1
|
Macela A, Kubelkova K. Why Does SARS-CoV-2 Infection Induce Autoantibody Production? Pathogens 2021; 10:380. [PMID: 33809954 PMCID: PMC8004127 DOI: 10.3390/pathogens10030380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/12/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
SARS-CoV-2 infection induces the production of autoantibodies, which is significantly associated with complications during hospitalization and a more severe prognosis in COVID-19 patients. Such a response of the patient's immune system may reflect (1) the dysregulation of the immune response or (2) it may be an attempt to regulate itself in situations where the non-infectious self poses a greater threat than the infectious non-self. Of significance may be the primary virus-host cell interaction where the surface-bound ACE2 ectoenzyme plays a critical role. Here, we present a brief analysis of recent findings concerning the immune recognition of SARS-CoV-2, which, we believe, favors the second possibility as the underlying reason for the production of autoantibodies during COVID-19.
Collapse
Affiliation(s)
| | - Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic;
| |
Collapse
|
2
|
Klimentova J, Pavkova I, Horcickova L, Bavlovic J, Kofronova O, Benada O, Stulik J. Francisella tularensis subsp. holarctica Releases Differentially Loaded Outer Membrane Vesicles Under Various Stress Conditions. Front Microbiol 2019; 10:2304. [PMID: 31649645 PMCID: PMC6795709 DOI: 10.3389/fmicb.2019.02304] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/20/2019] [Indexed: 11/16/2022] Open
Abstract
Francisella tularensis is a Gram-negative, facultative intracellular bacterium, causing a severe disease called tularemia. It secretes unusually shaped nanotubular outer membrane vesicles (OMV) loaded with a number of virulence factors and immunoreactive proteins. In the present study, the vesicles were purified from a clinical isolate of subsp. holarctica strain FSC200. We here provide a comprehensive proteomic characterization of OMV using a novel approach in which a comparison of OMV and membrane fraction is performed in order to find proteins selectively enriched in OMV vs. membrane. Only these proteins were further considered to be really involved in the OMV function and/or their exceptional structure. OMV were also isolated from bacteria cultured under various cultivation conditions simulating the diverse environments of F. tularensis life cycle. These included conditions mimicking the milieu inside the mammalian host during inflammation: oxidative stress, low pH, and high temperature (42°C); and in contrast, low temperature (25°C). We observed several-fold increase in vesiculation rate and significant protein cargo changes for high temperature and low pH. Further proteomic characterization of stress-derived OMV gave us an insight how the bacterium responds to the hostile environment of a mammalian host through the release of differentially loaded OMV. Among the proteins preferentially and selectively packed into OMV during stressful cultivations, the previously described virulence factors connected to the unique intracellular trafficking of Francisella were detected. Considerable changes were also observed in a number of proteins involved in the biosynthesis and metabolism of the bacterial envelope components like O-antigen, lipid A, phospholipids, and fatty acids. Data are available via ProteomeXchange with identifier PXD013074.
Collapse
Affiliation(s)
- Jana Klimentova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Czechia
| | - Ivona Pavkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Czechia
| | - Lenka Horcickova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Czechia
| | - Jan Bavlovic
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Czechia
| | - Olga Kofronova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Oldrich Benada
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia.,Faculty of Science, Jan Evangelista Purkyně University, Ústí nad Labem, Czechia
| | - Jiri Stulik
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Czechia
| |
Collapse
|
3
|
Fulton KM, Ananchenko A, Wolfraim L, Martin S, Twine SM. Classical Immunoproteomics: Serological Proteome Analysis (SERPA) for Antigen Identification. Methods Mol Biol 2019; 2024:59-78. [PMID: 31364042 DOI: 10.1007/978-1-4939-9597-4_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The study of the humoral immune response to infectious and chronic diseases is important for understanding the disease progression, identification of protective antigens, vaccine development, and discovery of biomarkers for early diagnosis. Proteomic approaches, including serological proteome analysis (SERPA), have been used to identify the repertoire of immunoreactive proteins in various diseases. In this chapter, we provide an outline of the SERPA approach, using the analysis of sera from mice vaccinated with a live attenuated tularemia vaccine as an example.
Collapse
Affiliation(s)
- Kelly M Fulton
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada.
| | - Anna Ananchenko
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada
| | | | | | - Susan M Twine
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada
| |
Collapse
|
4
|
Shahin K, Thompson KD, Inglis NF, Mclean K, Ramirez-Paredes JG, Monaghan SJ, Hoare R, Fontaine M, Metselaar M, Adams A. Characterization of the outer membrane proteome of Francisella noatunensis subsp. orientalis. J Appl Microbiol 2018; 125:686-699. [PMID: 29777634 DOI: 10.1111/jam.13918] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/03/2018] [Accepted: 05/11/2018] [Indexed: 11/28/2022]
Abstract
AIMS The aims of the current study were to characterize the outer membrane proteins (OMPs) of Francisella noatunensis subsp. orientalis (Fno) STIR-GUS-F2f7, and identify proteins recognized by sera from tilapia, Oreochromis niloticus, (L) that survived experimental challenge with Fno. METHODS AND RESULTS The composition of the OMPs of a virulent strain of Fno (STIR-GUS-F2f7), isolated from diseased red Nile tilapia in the United Kingdom, was examined. The sarcosine-insoluble OMPs fraction was screened with tilapia hyperimmune sera by western blot analysis following separation of the proteins by 1D SDS-PAGE. Liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) was used to identify the various proteins present in the OMP profile. Two hundred and thirty-nine proteins were identified, of which 44 were found in the immunogenic band recognized by the tilapia hyperimmune serum. In silico analysis was performed to predict the function and location of the OMPs identified by MS. CONCLUSIONS Using a powerful proteomic-based approach in conjugation with western immunoblotting, proteins comprising the outer membrane fraction of Fno STIR-GUS-F2f7 were identified, catalogued and screened for immune recognition by tilapia sera. SIGNIFICANCE AND IMPACT OF THE STUDY The current study is the first report on the characterization of Fno-OMPs. The findings here provide preliminary data on bacterial surface proteins that exist in direct contact with the host's immune defences during infection and offer an insight into the pathogenesis of Fno.
