1128/jb.183.19.5562-5570.2001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2001] [Accepted: 07/09/2001] [Indexed: 11/20/2022] Open
Abstract
Transcription factor PrfA controls the expression of virulence genes essential for Listeria monocytogenes pathogenesis. To gain insight into the structure-function relationship of PrfA, we devised a positive-selection system to isolate mutations reducing or abolishing transcriptional activity. The system is based on the observation that the listerial iap gene, encoding the p60 protein, is lethal if overexpressed in Bacillus subtilis. A plasmid in which the iap gene is placed under the control of the PrfA-dependent hly promoter was constructed and introduced into B. subtilis. This strain was rapidly killed when expression of iap was induced by introduction of a second plasmid carrying prfA. Two classes of B. subtilis survivor mutants were identified: one carried mutations in iap, and the second carried mutations in prfA. Sequence analysis of the defective prfA genes identified mutations in three regions of the PrfA protein: region A, between amino acids 58 and 67 in the beta-roll domain of PrfA; region B, between amino acids 169 and 193, which corresponds to the DNA-binding helix-turn-helix motif; and region C, comprising the 38 C-terminal amino acids of PrfA, which form a leucine zipper-like structure. PrfA proteins with mutations in regions B and C were unable to bind to the PrfA-binding site in the target DNA, while mutations in region A resulted in a protein still binding the target DNA but unable to form a stable complex with RNA polymerase and initiate transcription in vitro.
Collapse
Affiliation(s)
- M Herler
- Biocenter of the University of Würzburg (Microbiology), Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Vázquez-Boland JA, Kuhn M, Berche P, Chakraborty T, Domínguez-Bernal G, Goebel W, González-Zorn B, Wehland J, Kreft J. Listeria pathogenesis and molecular virulence determinants. Clin Microbiol Rev 2001; 14:584-640. [PMID: 11432815 PMCID: PMC88991 DOI: 10.1128/cmr.14.3.584-640.2001] [Citation(s) in RCA: 1506] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The gram-positive bacterium Listeria monocytogenes is the causative agent of listeriosis, a highly fatal opportunistic foodborne infection. Pregnant women, neonates, the elderly, and debilitated or immunocompromised patients in general are predominantly affected, although the disease can also develop in normal individuals. Clinical manifestations of invasive listeriosis are usually severe and include abortion, sepsis, and meningoencephalitis. Listeriosis can also manifest as a febrile gastroenteritis syndrome. In addition to humans, L. monocytogenes affects many vertebrate species, including birds. Listeria ivanovii, a second pathogenic species of the genus, is specific for ruminants. Our current view of the pathophysiology of listeriosis derives largely from studies with the mouse infection model. Pathogenic listeriae enter the host primarily through the intestine. The liver is thought to be their first target organ after intestinal translocation. In the liver, listeriae actively multiply until the infection is controlled by a cell-mediated immune response. This initial, subclinical step of listeriosis is thought to be common due to the frequent presence of pathogenic L. monocytogenes in food. In normal individuals, the continual exposure to listerial antigens probably contributes to the maintenance of anti-Listeria memory T cells. However, in debilitated and immunocompromised patients, the unrestricted proliferation of listeriae in the liver may result in prolonged low-level bacteremia, leading to invasion of the preferred secondary target organs (the brain and the gravid uterus) and to overt clinical disease. L. monocytogenes and L. ivanovii are facultative intracellular parasites able to survive in macrophages and to invade a variety of normally nonphagocytic cells, such as epithelial cells, hepatocytes, and endothelial cells. In all these cell types, pathogenic listeriae go through an intracellular life cycle involving early escape from the phagocytic vacuole, rapid intracytoplasmic multiplication, bacterially induced actin-based motility, and direct spread to neighboring cells, in which they reinitiate the cycle. In this way, listeriae disseminate in host tissues sheltered from the humoral arm of the immune system. Over the last 15 years, a number of virulence factors involved in key steps of this intracellular life cycle have been identified. This review describes in detail the molecular determinants of Listeria virulence and their mechanism of action and summarizes the current knowledge on the pathophysiology of listeriosis and the cell biology and host cell responses to Listeria infection. This article provides an updated perspective of the development of our understanding of Listeria pathogenesis from the first molecular genetic analyses of virulence mechanisms reported in 1985 until the start of the genomic era of Listeria research.