Collapse
Affiliation(s)
- K Shahin
- Faculty of Natural Sciences, Institute of Aquaculture, University of Stirling, Stirling, Scotland, UK.,Aquatic Animals Diseases Lab, Aquaculture Division, National Institute of Oceanography and Fisheries, Suez, Egypt
| | - K D Thompson
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, UK
| | - N F Inglis
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, UK
| | - K Mclean
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, UK
| | - J G Ramirez-Paredes
- Faculty of Natural Sciences, Institute of Aquaculture, University of Stirling, Stirling, Scotland, UK
| | - S J Monaghan
- Faculty of Natural Sciences, Institute of Aquaculture, University of Stirling, Stirling, Scotland, UK
| | - R Hoare
- Faculty of Natural Sciences, Institute of Aquaculture, University of Stirling, Stirling, Scotland, UK
| | - M Fontaine
- Benchmark Animal Health, Bush House, Edinburgh Technopole, Edinburgh, Midlothian, UK
| | - M Metselaar
- Benchmark Animal Health, Bush House, Edinburgh Technopole, Edinburgh, Midlothian, UK
| | - A Adams
- Faculty of Natural Sciences, Institute of Aquaculture, University of Stirling, Stirling, Scotland, UK
| |
Collapse
|
5
|
McCormick AA, Shakeel A, Yi C, Kaur H, Mansour AM, Bakshi CS. Intranasal administration of a two-dose adjuvanted multi-antigen TMV-subunit conjugate vaccine fully protects mice against Francisella tularensis LVS challenge. PLoS One 2018; 13:e0194614. [PMID: 29684046 PMCID: PMC5912714 DOI: 10.1371/journal.pone.0194614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/06/2018] [Indexed: 12/03/2022] Open
Abstract
Tularemia is a fatal human disease caused by Francisella tularensis, a Gram-negative encapsulated coccobacillus bacterium. Due to its low infectious dose, ease of aerosolized transmission, and lethal effects, the CDC lists F. tularensis as a Category A pathogen, the highest level for a potential biothreat agent. Previous vaccine studies have been conducted with live attenuated, inactivated, and subunit vaccines, which have achieved partial or full protection from F. tularensis live vaccine strain (LVS) challenge, but no vaccine has been approved for human use. We demonstrate the improved efficacy of a multi-antigen subunit vaccine by using Tobacco Mosaic virus (TMV) as an antigen carrier for the F. tularensis SchuS4 proteins DnaK, OmpA, SucB and Tul4 (DOST). The magnitude and quality of immune responses were compared after mice were immunized by subcutaneous or intranasal routes of administration with a TMV-DOST mixture, with or without four different adjuvants. Immune responses varied in magnitude and isotype profile, by antigen, by route of administration, and by protection in an F. tularensis LVS challenge model of disease. Interestingly, our analysis demonstrates an overwhelming IgG2 response to SucB after intranasal dosing, as well as a robust cellular response, which may account for the improved two-dose survival imparted by the tetravalent vaccine, compared to a previous study that tested efficacy of TMV-DOT. Our study provides evidence that potent humoral, cellular and mucosal immunity can be achieved by optimal antigen combination, delivery, adjuvant and appropriate route of administration, to improve vaccine potency and provide protection from pathogen challenge.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/analysis
- Antibodies, Bacterial/metabolism
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/metabolism
- Bacterial Vaccines/immunology
- Disease Models, Animal
- Female
- Francisella tularensis/immunology
- Immunity, Cellular
- Immunoglobulin G/analysis
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Immunoglobulin Isotypes/immunology
- Immunoglobulin Isotypes/metabolism
- Mice
- Mice, Inbred C57BL
- Survival Rate
- Tobacco Mosaic Virus/genetics
- Tobacco Mosaic Virus/metabolism
- Tularemia/immunology
- Tularemia/microbiology
- Tularemia/prevention & control
- Vaccines, Conjugate/immunology
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
| | - Aisha Shakeel
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Chris Yi
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Hardeep Kaur
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Ahd M. Mansour
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY
| | | |
Collapse
|
6
|
Holland KM, Rosa SJ, Kristjansdottir K, Wolfgeher D, Franz BJ, Zarrella TM, Kumar S, Sunagar R, Singh A, Bakshi CS, Namjoshi P, Barry EM, Sellati TJ, Kron SJ, Gosselin EJ, Reed DS, Hazlett KRO. Differential Growth of Francisella tularensis, Which Alters Expression of Virulence Factors, Dominant Antigens, and Surface-Carbohydrate Synthases, Governs the Apparent Virulence of Ft SchuS4 to Immunized Animals. Front Microbiol 2017; 8:1158. [PMID: 28690600 PMCID: PMC5479911 DOI: 10.3389/fmicb.2017.01158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 06/07/2017] [Indexed: 12/29/2022] Open
Abstract
The gram-negative bacterium Francisella tularensis (Ft) is both a potential biological weapon and a naturally occurring microbe that survives in arthropods, fresh water amoeba, and mammals with distinct phenotypes in various environments. Previously, we used a number of measurements to characterize Ft grown in Brain-Heart Infusion (BHI) broth as (1) more similar to infection-derived bacteria, and (2) slightly more virulent in naïve animals, compared to Ft grown in Mueller Hinton Broth (MHB). In these studies we observed that the free amino acids in MHB repress expression of select Ft virulence factors by an unknown mechanism. Here, we tested the hypotheses that Ft grown in BHI (BHI-Ft) accurately displays a full protein composition more similar to that reported for infection-derived Ft and that this similarity would make BHI-Ft more susceptible to pre-existing, vaccine-induced immunity than MHB-Ft. We performed comprehensive proteomic analysis of Ft grown in MHB, BHI, and BHI supplemented with casamino acids (BCA) and compared our findings to published “omics” data derived from Ft grown in vivo. Based on the abundance of ~1,000 proteins, the fingerprint of BHI-Ft is one of nutrient-deprived bacteria that—through induction of a stringent-starvation-like response—have induced the FevR regulon for expression of the bacterium's virulence factors, immuno-dominant antigens, and surface-carbohydrate synthases. To test the notion that increased abundance of dominant antigens expressed by BHI-Ft would render these bacteria more susceptible to pre-existing, vaccine-induced immunity, we employed a battery of LVS-vaccination and S4-challenge protocols using MHB- and BHI-grown Ft S4. Contrary to our hypothesis, these experiments reveal that LVS-immunization provides a barrier to infection that is significantly more effective against an MHB-S4 challenge than a BHI-S4 challenge. The differences in apparent virulence to immunized mice are profoundly greater than those observed with primary infection of naïve mice. Our findings suggest that tularemia vaccination studies should be critically evaluated in regard to the growth conditions of the challenge agent.
Collapse
Affiliation(s)
- Kristen M Holland
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Sarah J Rosa
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | | | - Donald Wolfgeher
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicago, IL, United States
| | - Brian J Franz
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Tiffany M Zarrella
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Raju Sunagar
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Anju Singh
- Trudeau InstituteSaranac Lake, NY, United States
| | - Chandra S Bakshi
- Department of Microbiology and Immunology, New York Medical CollegeValhalla, NY, United States
| | - Prachi Namjoshi
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Eileen M Barry
- School of Medicine, University of MarylandBaltimore, MD, United States
| | | | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicago, IL, United States
| | - Edmund J Gosselin
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Douglas S Reed
- Center for Vaccine Research, University of PittsburghPittsburgh, PA, United States
| | - Karsten R O Hazlett
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| |
Collapse
|
7
|
Gaur R, Alam SI, Kamboj DV. Immunoproteomic Analysis of Antibody Response of Rabbit Host Against Heat-Killed Francisella tularensis Live Vaccine Strain. Curr Microbiol 2017; 74:499-507. [PMID: 28233060 DOI: 10.1007/s00284-017-1217-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/10/2017] [Indexed: 11/24/2022]
Abstract
Francisella tularensis, the causative agent of tularemia, has attained the status of one of the high priority agents that could be used in the act of bioterrorism. Currently, there is no licensed vaccine for this highly infectious intracellular pathogen. Being a listed 'Category A' agent of the U.S. Center for Disease Control and Prevention (CDC), vaccines and therapeutics are immediately required against this pathogen. In this study, an immunoproteomic approach based on the techniques of 2-dimensional gel electrophoresis (2DE) and immunoblotting combined with mass spectrometry (MS) was used for elucidation of immunogenic components and putative vaccine candidates. Whole-cell soluble protein extract of F. tularensis LVS (Ft LVS) was separated by 2DE, and immunoblots were developed with sera raised in rabbit after immunization with heat-killed Ft LVS. A total of 28 immunoreactive proteins were identified by tandem mass spectrometry. Rabbit immunoproteome of F. tularensis was compared with those previously reported using sera from human patients and in murine model. Out of 28 immunoreactive proteins identified in this study, 12 and 17 overlapping proteins were recognized by human and murine sera, respectively. Nine proteins were found immunogenic in all the three hosts, while eight new immunogenic proteins were found in this study. Identified immunoreactive proteins may find application in design and development of protein subunit vaccine for tularemia.
Collapse
Affiliation(s)
- Ritu Gaur
- Biotechnology Division, Defence Research and Development Establishment, Gwalior, 474002, India
| | - Syed Imteyaz Alam
- Biotechnology Division, Defence Research and Development Establishment, Gwalior, 474002, India
| | - Dev Vrat Kamboj
- Biotechnology Division, Defence Research and Development Establishment, Gwalior, 474002, India.
| |
Collapse
|
8
|
Jia Q, Bowen R, Lee BY, Dillon BJ, Masleša-Galić S, Horwitz MA. Francisella tularensis Live Vaccine Strain deficient in capB and overexpressing the fusion protein of IglA, IglB, and IglC from the bfr promoter induces improved protection against F. tularensis respiratory challenge. Vaccine 2016; 34:4969-4978. [PMID: 27577555 DOI: 10.1016/j.vaccine.2016.08.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/18/2016] [Accepted: 08/12/2016] [Indexed: 10/21/2022]
Abstract
A safer and more effective vaccine than the unlicensed Francisella tularensis Live Vaccine Strain (LVS) is needed to protect against the biowarfare agent F. tularensis. Previously, we developed an LVS ΔcapB mutant that is significantly safer than LVS and provides potent protective immunity against F. tularensis respiratory challenge when administered intranasally but limited protection when administered intradermally unless as part of a prime-boost vaccination strategy. To improve the immunogenicity and efficacy of LVS ΔcapB, we developed recombinant LVS ΔcapB (rLVS ΔcapB) strains overexpressing various F. tularensis Francisella Pathogenicity Island (FPI) proteins - IglA, IglB and IglC, and a fusion protein (IglABC) comprising immunodominant epitopes of IglA, IglB, and IglC downstream of different Francisella promoters, including the bacterioferritin (bfr) promoter. We show that rLVS ΔcapB/bfr-iglA, iglB, iglC, and iglABC express more IglA, IglB, IglC or IglABC than parental LVS ΔcapB in broth and in human macrophages, and stably express FPI proteins in macrophages and mice absent antibiotic selection. In response to IglC and heat-inactivated LVS, spleen cells from mice immunized intradermally with rLVS ΔcapB/bfr-iglC or bfr-iglABC secrete greater amounts of interferon-gamma and/or interleukin-17 than those from mice immunized with LVS ΔcapB, comparable to those from LVS-immunized mice. Mice immunized with rLVS ΔcapB/bfr-iglA, iglB, iglC or iglABC produce serum antibodies at levels similar to LVS-immunized mice. Mice immunized intradermally with rLVS ΔcapB/bfr-iglABC and challenged intranasally with virulent F. tularensis Schu S4 survive longer than sham- and LVS ΔcapB-immunized mice. Mice immunized intranasally with rLVS ΔcapB/bfr-iglABC - but not with LVS - just before or after respiratory challenge with F. tularensis Schu S4 are partially protected; protection is correlated with induction of a strong innate immune response. Thus, rLVS ΔcapB/bfr-iglABC shows improved immunogenicity and protective efficacy compared with parental LVS ΔcapB and, in contrast to LVS, has partial efficacy as immediate pre- and post-exposure prophylaxis.