Collapse
Affiliation(s)
- J A Vázquez-Boland
- Grupo de Patogénesis Molecular Bacteriana, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hess J, Schaible U, Raupach B, Kaufmann SH. Exploiting the immune system: toward new vaccines against intracellular bacteria. Adv Immunol 2001; 75:1-88. [PMID: 10879281 DOI: 10.1016/s0065-2776(00)75001-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- J Hess
- Department of Immunology, Max-Planck-Institute for Infection Biology, Berlin, Germany
| | | | | | | |
Collapse
|
24
|
Mariani F, Cappelli G, Riccardi G, Colizzi V. Mycobacterium tuberculosis H37Rv comparative gene-expression analysis in synthetic medium and human macrophage. Gene 2000; 253:281-91. [PMID: 10940566 DOI: 10.1016/s0378-1119(00)00249-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Mycobacteria are intracellular pathogens that survive and grow in host macrophages. Following phagocytosis, sustained intracellular bacterial growth depends on its ability to avoid destruction by macrophage-mediated host defences such as lysosomal enzymes, reactive oxygen and the reactive nitrogen intermediates. This suggests that the interaction between host cell and microbe is delicately balanced, and can be tipped in favour of either organism. The identification of Mycobacterium tuberculosis H37Rv (MTB) genes expressed within host cells would contribute greatly to the development of new strategies to fight tuberculosis. In the present study, we compared MTB gene expression in the course of intra- (human macrophages) and extracellular growth (Sauton's medium) to ascertain whether differences might occur between gene-expression patterns in the two habitats of replication. Using reverse-transcriptase polymerase chain reaction (RT-PCR) on a group of 14 MTB-Complex-specific genes, we found that MT10Sa (a small stable RNA), 35 kDa (unknown), ahpC (alkyl hydroperoxide reductase, AhpC), sigF (alternative RNA Polymerase sigma factor), and katG (catalase-peroxidase, HPI) genes are expressed in both the environments, while Ag85B, Ag85C (members of the Antigen 85 Complex), rpoV (RNA Polymerase sigma factor) and ESAT6 (early secretory antigen, 6 kDa) are expressed only in the in vitro culture; on the other hand, Ag85A (Antigen 85 Complex), rpoB (RNA Polymerase beta sub-unit), pab (Protein antigen b), invA and invB genes (encoding proteins that show homologies with p60 of Listeria monocytogenes) are expressed only inside the macrophage. Positive RT-PCR products on cDNAs for these genomic regions were not obtained from approximately 1000-fold more bacteria grown in Laboratory Broth. Identification of M. tuberculosis genes expressed in response to phagocytosis by human macrophages increases our basic understanding of the host-pathogen interaction, and helps to identify bacterial factors necessary for in vivo survival and growth.
Collapse
Affiliation(s)
- F Mariani
- Experimental Medicine Institute-Rome/CNR/Tor Vergata Research Area, Via del Fosso del Cavaliere, 100, 00133, Rome, Italy.
| | | | | | | |
Collapse
|
25
|
Braunstein M, Griffin TJ IV, Kriakov JI, Friedman ST, Grindley ND, Jacobs WR. Identification of genes encoding exported Mycobacterium tuberculosis proteins using a Tn552'phoA in vitro transposition system. J Bacteriol 2000; 182:2732-40. [PMID: 10781540 PMCID: PMC101980 DOI: 10.1128/jb.182.10.2732-2740.2000] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Secreted and cell envelope-associated proteins are important to both Mycobacterium tuberculosis pathogenesis and the generation of protective immunity to M. tuberculosis. We used an in vitro Tn552'phoA transposition system to identify exported proteins of M. tuberculosis. The system is simple and efficient, and the transposon inserts randomly into target DNA. M. tuberculosis genomic libraries were targeted with Tn552'phoA transposons, and these libraries were screened in M. smegmatis for active PhoA translational fusions. Thirty-two different M. tuberculosis open reading frames were identified; eight contain standard signal peptides, six contain lipoprotein signal peptides, and seventeen contain one or more transmembrane domains. Four of these proteins had not yet been assigned as exported proteins in the M. tuberculosis databases. This collection of exported proteins includes factors that are known to participate in the immune response of M. tuberculosis and proteins with homologies, suggesting a role in pathogenesis. Nine of the proteins appear to be unique to mycobacteria and represent promising candidates for factors that participate in protective immunity and virulence. This technology of creating comprehensive fusion libraries should be applicable to other organisms.
Collapse
Affiliation(s)
- M Braunstein
- Howard Hughes Medical Institute, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | |
Collapse
|
26
|
Park JH, Lee YS, Lim YK, Kwon SH, Lee CU, Yoon BS. Specific binding of recombinant Listeria monocytogenes p60 protein to Caco-2 cells. FEMS Microbiol Lett 2000; 186:35-40. [PMID: 10779709 DOI: 10.1111/j.1574-6968.2000.tb09078.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The Listeria monocytogenes p60 is a major extracellular protein, which is believed to be involved in the invasion of these bacteria into their host cells. So far the mechanism by which p60 participates in the internalization or penetration of L. monocytogenes is still veiled. To determine the possibility of a direct interaction of p60 with the host cell surface, the iap gene was recombinantly expressed in Escherichia coli and used for binding studies with the enterocyte-like Caco-2 cells. Fluorescence activated flow cytometry and confocal laser scanning microscopy revealed a cell membrane specific staining with p60, which implications in Listeria virulence are discussed.