Collapse
Affiliation(s)
- Qingmei Jia
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California - Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1688, United States.
| | - Richard Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States.
| | - Bai-Yu Lee
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California - Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1688, United States.
| | - Barbara Jane Dillon
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California - Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1688, United States.
| | - Saša Masleša-Galić
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California - Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1688, United States.
| | - Marcus A Horwitz
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California - Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1688, United States.
| |
Collapse
|
9
|
Rowe HM, Huntley JF. From the Outside-In: The Francisella tularensis Envelope and Virulence. Front Cell Infect Microbiol 2015; 5:94. [PMID: 26779445 PMCID: PMC4688374 DOI: 10.3389/fcimb.2015.00094] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022] Open
Abstract
Francisella tularensis is a highly-infectious bacterium that causes the rapid, and often lethal disease, tularemia. Many studies have been performed to identify and characterize the virulence factors that F. tularensis uses to infect a wide variety of hosts and host cell types, evade immune defenses, and induce severe disease and death. This review focuses on the virulence factors that are present in the F. tularensis envelope, including capsule, LPS, outer membrane, periplasm, inner membrane, secretion systems, and various molecules in each of aforementioned sub-compartments. Whereas, no single bacterial molecule or molecular complex single-handedly controls F. tularensis virulence, we review here how diverse bacterial systems work in conjunction to subvert the immune system, attach to and invade host cells, alter phagosome/lysosome maturation pathways, replicate in host cells without being detected, inhibit apoptosis, and induce host cell death for bacterial release and infection of adjacent cells. Given that the F. tularensis envelope is the outermost layer of the bacterium, we highlight herein how many of these molecules directly interact with the host to promote infection and disease. These and future envelope studies are important to advance our collective understanding of F. tularensis virulence mechanisms and offer targets for future vaccine development efforts.
Collapse
Affiliation(s)
- Hannah M Rowe
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Jason F Huntley
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| |
Collapse
|
10
|
Development of a Multivalent Subunit Vaccine against Tularemia Using Tobacco Mosaic Virus (TMV) Based Delivery System. PLoS One 2015; 10:e0130858. [PMID: 26098553 PMCID: PMC4476615 DOI: 10.1371/journal.pone.0130858] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/26/2015] [Indexed: 11/21/2022] Open
Abstract
Francisella tularensis is a facultative intracellular pathogen, and is the causative agent of a fatal human disease known as tularemia. F. tularensis is classified as a Category A Biothreat agent by the CDC based on its use in bioweapon programs by several countries in the past and its potential to be used as an agent of bioterrorism. No licensed vaccine is currently available for prevention of tularemia. In this study, we used a novel approach for development of a multivalent subunit vaccine against tularemia by using an efficient tobacco mosaic virus (TMV) based delivery platform. The multivalent subunit vaccine was formulated to contain a combination of F. tularensis protective antigens: OmpA-like protein (OmpA), chaperone protein DnaK and lipoprotein Tul4 from the highly virulent F. tularensis SchuS4 strain. Two different vaccine formulations and immunization schedules were used. The immunized mice were challenged with lethal (10xLD100) doses of F. tularensis LVS on day 28 of the primary immunization and observed daily for morbidity and mortality. Results from this study demonstrate that TMV can be used as a carrier for effective delivery of multiple F. tularensis antigens. TMV-conjugate vaccine formulations are safe and multiple doses can be administered without causing any adverse reactions in immunized mice. Immunization with TMV-conjugated F. tularensis proteins induced a strong humoral immune response and protected mice against respiratory challenges with very high doses of F. tularensis LVS. This study provides a proof-of-concept that TMV can serve as a suitable platform for simultaneous delivery of multiple protective antigens of F. tularensis. Refinement of vaccine formulations coupled with TMV-targeting strategies developed in this study will provide a platform for development of an effective tularemia subunit vaccine as well as a vaccination approach that may broadly be applicable to many other bacterial pathogens.
Collapse
|
11
|
The sweet tooth of bacteria: common themes in bacterial glycoconjugates. Microbiol Mol Biol Rev 2015; 78:372-417. [PMID: 25184559 DOI: 10.1128/mmbr.00007-14] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Humans have been increasingly recognized as being superorganisms, living in close contact with a microbiota on all their mucosal surfaces. However, most studies on the human microbiota have focused on gaining comprehensive insights into the composition of the microbiota under different health conditions (e.g., enterotypes), while there is also a need for detailed knowledge of the different molecules that mediate interactions with the host. Glycoconjugates are an interesting class of molecules for detailed studies, as they form a strain-specific barcode on the surface of bacteria, mediating specific interactions with the host. Strikingly, most glycoconjugates are synthesized by similar biosynthesis mechanisms. Bacteria can produce their major glycoconjugates by using a sequential or an en bloc mechanism, with both mechanistic options coexisting in many species for different macromolecules. In this review, these common themes are conceptualized and illustrated for all major classes of known bacterial glycoconjugates, with a special focus on the rather recently emergent field of glycosylated proteins. We describe the biosynthesis and importance of glycoconjugates in both pathogenic and beneficial bacteria and in both Gram-positive and -negative organisms. The focus lies on microorganisms important for human physiology. In addition, the potential for a better knowledge of bacterial glycoconjugates in the emerging field of glycoengineering and other perspectives is discussed.
Collapse
|
12
|
Chandler JC, Sutherland MD, Harton MR, Molins CR, Anderson RV, Heaslip DG, Bosio CM, Belisle JT. Francisella tularensis LVS surface and membrane proteins as targets of effective post-exposure immunization for tularemia. J Proteome Res 2014; 14:664-75. [PMID: 25494920 PMCID: PMC4324441 DOI: 10.1021/pr500628k] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Francisella tularensis causes disease (tularemia)
in a large number of mammals, including man. We previously demonstrated
enhanced efficacy of conventional antibiotic therapy for tularemia
by postexposure passive transfer of immune sera developed against
a F. tularensis LVS membrane protein fraction (MPF).
However, the protein composition of this immunogenic fraction was
not defined. Proteomic approaches were applied to define the protein
composition and identify the immunogens of MPF. MPF consisted of at
least 299 proteins and 2-D Western blot analyses using sera from MPF-immunized
and F. tularensis LVS-vaccinated mice coupled to
liquid chromatography–tandem mass spectrometry identified 24
immunoreactive protein spots containing 45 proteins. A reverse vaccinology
approach that applied labeling of F. tularensis LVS
surface proteins and bioinformatics was used to reduce the complexity
of potential target immunogens. Bioinformatics analyses of the immunoreactive
proteins reduced the number of immunogen targets to 32. Direct surface
labeling of F. tularensis LVS resulted in the identification
of 31 surface proteins. However, only 13 of these were reactive with
MPF and/or F. tularensis LVS immune sera. Collectively,
this use of orthogonal proteomic approaches reduced the complexity
of potential immunogens in MPF by 96% and allowed for prioritization
of target immunogens for antibody-based immunotherapies against tularemia.
Collapse
Affiliation(s)
- Jeffrey C Chandler
- Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Department of Microbiology, Immunology, and Pathology, Colorado State University , Campus Delivery 0922, Fort Collins 80523, Colorado, United States
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Lu Z, Rynkiewicz MJ, Madico G, Li S, Yang CY, Perkins HM, Sompuram SR, Kodela V, Liu T, Morris T, Wang D, Roche MI, Seaton BA, Sharon J. B-cell epitopes in GroEL of Francisella tularensis. PLoS One 2014; 9:e99847. [PMID: 24968190 PMCID: PMC4072690 DOI: 10.1371/journal.pone.0099847] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/16/2014] [Indexed: 01/01/2023] Open
Abstract
The chaperonin protein GroEL, also known as heat shock protein 60 (Hsp60), is a prominent antigen in the human and mouse antibody response to the facultative intracellular bacterium Francisella tularensis (Ft), the causative agent of tularemia. In addition to its presumed cytoplasmic location, FtGroEL has been reported to be a potential component of the bacterial surface and to be released from the bacteria. In the current study, 13 IgG2a and one IgG3 mouse monoclonal antibodies (mAbs) specific for FtGroEL were classified into eleven unique groups based on shared VH-VL germline genes, and seven crossblocking profiles revealing at least three non-overlapping epitope areas in competition ELISA. In a mouse model of respiratory tularemia with the highly pathogenic Ft type A strain SchuS4, the Ab64 and N200 IgG2a mAbs, which block each other’s binding to and are sensitive to the same two point mutations in FtGroEL, reduced bacterial burden indicating that they target protective GroEL B-cell epitopes. The Ab64 and N200 epitopes, as well as those of three other mAbs with different crossblocking profiles, Ab53, N3, and N30, were mapped by hydrogen/deuterium exchange–mass spectrometry (DXMS) and visualized on a homology model of FtGroEL. This model was further supported by its experimentally-validated computational docking to the X-ray crystal structures of Ab64 and Ab53 Fabs. The structural analysis and DXMS profiles of the Ab64 and N200 mAbs suggest that their protective effects may be due to induction or stabilization of a conformational change in FtGroEL.
Collapse
Affiliation(s)
- Zhaohua Lu
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Michael J. Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Guillermo Madico
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sheng Li
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Chiou-Ying Yang
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Hillary M. Perkins
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Seshi R. Sompuram
- Medical Discovery Partners, LLC, Boston, Massachusetts, United States of America
| | - Vani Kodela
- Medical Discovery Partners, LLC, Boston, Massachusetts, United States of America
| | - Tong Liu
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Timothy Morris
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Daphne Wang
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Marly I. Roche
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Barbara A. Seaton
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jacqueline Sharon
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
14
|
Pávková I, Brychta M, Strašková A, Schmidt M, Macela A, Stulík J. Comparative proteome profiling of host–pathogen interactions: insights into the adaptation mechanisms of Francisella tularensis in the host cell environment. Appl Microbiol Biotechnol 2013; 97:10103-15. [DOI: 10.1007/s00253-013-5321-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 09/30/2013] [Accepted: 10/09/2013] [Indexed: 11/30/2022]
|
15
|
A heterologous prime-boost vaccination strategy comprising the Francisella tularensis live vaccine strain capB mutant and recombinant attenuated Listeria monocytogenes expressing F. tularensis IglC induces potent protective immunity in mice against virulent F. tularensis aerosol challenge. Infect Immun 2013; 81:1550-61. [PMID: 23439306 DOI: 10.1128/iai.01013-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Francisella tularensis, the causative agent of tularemia, is a category A bioterrorism agent. A vaccine that is safer and more effective than the currently available unlicensed F. tularensis live vaccine strain (LVS) is needed to protect against intentional release of aerosolized F. tularensis, the most dangerous type of exposure. In this study, we employed a heterologous prime-boost vaccination strategy comprising intradermally administered LVS ΔcapB (highly attenuated capB-deficient LVS mutant) as the primer vaccine and rLm/iglC (recombinant attenuated Listeria monocytogenes expressing the F. tularensis immunoprotective antigen IglC) as the booster vaccine. Boosting LVS ΔcapB-primed mice with rLm/iglC significantly enhanced T cell immunity; their splenic T cells secreted significantly more gamma interferon (IFN-γ) and had significantly more cytokine (IFN-γ and/or tumor necrosis factor [TNF] and/or interleukin-2 [IL-2])-producing CD4(+) and CD8(+) T cells upon in vitro IglC stimulation. Importantly, mice primed with LVS ΔcapB or rLVS ΔcapB/IglC, boosted with rLm/iglC, and subsequently challenged with 10 50% lethal doses (LD50) of aerosolized highly virulent F. tularensis Schu S4 had a significantly higher survival rate and mean survival time than mice immunized with only LVS ΔcapB (P < 0.0001); moreover, compared with mice immunized once with LVS, primed-boosted mice had a higher survival rate (75% versus 62.5%) and mean survival time during the first 21 days postchallenge (19 and 20 days for mice boosted after being primed with LVS ΔcapB and rLVS ΔcapB/IglC, respectively, versus 17 days for mice immunized with LVS) and maintained their weight significantly better (P < 0.01). Thus, the LVS ΔcapB-rLm/iglC prime-boost vaccination strategy holds substantial promise for a vaccine that is safer and at least as potent as LVS.
Collapse
|
16
|
Abstract
The varied landscape of the adaptive immune response is determined by the peptides presented by immune cells, derived from viral or microbial pathogens or cancerous cells. The study of immune biomarkers or antigens is not new and classical methods such as agglutination, enzyme-linked immunosorbent assay, or Western blotting have been used for many years to study the immune response to vaccination or disease. However, in many of these traditional techniques, protein or peptide identification has often been the bottleneck. Recent advances in genomics and proteomics, has led to many of the rapid advances in proteomics approaches. Immunoproteomics describes a rapidly growing collection of approaches that have the common goal of identifying and measuring antigenic peptides or proteins. This includes gel based, array based, mass spectrometry, DNA based, or in silico approaches. Immunoproteomics is yielding an understanding of disease and disease progression, vaccine candidates, and biomarkers. This review gives an overview of immunoproteomics and closely related technologies that are used to define the full set of antigens targeted by the immune system during disease.
Collapse
Affiliation(s)
- Kelly M Fulton
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | | |
Collapse
|
17
|
Fulton KM, Martin SS, Wolfraim L, Twine SM. Methods and applications of serological proteome analysis. Methods Mol Biol 2013; 1061:97-112. [PMID: 23963932 DOI: 10.1007/978-1-62703-589-7_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The study of the humoral response to infectious diseases and chronic diseases, such as cancer, is important for many reasons, including understanding the host response to disease, identification of protective antigens, vaccine development, and discovery of biomarkers for early diagnosis. During the past decade, proteomic approaches, such as serological proteome analysis (SERPA), have been used to identify the repertoire of immunoreactive proteins in various diseases. In this chapter, we provide an outline of the SERPA approach, using the analysis of sera from mice vaccinated with a live attenuated tularemia vaccine as an example.
Collapse
Affiliation(s)
- Kelly M Fulton
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | | | | | | |
Collapse
|
18
|
Production of outer membrane vesicles and outer membrane tubes by Francisella novicida. J Bacteriol 2012; 195:1120-32. [PMID: 23264574 DOI: 10.1128/jb.02007-12] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella spp. are highly infectious and virulent bacteria that cause the zoonotic disease tularemia. Knowledge is lacking for the virulence factors expressed by Francisella and how these factors are secreted and delivered to host cells. Gram-negative bacteria constitutively release outer membrane vesicles (OMV), which may function in the delivery of virulence factors to host cells. We identified growth conditions under which Francisella novicida produces abundant OMV. Purification of the vesicles revealed the presence of tube-shaped vesicles in addition to typical spherical OMV, and examination of whole bacteria revealed the presence of tubes extending out from the bacterial surface. Recently, both prokaryotic and eukaryotic cells have been shown to produce membrane-enclosed projections, termed nanotubes, which appear to function in cell-cell communication and the exchange of molecules. In contrast to these previously characterized structures, the F. novicida tubes are produced in liquid as well as on solid medium and are derived from the OM rather than the cytoplasmic membrane. The production of the OMV and tubes (OMV/T) by F. novicida was coordinately regulated and responsive to both growth medium and growth phase. Proteomic analysis of purified OMV/T identified known Francisella virulence factors among the constituent proteins, suggesting roles for the vesicles in pathogenesis. In support of this, production of OM tubes by F. novicida was stimulated during infection of macrophages and addition of purified OMV/T to macrophages elicited increased release of proinflammatory cytokines. Finally, vaccination with purified OMV/T protected mice from subsequent challenge with highly lethal doses of F. novicida.