Collapse
Affiliation(s)
- J H Park
- Peptide Engineering Research Unit, Korea Research Institute of Bioscience and Biotechnology, Yusong, P.O. Box 115, Taejon, South Korea
| | | | | | | | | | | |
Collapse
|
27
|
Kozarov E, Miyashita N, Burks J, Cerveny K, Brown TA, McArthur WP, Progulske-Fox A. Expression and immunogenicity of hemagglutinin A from Porphyromonas gingivalis in an avirulent Salmonella enterica serovar typhimurium vaccine strain. Infect Immun 2000; 68:732-9. [PMID: 10639440 PMCID: PMC97199 DOI: 10.1128/iai.68.2.732-739.2000] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gingivalis is a major etiologic agent of periodontitis, a chronic inflammatory disease that ultimately results in the loss of the supporting tissues of the teeth. Previous work has demonstrated the usefulness of avirulent Salmonella enterica serovar Typhimurium strains as antigen delivery systems for protective antigens of pathogens that colonize or cross mucosal surfaces. In this study, we constructed and characterized a recombinant S. enterica serovar Typhimurium avirulent vaccine strain which expresses hemagglutinin A and carries no antibiotic resistance markers. HagA, a major virulence-associated surface protein, is a potentially useful immunogen that contains an antigenic epitope which, in humans, elicits an immune response that is protective against subsequent colonization by P. gingivalis. The hagA gene, including its promoter, was cloned into a balanced-lethal Salmonella vector and transferred to the vaccine strain. Heterologous expression of HagA was demonstrated in both Escherichia coli JM109 and S. enterica serovar Typhimurium vaccine strain chi4072. The HagA epitope was present in its native configuration as determined by immunochemistry and immunoelectron microscopy. Purified recombinant HagA was recognized by sera from mice immunized with the S. enterica serovar Typhimurium vaccine strain. The HagA-specific antigen of the vaccine was also found to be recognized by serum from a periodontal patient. This vaccine strain, which expresses the functional hemagglutinin protein, induces a humoral immune response against HagA and may be useful for developing a protective vaccine against periodontal diseases associated with P. gingivalis.
Collapse
Affiliation(s)
- E Kozarov
- Department of Oral Biology and the Periodontal Disease Research Center, University of Florida, Gainesville, Florida 32610, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Appelberg R, Leal IS. Mutants of Listeria monocytogenes defective in In vitro invasion and cell-to-cell spreading still invade and proliferate in hepatocytes of neutropenic mice. Infect Immun 2000; 68:912-4. [PMID: 10639462 PMCID: PMC97221 DOI: 10.1128/iai.68.2.912-914.2000] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Listeria monocytogenes mutants defective in the actA gene, the plcB gene, and the inlA and inlB genes were less virulent when injected intravenously into BALB/c mice. The growth of these strains as well as of the virulent wild-type strains was increased by treating mice with a neutrophil-specific depleting monoclonal antibody, RB6-8C5. Histologic examination of the livers of the treated animals showed intrahepatocytic proliferation of the listeriae in all cases. Our data show that more than one pathway exists that allows L. monocytogenes to invade parenchymal cells. One pathway most likely involves the actA and plcB gene products, and a second one probably involves the internalins.
Collapse
Affiliation(s)
- R Appelberg
- Laboratory of Microbiology and Immunology of Infection, Institute for Molecular and Cell Biology, University of Porto, Porto, Portugal.
| | | |
Collapse
|
29
|
Abstract
Although empirical vaccine development was highly successful, it has now reached its limits. Vaccines are only efficacious against those pathogens which are primarily controlled by antibodies. Protection against many infectious agents, however, strongly depends on T lymphocytes. Thus, novel vaccines have to stimulate the combination of T lymphocytes that is required for an optimum protective immune response. Although identification of antigens remains crucial, novel vaccine design also needs to consider the best way of introducing these antigens to the immune system. Intracellular antigen compartmentalisation, the early cytokine milieu and the appropriate surface expression of co-stimulatory molecules are of major relevance for understanding how novel vaccines could induce a protective immune response mediated by T lymphocytes. Intracellular bacteria are controlled by T lymphocytes and efficacious vaccines against these pathogens are not available yet. In this treatise, two experimental vaccination strategies will be described in more detail. These encompass recombinant vaccine carriers expressing, and naked DNA constructs encoding, heterologous antigens. Both vaccination strategies proved to be protective in the model of experimental listeriosis of mice.