Collapse
|
19
|
Twine S, Shen H, Harris G, Chen W, Sjostedt A, Ryden P, Conlan W. BALB/c mice, but not C57BL/6 mice immunized with a ΔclpB mutant of Francisella tularensis subspecies tularensis are protected against respiratory challenge with wild-type bacteria: association of protection with post-vaccination and post-challenge immune responses. Vaccine 2012; 30:3634-45. [PMID: 22484348 DOI: 10.1016/j.vaccine.2012.03.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 02/23/2012] [Accepted: 03/16/2012] [Indexed: 01/08/2023]
Abstract
Francisella tularensis subspecies tularensis is highly virulent for humans especially when it is inhaled. Therefore, it has the potential to be used as a biothreat agent. Vaccines against F. tularensis will need to be approved in accordance with the FDA Animal Rule. This will require identification of robust correlates of protection in experimental animals and the demonstration that similar immune responses are generated in vaccinated humans. Towards this goal, we have developed an experimental live vaccine strain by deleting the gene, clpB, encoding a heat shock protein from virulent subsp. tularensis strain, SCHU S4. SCHU S4ΔclpB administered intradermally protects BALB/c, but not C57BL/6 mice from subsequent respiratory challenge with wildtype SCHU S4. A comparison of post-vaccination and post-challenge immune responses in these two mouse strains shows an association between several antibody and cytokine responses and protection. In particular, elevated IFNγ levels in the skin 2 days after vaccination, sero-conversion to hypothetical membrane protein FTT_1778c, and to 30S ribosomal protein S1 (FTT_0183c) of F. tularensis after 30 days of vaccination, and elevated levels of pulmonary IL-17 on day 7 after respiratory challenge with SCHU S4 were all associated with protection.
Collapse
Affiliation(s)
- Susan Twine
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ontario, K1A 0R6, Canada
| | | | | | | | | | | | | |
Collapse
|
20
|
De Pascalis R, Chou AY, Bosio CM, Huang CY, Follmann DA, Elkins KL. Development of functional and molecular correlates of vaccine-induced protection for a model intracellular pathogen, F. tularensis LVS. PLoS Pathog 2012; 8:e1002494. [PMID: 22275868 PMCID: PMC3262015 DOI: 10.1371/journal.ppat.1002494] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 12/06/2011] [Indexed: 11/19/2022] Open
Abstract
In contrast with common human infections for which vaccine efficacy can be evaluated directly in field studies, alternative strategies are needed to evaluate efficacy for slowly developing or sporadic diseases like tularemia. For diseases such as these caused by intracellular bacteria, serological measures of antibodies are generally not predictive. Here, we used vaccines varying in efficacy to explore development of clinically useful correlates of protection for intracellular bacteria, using Francisella tularensis as an experimental model. F. tularensis is an intracellular bacterium classified as Category A bioterrorism agent which causes tularemia. The primary vaccine candidate in the U.S., called Live Vaccine Strain (LVS), has been the subject of ongoing clinical studies; however, safety and efficacy are not well established, and LVS is not licensed by the U.S. FDA. Using a mouse model, we compared the in vivo efficacy of a panel of qualitatively different Francisella vaccine candidates, the in vitro functional activity of immune lymphocytes derived from vaccinated mice, and relative gene expression in immune lymphocytes. Integrated analyses showed that the hierarchy of protection in vivo engendered by qualitatively different vaccines was reflected by the degree of lymphocytes' in vitro activity in controlling the intramacrophage growth of Francisella. Thus, this assay may be a functional correlate. Further, the strength of protection was significantly related to the degree of up-regulation of expression of a panel of genes in cells recovered from the assay. These included IFN-γ, IL-6, IL-12Rβ2, T-bet, SOCS-1, and IL-18bp. Taken together, the results indicate that an in vitro assay that detects control of bacterial growth, and/or a selected panel of mediators, may ultimately be developed to predict the outcome of vaccine efficacy and to complement clinical trials. The overall approach may be applicable to intracellular pathogens in general. Diseases such as tuberculosis (caused by Mycobacterium tuberculosis) or tularemia (caused by Francisella tularensis) result from infections by microbes that live within cells of a person's body. New vaccines are being developed against such intracellular pathogens, but some will be difficult to test, because disease takes a long time to develop (e.g., tuberculosis) or because outbreaks are unpredictable (e.g., tularemia). Usually such infections are controlled by activities of T cells. However, there are no accepted measures of T cell function that reliably predict vaccine-induced protection. We studied two new ways to do so. We used a group of vaccine candidates against tularemia that stimulated good, fair, or poor protection of mice against Francisella challenge. We then measured whether Francisella–immune cells from vaccinated mice controlled the growth of bacteria inside cells, and/or whether the expression of immune genes in Francisella–immune cells was increased. We found that the degree of protection was matched by the degree of the cells' function in controlling intramacrophage bacterial growth. Further, the degree was predicted by relative amounts of gene expression for several immune mediators. Thus the two new options explored here may help predict protection, without waiting for the onset of disease.
Collapse
Affiliation(s)
- Roberto De Pascalis
- Laboratory of Mycobacterial Diseases and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Rockville, Maryland, United States of America
| | - Alicia Y. Chou
- Laboratory of Mycobacterial Diseases and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Rockville, Maryland, United States of America
| | - Catharine M. Bosio
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, Montana, United States of America
| | - Chiung-Yu Huang
- Biostatistics Research Branch, Division of Clinical Research, NIAID/NIH, Bethesda, Maryland, United States of America
| | - Dean A. Follmann
- Biostatistics Research Branch, Division of Clinical Research, NIAID/NIH, Bethesda, Maryland, United States of America
| | - Karen L. Elkins
- Laboratory of Mycobacterial Diseases and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
21
|
Conlan JW. Tularemia vaccines: recent developments and remaining hurdles. Future Microbiol 2011; 6:391-405. [PMID: 21526941 DOI: 10.2217/fmb.11.22] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Francisella tularensis subsp. tularensis is a facultative intracellular bacterial pathogen of humans and other mammals. Its inhaled infectious dose is very low and can result in very high mortality. Historically, subsp. tularensis was developed as a biological weapon and there are now concerns about its abuse as such by terrorists. A live attenuated vaccine developed pragmatically more than half a century ago from the less virulent holarctica subsp. is the sole prophylactic available, but it remains unlicensed. In recent years several other potential live, killed and subunit vaccine candidates have been developed and tested in mice for their efficacy against respiratory challenge with subsp. tularensis. This article will review these vaccine candidates and the development hurdles they face.
Collapse
Affiliation(s)
- J Wayne Conlan
- National Research Council, Institute for Biological Sciences, Ottawa, Ontario, Canada.
| |
Collapse
|
22
|
Fulton KM, Zhao X, Petit MD, Kilmury SLN, Wolfraim LA, House RV, Sjostedt A, Twine SM. Immunoproteomic analysis of the human antibody response to natural tularemia infection with Type A or Type B strains or LVS vaccination. Int J Med Microbiol 2011; 301:591-601. [PMID: 21873113 DOI: 10.1016/j.ijmm.2011.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 05/24/2011] [Accepted: 07/03/2011] [Indexed: 10/17/2022] Open
Abstract
Francisella tularensis is pathogenic for many mammalian species including humans, causing a spectrum of diseases called tularemia. The highly virulent Type A strains have associated mortality rates of up to 60% if inhaled. An attenuated live vaccine strain (LVS) is the only vaccine to show efficacy in humans, but suffers several barriers to licensure, including the absence of a correlate of protection. An immunoproteomics approach was used to survey the repertoire of antibodies in sera from individuals who had contracted tularemia during two outbreaks and individuals from two geographical areas who had been vaccinated with NDBR Lot 11 or Lot 17 LVS. These data showed a large overlap in the antibodies generated in response to tularemia infection or LVS vaccination. A total of seven proteins were observed to be reactive with 60% or more sera from vaccinees and convalescents. A further four proteins were recognised by 30-60% of the sera screened. These proteins have the potential to serve as markers of vaccination or candidates for subunit vaccines.
Collapse
Affiliation(s)
- Kelly M Fulton
- National Research Council Institute for Biological Sciences, Ottawa, Canada
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Pierson T, Matrakas D, Taylor YU, Manyam G, Morozov VN, Zhou W, van Hoek ML. Proteomic Characterization and Functional Analysis of Outer Membrane Vesicles of Francisella novicida Suggests Possible Role in Virulence and Use as a Vaccine. J Proteome Res 2011; 10:954-67. [DOI: 10.1021/pr1009756] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Tony Pierson
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
| | - Demetrios Matrakas
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
| | - Yuka U. Taylor
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
| | - Ganiraju Manyam
- Department of Bioinformatics & Computational Biology, The UT MD Anderson Cancer Center, Houston, Texas, United States
| | - Victor N. Morozov
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia 20110, United States
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia 20110, United States
| | - Monique L. van Hoek
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia 20110, United States
| |
Collapse
|
24
|
Kilmury SLN, Twine SM. The francisella tularensis proteome and its recognition by antibodies. Front Microbiol 2011; 1:143. [PMID: 21687770 PMCID: PMC3109489 DOI: 10.3389/fmicb.2010.00143] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 12/18/2010] [Indexed: 01/31/2023] Open
Abstract
Francisella tularensis is the causative agent of a spectrum of diseases collectively known as tularemia. The extreme virulence of the pathogen in humans, combined with the low infectious dose and the ease of dissemination by aerosol have led to concerns about its abuse as a bioweapon. Until recently, nothing was known about the virulence mechanisms and even now, there is still a relatively poor understanding of pathogen virulence. Completion of increasing numbers of Francisella genome sequences, combined with comparative genomics and proteomics studies, are contributing to the knowledge in this area. Tularemia may be treated with antibiotics, but there is currently no licensed vaccine. An attenuated strain, the Live Vaccine Strain (LVS) has been used to vaccinate military and at risk laboratory personnel, but safety concerns mean that it is unlikely to be licensed by the FDA for general use. Little is known about the protective immunity induced by vaccination with LVS, in humans or animal models. Immunoproteomics studies with sera from infected humans or vaccinated mouse strains, are being used in gel-based or proteome microarray approaches to give insight into the humoral immune response. In addition, these data have the potential to be exploited in the identification of new diagnostic or protective antigens, the design of next generation live vaccine strains, and the development of subunit vaccines. Herein, we briefly review the current knowledge from Francisella comparative proteomics studies and then focus upon the findings from immunoproteomics approaches.
Collapse
Affiliation(s)
- Sara L. N. Kilmury
- Institute for Biological Sciences, National Research Council CanadaOttawa, ON, Canada
| | - Susan M. Twine
- Institute for Biological Sciences, National Research Council CanadaOttawa, ON, Canada
| |
Collapse
|
25
|
Immunoproteomic analysis of human serological antibody responses to vaccination with whole-cell pertussis vaccine (WCV). PLoS One 2010; 5:e13915. [PMID: 21170113 PMCID: PMC2976700 DOI: 10.1371/journal.pone.0013915] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 10/18/2010] [Indexed: 11/29/2022] Open
Abstract
Background Pertussis (whooping cough) caused by Bordetella pertussis
(B.p), continues to be a serious public health threat.
Vaccination is the most economical and effective strategy for preventing and
controlling pertussis. However, few systematic investigations of actual
human immune responses to pertussis vaccines have been performed. Therefore,
we utilized a combination of two-dimensional electrophoresis (2-DE),
immunoblotting, and mass spectrometry to reveal the entire antigenic
proteome of whole-cell pertussis vaccine (WCV) targeted by the human immune
system as a first step toward evaluating the repertoire of human humoral
immune responses against WCV. Methodology/Principal Findings Immunoproteomic profiling of total membrane enriched proteins and
extracellular proteins of Chinese WCV strain 58003 identified a total of 30
immunoreactive proteins. Seven are known pertussis antigens including
Pertactin, Serum resistance protein, chaperonin GroEL and two OMP porins.
Sixteen have been documented to be immunogenic in other pathogens but not in
B.p, and the immunogenicity of the last seven proteins
was found for the first time. Furthermore, by comparison of the human and
murine immunoproteomes of B.p, with the exception of four
human immunoreactive proteins that were also reactive with mouse immune
sera, a unique group of antigens including more than 20 novel immunoreactive
proteins that uniquely reacted with human immune serum was confirmed. Conclusions/Significance This study is the first time that the repertoire of human serum antibody
responses against WCV was comprehensively investigated, and a small number
of previously unidentified antigens of WCV were also found by means of the
classic immunoproteomic strategy. Further research on these newly identified
predominant antigens of B.p exclusively against humans will
not only remarkably accelerate the development of diagnostic biomarkers and
subunit vaccines but also provide detailed insight into human immunity
mechanisms against WCV. In particular, this work highlights the
heterogeneity of the B.p immunoreactivity patterns of the
mouse model and the human host.
Collapse
|
26
|
Deringer JR, Chen C, Samuel JE, Brown WC. Immunoreactive Coxiella burnetii Nine Mile proteins separated by 2D electrophoresis and identified by tandem mass spectrometry. MICROBIOLOGY-SGM 2010; 157:526-542. [PMID: 21030434 PMCID: PMC3090129 DOI: 10.1099/mic.0.043513-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Coxiella burnetii is a Gram-negative obligate intracellular pathogen and the causative agent of Q fever in humans. Q fever causes acute flu-like symptoms and may develop into a chronic disease leading to endocarditis. Its potential as a bioweapon has led to its classification as a category B select agent. An effective inactivated whole-cell vaccine (WCV) currently exists but causes severe granulomatous/necrotizing reactions in individuals with prior exposure, and is not licensed for use in most countries. Current efforts to reduce or eliminate the deleterious reactions associated with WCVs have focused on identifying potential subunit vaccine candidates. Both humoral and T cell-mediated responses are required for protection in animal models. In this study, nine novel immunogenic C. burnetii proteins were identified in extracted whole-cell lysates using 2D electrophoresis, immunoblotting with immune guinea pig sera, and tandem MS. The immunogenic C. burnetii proteins elicited antigen-specific IgG in guinea pigs vaccinated with whole-cell killed Nine Mile phase I vaccine, suggesting a T cell-dependent response. Eleven additional proteins previously shown to react with immune human sera were also antigenic in guinea pigs, showing the relevance of the guinea pig immunization model for antigen discovery. The antigens described here warrant further investigation to validate their potential use as subunit vaccine candidates.
Collapse
Affiliation(s)
- James R Deringer
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA
| | - Chen Chen
- Department of Microbial and Molecular Pathogenesis, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - James E Samuel
- Department of Microbial and Molecular Pathogenesis, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Wendy C Brown
- School for Global Animal Health, Washington State University, Pullman, WA 99164, USA.,Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
27
|
Balonova L, Hernychova L, Mann BF, Link M, Bilkova Z, Novotny MV, Stulik J. Multimethodological approach to identification of glycoproteins from the proteome of Francisella tularensis, an intracellular microorganism. J Proteome Res 2010; 9:1995-2005. [PMID: 20175567 DOI: 10.1021/pr9011602] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
It appears that most glycoproteins found in pathogenic bacteria are associated with virulence. Despite the recent identification of novel virulence factors, the mechanisms of virulence in Francisella tularensis are poorly understood. In spite of its importance, questions about glycosylation of proteins in this bacterium and its potential connection with bacterial virulence have not been answered yet. In the present study, several putative Francisella tularensis glycoproteins were characterized through the combination of carbohydrate-specific detection and lectin affinity with highly sensitive mass spectrometry utilizing the bottom-up proteomic approach. The protein PilA that was recently found as being possibly glycosylated, as well as other proteins with designation as novel factors of virulence, were among the proteins identified in this study. The reported data compile the list of potential glycoproteins that may serve as a takeoff platform for a further definition of proteins modified by glycans, faciliting a better understanding of the function of protein glycosylation in pathogenicity of Francisella tularensis.
Collapse
Affiliation(s)
- Lucie Balonova
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
28
|
Twine SM, Petit MD, Fulton KM, House RV, Conlan JW. Immunoproteomics analysis of the murine antibody response to vaccination with an improved Francisella tularensis live vaccine strain (LVS). PLoS One 2010; 5:e10000. [PMID: 20368994 PMCID: PMC2848853 DOI: 10.1371/journal.pone.0010000] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 03/05/2010] [Indexed: 11/18/2022] Open
Abstract
Background Francisella tularensis subspecies tularensis is the causative agent of a spectrum of diseases collectively known as tularemia. An attenuated live vaccine strain (LVS) has been shown to be efficacious in humans, but safety concerns have prevented its licensure by the FDA. Recently, F. tularensis LVS has been produced under Current Good Manufacturing Practice (CGMP guidelines). Little is known about the immunogenicity of this new vaccine preparation in comparison with extensive studies conducted with laboratory passaged strains of LVS. Thus, the aim of the current work was to evaluate the repertoire of antibodies produced in mouse strains vaccinated with the new LVS vaccine preparation. Methodology/Principal Findings In the current study, we used an immunoproteomics approach to examine the repertoire of antibodies induced following successful immunization of BALB/c versus unsuccessful vaccination of C57BL/6 mice with the new preparation of F. tularensis LVS. Successful vaccination of BALB/c mice elicited antibodies to nine identified proteins that were not recognized by antisera from vaccinated but unprotected C57BL/6 mice. In addition, the CGMP formulation of LVS stimulated a greater repertoire of antibodies following vaccination compared to vaccination with laboratory passaged ATCC LVS strain. A total of 15 immunoreactive proteins were identified in both studies, however, 16 immunoreactive proteins were uniquely reactive with sera from the new formulation of LVS. Conclusions/Significance This is the first report characterising the antibody based immune response of the new formulation of LVS in the widely used murine model of tularemia. Using two mouse strains, we show that successfully vaccinated mice can be distinguished from unsuccessfully vaccinated mice based upon the repertoire of antibodies generated. This opens the door towards downselection of antigens for incorporation into tularemia subunit vaccines. In addition, this work also highlights differences in the humoral immune response to vaccination with the commonly used laboratory LVS strain and the new vaccine formulation of LVS.