Collapse
Affiliation(s)
- S H Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany.
| | | | | |
Collapse
|
30
|
Abstract
Listeria monocytogenes is a gram-positive, nonsporulating, food-borne pathogen of humans and animals that is able to invade many eukaryotic cells. Several listerial surface components have been reported to interact with eukaryotic cell receptors, but the complete mechanism by which the bacteria interact with all of these cell types remains largely unknown. In this work, we found that L. monocytogenes binds to human fibronectin, a 450,000-Da dimeric glycoprotein found in body fluids, on the surface of cells and in an insoluble component of the extracellular matrix. The binding of fibronectin to L. monocytogenes was found to be saturable and dependent on proteinaceous receptors. Five fibronectin-binding proteins of 55.3, 48.6, 46.7, 42.4, and 26.8 kDa were identified. The 55.3-kDa protein was proved to be present at the bacterial cell surface. The binding of L. monocytogenes to fibronectin adds to the number of molecules to which the bacterium is able to adhere and emphasizes the complexity of host-pathogen interactions.
Collapse
Affiliation(s)
- P Gilot
- Department of Virology, Pasteur Institute, Brussels, Belgium.
| | | | | |
Collapse
|
31
|
Orr N, Galen JE, Levine MM. Expression and immunogenicity of a mutant diphtheria toxin molecule, CRM(197), and its fragments in Salmonella typhi vaccine strain CVD 908-htrA. Infect Immun 1999; 67:4290-4. [PMID: 10417208 PMCID: PMC96741 DOI: 10.1128/iai.67.8.4290-4294.1999] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mutant diphtheria toxin molecule CRM(197) and fragments thereof were expressed in attenuated Salmonella typhi CVD 908-htrA, and the constructs were tested for their ability to induce serum antitoxin. Initially, expressed proteins were insoluble, and the constructs failed to induce neutralizing antitoxin. Soluble CRM(197) was expressed at low levels by utilizing the hemolysin A secretion system from Escherichia coli.
Collapse
Affiliation(s)
- N Orr
- Department of Pediatrics, Division of Infectious Diseases and Tropical Pediatrics, Center for Vaccine Development, Department of Medicine, Division of Geographic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
32
|
Geginat G, Nichterlein T, Kretschmar M, Schenk S, Hof H, Lalic-Mülthaler M, Goebel W, Bubert A. Enhancement of the Listeria monocytogenes p60-Specific CD4 and CD8 T Cell Memory by Nonpathogenic Listeria innocua. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.8.4781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The contact of T cells to cross-reactive antigenic determinants expressed by nonpathogenic environmental micro-organisms may contribute to the induction or maintenance of T cell memory. This hypothesis was evaluated in the model of murine Listeria monocytogenes infection. The influence of nonpathogenic L. innocua on the L. monocytogenes p60-specific T cell response was analyzed. We show that some CD4 T cell clones raised against purified p60 from L. monocytogenes cross-react with p60 purified from L. innocua. The L. monocytogenes p60-specific CD4 T cell clone 1A recognized the corresponding L. innocua p60 peptide QAAKPAPAPSTN, which differs only in the first amino acid residue. In vitro experiments revealed that after L. monocytogenes infection of APCs, MHC class I-restricted presentation of p60 occurs, while MHC class II-restricted p60 presentation is inhibited. L. innocua-infected cells presented p60 more weakly but equally well in the context of both MHC class I and MHC class II. In contrast to these in vitro experiments the infection of mice with L. monocytogenes induced a strong p60-specific CD4 and CD8 T cell response, while L. innocua infection failed to induce p60-specific T cells. L. innocua booster infection, however, expanded p60-specific memory T cells induced by previous L. monocytogenes infection. In conclusion, these findings suggest that infection with a frequently occurring environmental bacterium such as L. innocua, which is nonpathogenic and not adapted to intracellular replication, can contribute to the maintenance of memory T cells specific for a related intracellular pathogen.
Collapse
Affiliation(s)
- Gernot Geginat
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and
| | - Thomas Nichterlein
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and
| | - Marianne Kretschmar
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and
| | - Simone Schenk
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and
| | - Herbert Hof
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and
| | - Mio Lalic-Mülthaler
- †Lehrstuhl für Mikrobiologie, Theodor-Boveri-Institut für Biowissenschaften, Am Hubland, Würzburg, Germany
| | - Werner Goebel
- †Lehrstuhl für Mikrobiologie, Theodor-Boveri-Institut für Biowissenschaften, Am Hubland, Würzburg, Germany
| | - Andreas Bubert
- †Lehrstuhl für Mikrobiologie, Theodor-Boveri-Institut für Biowissenschaften, Am Hubland, Würzburg, Germany
| |
Collapse
|
33
|
Affiliation(s)
- J C Whisstock
- Dept of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Melbourne, Victoria 3168, Australia
| | | |
Collapse
|
34
|
Garduño RA, Garduño E, Hoffman PS. Surface-associated hsp60 chaperonin of Legionella pneumophila mediates invasion in a HeLa cell model. Infect Immun 1998; 66:4602-10. [PMID: 9746556 PMCID: PMC108567 DOI: 10.1128/iai.66.10.4602-4610.1998] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HeLa cells have been previously used to demonstrate that virulent strains of Legionella pneumophila (but not salt-tolerant avirulent strains) efficiently invade nonphagocytic cells. Hsp60, a member of the GroEL family of chaperonins, is displayed on the surface of virulent L. pneumophila (R. A. Garduño et al., J. Bacteriol. 180:505-513, 1988). Because Hsp60 is largely involved in protein-protein interactions, we investigated its role in adherence-invasion in the HeLa cell model. Hsp60-specific antibodies inhibited the adherence and invasiveness of two virulent L. pneumophila strains in a dose-dependent manner but had no effect on the association of their salt-tolerant avirulent derivatives with HeLa cells. A monospecific anti-OmpS (major outer membrane protein) serum inhibited the association of both virulent and avirulent strains of L. pneumophila to HeLa cells, suggesting that while both Hsp60 and OmpS may mediate bacterial association to HeLa cells, only virulent strains selectively displayed Hsp60 on their surfaces. Furthermore, the surface-associated Hsp60 of virulent bacterial cells was susceptible to the action of trypsin, which rendered the bacteria noninvasive. Additionally, pretreatment of HeLa cells with purified Hsp60 or precoating of the plastic surface where HeLa cells attached with Hsp60 reduced the adherence and invasiveness of the two virulent strains. Finally, recombinant Hsp60 covalently bound to latex beads promoted the early association of beads with HeLa cells by a factor of 20 over bovine serum albumin (BSA)-coated beads and competed with virulent strains for association with HeLa cells. Hsp60-coated beads were internalized in large numbers by HeLa cells and remained in tight endosomes that did not fuse with other vesicles, whereas internalized BSA-coated beads, for which endocytic trafficking is well established, resided in more loose or elongated endosomes. Mature intracellular forms of L. pneumophila, which were up to 100-fold more efficient than agar-grown bacteria at associating with HeLa cells, were enriched for Hsp60 on the bacterial surface, as determined by immunolocalization techniques. Collectively, these results establish a role for surface-exposed Hsp60 in invasion of HeLa cells by L. pneumophila.
Collapse
Affiliation(s)
- R A Garduño
- Department of Microbiology and Immunology and Department of Medicine, Division of Infectious Diseases, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4H7
| | | | | |
Collapse
|
35
|
Geginat G, Lalic M, Kretschmar M, Goebel W, Hof H, Palm D, Bubert A. Th1 Cells Specific for a Secreted Protein of Listeria monocytogenes Are Protective In Vivo. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.12.6046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
In the present study we have investigated the role of the secreted p60 protein from Listeria monocytogenes as an Ag for CD4 T cells. The p60 protein is an abundant extracellular protein that is highly conserved within the members of the genus Listeria. Our results show that L. monocytogenes infection induces a potent p60-specific Th1 immune response. Remarkably, we found that p60-specific Th1 clones mediate significant protection against L. monocytogenes infection. For one p60-specific clone, the peptide epitope was defined. This clone recognized p60 301-312 (EAAKPAPAPSTN) in the context of the H-2Ad molecule. Despite the fact that acquired immunity against L. monocytogenes is primarily mediated by cytotoxic CD8 T lymphocytes, our data clearly demonstrate that secreted bacterial proteins are important CD4 T cell Ags and that Th1 clones specific for a secreted bacterial protein can contribute to the protection against an intracellular pathogen such as L. monocytogenes.