Collapse
Affiliation(s)
- Susan M Twine
- National Research Council Institute for Biological Sciences, Ottawa, Ontario, Canada.
| | | | | | | | | |
Collapse
|
29
|
Gregory SH, Chen WH, Mott S, Palardy JE, Parejo NA, Heninger S, Anderson CA, Artenstein AW, Opal SM, Cross AS. Detoxified endotoxin vaccine (J5dLPS/OMP) protects mice against lethal respiratory challenge with Francisella tularensis SchuS4. Vaccine 2010; 28:2908-15. [PMID: 20170768 DOI: 10.1016/j.vaccine.2010.01.067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 12/23/2009] [Accepted: 01/10/2010] [Indexed: 10/19/2022]
Abstract
Francisella tularensis is a category A select agent. J5dLPS/OMP is a novel vaccine construct consisting of detoxified, O-polysaccharide side chain-deficient, lipopolysaccharide non-covalently complexed with the outer membrane protein of N. meningitidis group B. Immunization elicits high-titer polyclonal antibodies specific for the highly-conserved epitopes expressed within the glycolipid core that constitutes gram-negative bacteria (e.g., F. tularensis). Mice immunized intranasally with J5dLPS/OMP exhibited protective immunity to intratracheal challenge with the live vaccine strain, as well as the highly-virulent SchuS4 strain, of F. tularensis. The efficacy of J5dLPS/OMP vaccine suggests its potential utility in immunizing the general population against several different gram-negative select agents concurrently.
Collapse
Affiliation(s)
- Stephen H Gregory
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, 55 Claverick Street, Providence, RI 02903, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Francisella tularensis is a Category A select agent for which vaccine and countermeasure development are a priority. In the past eight years, renewed interest in this pathogen has led to the generation of an enormous amount of new data on both the pathogen itself and its interaction with host cells. This information has fostered the development of various vaccine candidates including acellular subunit, killed whole cell and live attenuated. This review summarizes the progress and promise of these various candidates.
Collapse
Affiliation(s)
- Eileen M Barry
- University of Maryland School of Medicine, Center for Vaccine Development, Baltimore, MD, USA.
| | | | | |
Collapse
|
31
|
Zhou M, Zhang A, Guo Y, Liao Y, Chen H, Jin M. A comprehensive proteome map of the Haemophilus parasuis serovar 5. Proteomics 2009; 9:2722-39. [PMID: 19405026 DOI: 10.1002/pmic.200800717] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Haemophilus parasuis is the causative agent of Glässer's disease of pigs, a disease associated with fibrinous polyserositis, polyarthritis and meningitis. Systematic reference maps of outer membrane, intracellular and extracellular proteome fractions of the clinical isolate H. parasuis SH0165 were examined by 2-DE coupled with MALDI-TOF MS. A total of 539 proteins spots were successfully identified, corresponding to 317 different proteins that were classified into functional categories. The majority of these proteins were linked to housekeeping functions in amino acid transport and metabolism, secondary metabolites biosynthesis, transport and catabolism and post-translational modification, protein turnover and chaperones. A significant number of outer membrane proteins were identified, such as Wza, Omp2, Omp5, D15 and PalA, which were supposed to play important roles in basic physiology of H. parasuis. In addition, several virulence-associated proteins involved in type I (TolC), type III (DsbA and DsbC) and type V (Autotransporter adhesins) secretion systems, and solute-binding proteins participating in iron-uptake systems were also identified in the present study.
Collapse
Affiliation(s)
- Mingguang Zhou
- Unit of Animal Infectious Diseases, National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, Hubei, P. R. China
| | | | | | | | | | | |
Collapse
|
32
|
Pelletier N, Raoult D, La Scola B. Specific recognition of the major capsid protein of Acanthamoeba polyphaga mimivirus by sera of patients infected by Francisella tularensis. FEMS Microbiol Lett 2009; 297:117-23. [PMID: 19538509 DOI: 10.1111/j.1574-6968.2009.01675.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Francisella tularensis, a Gram-negative cocobacillus responsible for tularemia, especially severe pneumonia, is a facultative intracellular bacterium classified as a biological agent of category A. Acanthamoeba polyphaga mimivirus (APM) is a recently discovered giant virus suspected to be an agent of both community- and hospital-acquired pneumonia. During specificity testing of antibody to APM detection, it was observed that nearly all patients infected by F. tularensis had elevated antibody titers to APM. In the present study, we investigated this cross-reactivity by immunoproteomics. Apart from the detection of antibodies reactive to new immunoreactive proteins in patients infected by F. tularensis, we showed that the sera of those patients recognize specifically two proteins of APM: the capsid protein and another protein of unknown function. No common protein motif can be detected in silico based on genome analysis of the involved protein. Furthermore, this cross-reactivity was confirmed with the recombinant capsid protein expressed in Escherichia coli. This emphasizes the pitfalls of a serological diagnosis of pneumonia.
Collapse
Affiliation(s)
- Nicolas Pelletier
- URMITE, CNRS UMR IRD 6236, IFR 48, Faculté de Médecine, Marseille, France
| | | | | |
Collapse
|
33
|
Francisella tularensis infection-derived monoclonal antibodies provide detection, protection, and therapy. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:414-22. [PMID: 19176692 DOI: 10.1128/cvi.00362-08] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Francisella tularensis is the causative agent of tularemia and a potential agent of biowarfare. As an easily transmissible infectious agent, rapid detection and treatment are necessary to provide a positive clinical outcome. As an agent of biowarfare, there is an additional need to prevent infection. We made monoclonal antibodies to the F. tularensis subsp. holarctica live vaccine strain (F. tularensis LVS) by infecting mice with a sublethal dose of bacteria and, following recovery, by boosting the mice with sonicated organisms. The response to the initial and primary infection was restricted to immunoglobulin M antibody directed solely against lipopolysaccharide (LPS). After boosting with sonicated organisms, the specificity repertoire broadened against protein antigens, including DnaK, LpnA, FopA, bacterioferritin, the 50S ribosomal protein L7/L12, and metabolic enzymes. These monoclonal antibodies detect F. tularensis LVS by routine immunoassays, including enzyme-linked immunosorbent assay, Western blot analysis, and immunofluorescence. The ability of the antibodies to protect mice from intradermal infection, both prophylactically and therapeutically, was examined. An antibody to LPS which provides complete protection from infection with F. tularensis LVS and partial protection from infection with F. tularensis subsp. tularensis strain SchuS4 was identified. There was no bacteremia and reduced organ burden within the first 24 h when mice were protected from F. tularensis LVS infection with the anti-LPS antibody. No antibody that provided complete protection when administered therapeutically was identified; however, passive transfer of antibodies against LPS, FopA, and LpnA resulted in 40 to 50% survival of mice infected with F. tularensis LVS.
Collapse
|
34
|
Barel M, Hovanessian AG, Meibom K, Briand JP, Dupuis M, Charbit A. A novel receptor - ligand pathway for entry of Francisella tularensis in monocyte-like THP-1 cells: interaction between surface nucleolin and bacterial elongation factor Tu. BMC Microbiol 2008; 8:145. [PMID: 18789156 PMCID: PMC2551611 DOI: 10.1186/1471-2180-8-145] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Accepted: 09/12/2008] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Francisella tularensis, the causative agent of tularemia, is one of the most infectious human bacterial pathogens. It is phagocytosed by immune cells, such as monocytes and macrophages. The precise mechanisms that initiate bacterial uptake have not yet been elucidated. Participation of C3, CR3, class A scavenger receptors and mannose receptor in bacterial uptake have been already reported. However, contribution of an additional, as-yet-unidentified receptor for F. tularensis internalization has been suggested. RESULTS We show here that cell-surface expressed nucleolin is a receptor for Francisella tularensis Live Vaccine Strain (LVS) and promotes LVS binding and infection of human monocyte-like THP-1 cells. The HB-19 pseudopeptide that binds specifically carboxy-terminal RGG domain of nucleolin inhibits LVS binding and infection of monocyte-like THP-1 cells. In a pull-down assay, elongation factor Tu (EF-Tu), a GTP-binding protein involved in protein translation, usually found in cytoplasm, was recovered among LVS bacterial membrane proteins bound on RGG domain of nucleolin. A specific polyclonal murine antibody was raised against recombinant LVS EF-Tu. By fluorescence and electron microscopy experiments, we found that a fraction of EF-Tu could be detected at the bacterial surface. Anti-EF-Tu antibodies reduced LVS binding to monocyte-like THP-1 cells and impaired infection, even in absence of complement and complement receptors. Interaction between EF-Tu and nucleolin was illustrated by two different pull-down assays using recombinant EF-Tu proteins and either RGG domain of nucleolin or cell solubilized nucleolin. DISCUSSION Altogether, our results demonstrate that the interaction between surface nucleolin and its bacterial ligand EF-Tu plays an important role in Francisella tularensis adhesion and entry process and may therefore facilitate invasion of host tissues. Since phagosomal escape and intra-cytosolic multiplication of LVS in infected monocytes are very similar to those of human pathogenic F. tularensis ssp tularensis, the mechanism of entry into monocyte-like THP-1 cells, involving interaction between EF-Tu and nucleolin, might be similar in the two subspecies. Thus, the use of either nucleolin-specific pseudopeptide HB-19 or recombinant EF-Tu could provide attractive therapeutic approaches for modulating F. tularensis infection.