Collapse
Affiliation(s)
- Gernot Geginat
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and Lehrstuhl für
| | | | - Marianne Kretschmar
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and Lehrstuhl für
| | | | - Herbert Hof
- *Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany; and Lehrstuhl für
| | - Dieter Palm
- ‡Physiologische Chemie, Theodor-Boveri-Institut für Biowissenschaften, Würzburg, Germany
| | | |
Collapse
|
36
|
Gentschev I, Glaser I, Goebel W, McKeever DJ, Musoke A, Heussler VT. Delivery of the p67 sporozoite antigen of Theileria parva by using recombinant Salmonella dublin: secretion of the product enhances specific antibody responses in cattle. Infect Immun 1998; 66:2060-4. [PMID: 9573089 PMCID: PMC108163 DOI: 10.1128/iai.66.5.2060-2064.1998] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The p67 sporozoite antigen of Theileria parva has been fused to the C-terminal secretion signal of Escherichia coli hemolysin and expressed in secreted form by attenuated Salmonella dublin aroA strain SL5631. The recombinant p67 antigen was detected in the supernatant of transformed bacterial cultures. Immunization trials in cattle revealed that SL5631 secreting the antigen provoked a 10-fold-higher antibody response to p67 than recombinant SL5631 expressing but not secreting p67. Immunized calves were challenged with a 80% lethal dose of T. parva sporozoites and monitored for the development of infection. Two of three calves immunized intramuscularly with the p67-secreting SL5631 strain were found to be protected, whereas only one of three animals immunized with the nonsecreting p67-expressing SL5631 strain was protected. This is the first demonstration that complete eukaryotic antigens fused to the C-terminal portion of E. coli hemolysin can be exported from attenuated Salmonella strains and that such exported antigens can protect cattle against subsequent parasite challenge.
Collapse
Affiliation(s)
- I Gentschev
- Lehrstuhl für Mikrobiologie, Theodor-Boveri-Institut für Biowissenschaften, Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
37
|
Kuhn M. The microtubule depolymerizing drugs nocodazole and colchicine inhibit the uptake of Listeria monocytogenes by P388D1 macrophages. FEMS Microbiol Lett 1998; 160:87-90. [PMID: 9495017 DOI: 10.1111/j.1574-6968.1998.tb12895.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Uptake of Listeria monocytogenes by different mammalian cells like macrophages and epithelial cells is dependent on functional actin filaments and hence susceptible to inhibition by cytochalasin. Here we show that phagocytic uptake of L. monocytogenes by P388D1 macrophages is also highly sensitive to treatment with the microtubule depolymerizing drugs nocodazole and colchicine. This sensitivity is cell type specific and much less pronounced in bone marrow-derived macrophages and Caco-2 epithelial cells. In contrast to nocodazole and colchicine, the microtubule stabilizing drug taxol has no significant effect on the uptake of L. monocytogenes by all three cell types tested.
Collapse
Affiliation(s)
- M Kuhn
- Theodor-Boveri-Institut für Biowissenschaften, Universität Würzburg, Lehrstuhl für Mikrobiologie, Germany.
| |
Collapse
|
38
|
Gregory SH, Sagnimeni AJ, Wing EJ. Internalin B promotes the replication of Listeria monocytogenes in mouse hepatocytes. Infect Immun 1997; 65:5137-41. [PMID: 9393807 PMCID: PMC175740 DOI: 10.1128/iai.65.12.5137-5141.1997] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The uptake of Listeria monocytogenes by a variety of cell types in vitro is facilitated by the protein products of the inlAB (internalin) operon expressed by the organism. In the case of mouse hepatocytes, the extent to which inlAB expression influenced the uptake of Listeria in vitro was markedly dependent upon the ratio of bacteria to cells. At a ratio of 100:1, greater than 40-fold fewer transposon-induced inl4B mutant listeriae entered hepatocytes compared to the isogenic wild-type control; the difference was only fourfold, however, in cultures inoculated at a 1:1 ratio. Similarly, the uptake of in-frame inlB or inlAB deletion mutants differed only fourfold from the uptake of wild-type or inlA mutant Listeria at a 1:1 multiplicity of infection. Mutations affecting inlB or inlAB, on the other hand, resulted in a marked decrease in the capacity of Listeria to proliferate within mouse hepatocytes in vivo and in vitro. Electron micrographs of Listeria-infected hepatocytes demonstrated the impaired capacity of inlB mutants to escape from endocytic vacuoles and to enter the cytoplasm where proliferation occurs. These findings indicate that the protein product of inlB exerts a significant effect on the intracellular replication of Listeria.
Collapse
Affiliation(s)
- S H Gregory
- Department of Medicine, University of Pittsburgh Medical Center, Pennsylvania, USA
| | | | | |
Collapse
|
39
|
Hess J, Dietrich G, Gentschev I, Miko D, Goebel W, Kaufmann SH. Protection against murine listeriosis by an attenuated recombinant Salmonella typhimurium vaccine strain that secretes the naturally somatic antigen superoxide dismutase. Infect Immun 1997; 65:1286-92. [PMID: 9119463 PMCID: PMC175129 DOI: 10.1128/iai.65.4.1286-1292.1997] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A recombinant (r)-Salmonella typhimurium aroA vaccine strain was constructed which secretes the naturally somatic protein of Listeria monocytogenes, superoxide dismutase (SOD), by the HlyB/HlyD/TolC export machinery. Vaccine efficacy of the SOD-bearing carrier strain was compared with that of the p60-secreting construct, S. typhimurium p60s (J. Hess, I. Gentschev, D. Miko, M. Welzel, C. Ladel, W. Goebel, and S. H. E. Kaufmann, Proc. Natl. Acad. Sci. USA 93:1458-1463, 1996). Vaccination of mice with both constructs induced protection against a lethal challenge with the intracellular pathogen, L. monocytogenes. While the somatic listerial antigen, SOD, is immunologically uncharacterized, the naturally secreted protein of L. monocytogenes, p60, is known to be highly immunogenic. Our data emphasize the high vaccine potential of r-Salmonella constructs secreting antigens of somatic or secreted origin. Moreover, they suggest that the HlyB/HlyD/TolC-based antigen delivery system with attenuated Salmonella spp. as the carrier is capable of potentiating the immune response against foreign proteins independent from their immunogenicity in and display by the natural host.
Collapse
Affiliation(s)
- J Hess
- Department of Immunology, University of Ulm, Germany.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Listeria monocytogenes is one of the leading foodborne pathogens and has been implicated in numerous outbreaks in the last 2 decades. Immunocompromised populations are usually the most susceptible to Listeria infections. Although the pathogenic mechanism is a complex process, significant progress has been made in unravelling the mechanism in recent years. It is now clear that numerous extracellular and cell-associated proteins, such as internalin, listeriolysin, actin polymerization protein, phospholipase, metalloprotease, and possibly p60 proteins, are essential for L. monocytogenes entry into mammalian cells, survival inside the phagosome, escape into the cytoplasm, and cell-to-cell spread. Other proteins may be responsible for growth and physiology or to maintain the structural integrity of the bacteria. Monoclonal and polyclonal antibodies have been developed against many of those antigens or their synthetic derivatives that have helped greatly to determine the structure and function of these antigens. The antibodies were also used for the diagnosis and detection, immunocytochemical staining, and serotyping of Listeria. Humoral immune response to live L. monocytogenes cells was examined in naturally or experimentally infected hosts. Studies revealed that only extracellular antigens induced the humoral response, whereas cell-associated antigens had apparently no response. It is speculated that during the occasional bacteremic phase, L. monocytogenes releases extracellular antigens that are then processed by the immune system for antibody production. As L. monocytogenes is an intracellular pathogen, the cell-associated antigens are not persistent in the blood circulation and thus fail to stimulate the humoral immune response.
Collapse
Affiliation(s)
- A K Bhunia
- Department of Food Science and Animal Industries, Alabama A&M University, Huntsville 35762, USA
| |
Collapse
|
41
|
Gentschev I, Mollenkopf H, Sokolovic Z, Hess J, Kaufmann SH, Goebel W. Development of antigen-delivery systems, based on the Escherichia coli hemolysin secretion pathway. Gene 1996; 179:133-40. [PMID: 8955639 DOI: 10.1016/s0378-1119(96)00424-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We describe the development of plasmid vectors carrying the expression sites, an hlyA cassette and the secretion genes of Escherichia coli hemolysin. These allow the synthesis and secretion of heterologous microbial antigens in E. coli and attenuated Salmonella aroA strains. Genes or gene fragments encoding microbial antigens are inserted in-frame into a residual part of the hlyA gene which essentially encodes the HlyA secretion signal (HlyAs). In general, the fused genes, carrying the hlyAs sequence at the 3' terminus, are efficiently expressed, and the synthesized antigens are secreted into the culture supernatant of the producing strain. Attenuated Salmonella strains synthesizing either HlyAs-fused listeriolysin or p60 of Listeria monocytogenes were constructed by this procedure and shown to provide protective immunity against L. monocytogenes in mice. The most effective protection was obtained when these microbial antigens were secreted by the attenuated Salmonella strains. We further present new approaches which may allow the application of this antigen-delivery system to any microbial antigen.
Collapse
Affiliation(s)
- I Gentschev
- Lehrstuhl für Mikrobiologie, Theodor-Boveri-Institut für Biowissenschaften, Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Gregory SH, Sagnimeni AJ, Wing EJ. Expression of the inlAB operon by Listeria monocytogenes is not required for entry into hepatic cells in vivo. Infect Immun 1996; 64:3983-6. [PMID: 8926058 PMCID: PMC174326 DOI: 10.1128/iai.64.10.3983-3986.1996] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Listeria monocytogenes injected intravenously into mice is taken up in the liver, where hepatocytes serve as the principal site of intracellular replication. The factors effecting entry of L. monocytogenes into hepatic cells remain to be determined. Others have shown that the protein products of the inlAB (internalin) operon are required for maximum entry of L. monocytogenes into a number of cell lines in vitro. Likewise, we report here that expression of the inlAB operon was required for maximum uptake of L. monocytogenes by primary cultures of mouse hepatocytes. Uptake of an inlAB mutant strain of L. monocytogenes was approximately 10-fold less than that of the isogenic wild-type control. In contrast, inlAB expression was not a factor in (i) clearance of L. monocytogenes injected intravenously into mice and taken up in the liver, (ii) the distribution of L. monocytogenes among hepatocytes and nonparenchymal cells in the liver, or (iii) internalization of L. monocytogenes by hepatic cells in vivo. These latter findings suggest that infection of hepatic cells by L. monocytogenes in vivo does not require the protein products of the inlAB operon.
Collapse
Affiliation(s)
- S H Gregory
- Department of Medicine, University of Pittsburgh Medical Center, Pennsylvania, USA
| | | | | |
Collapse
|
43
|
Hess J, Dreher A, Gentschev I, Goebel W, Ladel C, Miko D, Kaufmann SH. Protein p60 participates in intestinal host invasion by Listeria monocytogenes. ZENTRALBLATT FUR BAKTERIOLOGIE : INTERNATIONAL JOURNAL OF MEDICAL MICROBIOLOGY 1996; 284:263-72. [PMID: 8837387 DOI: 10.1016/s0934-8840(96)80102-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The role of p60 in intestinal invasion by Listeria monocytogenes was assessed after oral infection of mice with the p60 low-expressing mutant RIII, or with anti-p60 antibody coated wild-type EGD. Invasion by L. monocytogenes RIII bacteria has been unimpaired suggesting that a low density of p60 suffices for entry. Up to 24 h post infection (p.i.), intestinal penetration by L. monocytogenes EGD bacteria was markedly reduced by coating with anti-p60 antibodies. In histological sections, anti-p60 antibody-treated L. monocytogenes EGD, but not uncoated listeriae were still detectable 24 h p.i. at the apical surface of enterocytes in the intestine. We conclude that p60 contributes to host invasion through the natural port of listerial entry, the intestinal epithelium.
Collapse
Affiliation(s)
- J Hess
- Department of Immunology, University of Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Zwickey HL, Potter TA. Peptide epitopes from noncytosolic Listeria monocytogenes can be presented by major histocompatibility complex class I molecules. Infect Immun 1996; 64:1870-2. [PMID: 8613407 PMCID: PMC174008 DOI: 10.1128/iai.64.5.1870-1872.1996] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Listeria monocytogenes is an intracellular pathogen which escapes the phagosome and resides in the cytosol of the host cell. Using Listeria innocua and a mutant strain of L. monocytogenes (listeriolysin O negative), which do not enter the cytosol of the host cell, we demonstrate class I presentation of an epitope of p60, a protein secreted by L. monocytogenes, to a class I-restricted CD8+ cytotoxic T lymphocyte clone.
Collapse
Affiliation(s)
- H L Zwickey
- Department of Medicine, National Jewish Center for Immunology and Respiratory Medicine, Denver, CO 80206-2761, USA
| | | |
Collapse
|
45
|
Hess J, Gentschev I, Miko D, Welzel M, Ladel C, Goebel W, Kaufmann SH. Superior efficacy of secreted over somatic antigen display in recombinant Salmonella vaccine induced protection against listeriosis. Proc Natl Acad Sci U S A 1996; 93:1458-63. [PMID: 8643654 PMCID: PMC1079202 DOI: 10.1073/pnas.93.4.1458] [Citation(s) in RCA: 168] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Vaccination provides the most potent measure against infectious disease, and recombinant (r) viable vaccines expressing defined pathogen-derived antigens represent powerful candidates for future vaccination strategies. In a new approach we constructed r-aroA- Salmonella typhimurium displaying p60 or listeriolysin (Hly) antigen of Listeria monocytogenes in secreted or somatic form in the host cell. Vaccination of mice with r-aroA- S. typhimurium induced protection against the intracellular pathogen L. monocytogenes only with secreted and not with somatic antigen. Secreted Hly was slightly more potent in inducing protective immunity than secreted p60. Both r-aroA- S. typhimurium secreting p60 in the endosome and r-aroA- S. typhimurium secreting Hly in the cytosol induced protective CD4+ and CD8+ T-cells suggesting CD8+ T-cell stimulation independent from intracellular residence of r-aroA- S. typhimurium carriers. Hence, not only the type of antigen but also its display by the r-carrier within the host cell critically influences vaccine efficacy.
Collapse
Affiliation(s)
- J Hess
- Department of Immunology, University of Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|