Collapse
Affiliation(s)
- Monique Barel
- INSERM U570, Unité de Pathogénie des Infections Systémiques, Université Paris Descartes, Faculté de Médecine Necker Enfants-Malades, 156 rue de Vaugirard, 75730, Paris Cedex 15, France
| | - Ara G Hovanessian
- UPR 2228 CNRS, Régulation de la transcription et maladies génétiques, UFR Biomédicale des Saints-Pères, 45 rue des Saints Pères, 75270, Paris Cedex 06, France
| | - Karin Meibom
- INSERM U570, Unité de Pathogénie des Infections Systémiques, Université Paris Descartes, Faculté de Médecine Necker Enfants-Malades, 156 rue de Vaugirard, 75730, Paris Cedex 15, France
| | - Jean-Paul Briand
- UPR 9021 CNRS, Immunologie et Chimie Thérapeutiques, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, 67084, Strasbourg Cedex, France
| | - Marion Dupuis
- INSERM U570, Unité de Pathogénie des Infections Systémiques, Université Paris Descartes, Faculté de Médecine Necker Enfants-Malades, 156 rue de Vaugirard, 75730, Paris Cedex 15, France
| | - Alain Charbit
- INSERM U570, Unité de Pathogénie des Infections Systémiques, Université Paris Descartes, Faculté de Médecine Necker Enfants-Malades, 156 rue de Vaugirard, 75730, Paris Cedex 15, France
| |
Collapse
|
35
|
Rohmer L, Guina T, Chen J, Gallis B, Taylor GK, Shaffer SA, Miller SI, Brittnacher MJ, Goodlett DR. Determination and Comparison of the Francisella tularensis subsp.novicida U112 Proteome to Other Bacterial Proteomes. J Proteome Res 2008; 7:2016-24. [DOI: 10.1021/pr700760z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Laurence Rohmer
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - Tina Guina
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - Jinzhi Chen
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - Byron Gallis
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - Greg K. Taylor
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - Scott A. Shaffer
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - Samuel I. Miller
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - Mitchell J. Brittnacher
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| | - David R. Goodlett
- Department of Genome Sciences, Microbiology, Medicine, Medicinal Chemistry, and Department of Pediatrics, Division of Infectious Diseases, University of Washington, Seattle, Washington 98195
| |
Collapse
|
36
|
Meibom KL, Dubail I, Dupuis M, Barel M, Lenco J, Stulik J, Golovliov I, Sjöstedt A, Charbit A. The heat-shock protein ClpB of Francisella tularensis is involved in stress tolerance and is required for multiplication in target organs of infected mice. Mol Microbiol 2008; 67:1384-401. [PMID: 18284578 DOI: 10.1111/j.1365-2958.2008.06139.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Intracellular bacterial pathogens generally express chaperones such as Hsp100s during multiplication in host cells, allowing them to survive potentially hostile conditions. Francisella tularensis is a highly infectious bacterium causing the zoonotic disease tularaemia. The ability of F. tularensis to multiply and survive in macrophages is considered essential for its virulence. Although previous mutant screens in Francisella have identified the Hsp100 chaperone ClpB as important for intracellular survival, no detailed study has been performed. We demonstrate here that ClpB of F. tularensis live vaccine strain (LVS) is important for resistance to cellular stress. Promoter analysis shows that the transcriptional start is preceded by a sigma32-like promoter sequence and we demonstrate that expression of clpB is induced by heat shock. This indicates that expression of clpB is dependent on the heat-shock response mediated by sigma32, the only alternative sigma-factor present in Francisella. Our studies demonstrate that ClpB contributes to intracellular multiplication in vitro, but is not essential. However, ClpB is absolutely required for Francisella to replicate in target organs and induce disease in mice. Proteomic analysis of membrane-enriched fractions shows that five proteins are recovered at lower levels in the mutant strain. The crucial role of ClpB for in vivo persistence of Francisella may be linked to its assumed function in reactivation of aggregated proteins under in vivo stress conditions.
Collapse
Affiliation(s)
- Karin L Meibom
- Université Paris Descartes, Faculté de Médecine René Descartes, Paris F-75015, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Thakran S, Li H, Lavine CL, Miller MA, Bina JE, Bina XR, Re F. Identification of Francisella tularensis lipoproteins that stimulate the toll-like receptor (TLR) 2/TLR1 heterodimer. J Biol Chem 2007; 283:3751-60. [PMID: 18079113 DOI: 10.1074/jbc.m706854200] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The innate immune response to Francisella tularensis is primarily mediated by TLR2, though the bacterial products that stimulate this receptor remain unknown. Here we report the identification of two Francisella lipoproteins, TUL4 and FTT1103, which activate TLR2. We demonstrate that TUL4 and FTT1103 stimulate chemokine production in human and mouse cells in a TLR2-dependent way. Using an assay that relies on chimeric TLR proteins, we show that TUL4 and FTT1103 stimulate exclusively the TLR2/TLR1 heterodimer. Our results also show that yet unidentified Francisella proteins, possibly unlipi-dated, have the ability to stimulate the TLR2/TLR6 heterodimer. Through domain-exchange analysis, we determined that an extended region that comprises LRR 9-17 in the extra-cellular portion of TLR1 mediates response to Francisella lipoproteins and triacylated lipopeptide. Substitution of the corresponding LRR of TLR6 with the LRR derived from TLR1 enables TLR6 to recognize TUL4, FTT1103, and triacylated lipopeptide. This study identifies for the first time specific Fran-cisella products capable of stimulating a proinflammatory response and the cellular receptors they trigger.
Collapse
Affiliation(s)
- Shalini Thakran
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Lu Z, Roche MI, Hui JH, Unal B, Felgner PL, Gulati S, Madico G, Sharon J. Generation and characterization of hybridoma antibodies for immunotherapy of tularemia. Immunol Lett 2007; 112:92-103. [PMID: 17764754 PMCID: PMC2128743 DOI: 10.1016/j.imlet.2007.07.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 07/08/2007] [Accepted: 07/09/2007] [Indexed: 12/20/2022]
Abstract
Tularemia is caused by the Gram-negative facultative intracellular bacterium Francisella tularensis, which has been classified as a category A select agent-a likely bioweapon. The high virulence of F. tularensis and the threat of engineered antibiotic resistant variants warrant the development of new therapies to combat this disease. We have characterized 14 anti-Francisella hybridoma antibodies derived from mice infected with F. tularensis live vaccine strain (LVS) for potential use as immunotherapy of tularemia. All 14 antibodies cross-reacted with virulent F. tularensis type A clinical isolates, 8 bound to a purified preparation of LVS LPS, and 6 bound to five protein antigens, identified by proteome microarray analysis. An IgG2a antibody, reactive with the LPS preparation, conferred full protection when administered either systemically or intranasally to BALB/c mice post challenge with a lethal dose of intranasal LVS; three other antibodies prolonged survival. These anti-Francisella hybridoma antibodies could be converted to chimeric versions with mouse V regions and human C regions to serve as components of a recombinant polyclonal antibody for clinical testing as immunotherapy of tularemia. The current study is the first to employ proteome microarrays to identify the target antigens of anti-Francisella monoclonal antibodies and the first to demonstrate the systemic and intranasal efficacy of monoclonal antibodies for post-exposure treatment of respiratory tularemia.
Collapse
MESH Headings
- Administration, Intranasal
- Adoptive Transfer
- Animals
- Antibodies, Bacterial/classification
- Antibodies, Bacterial/immunology
- Antibodies, Bacterial/therapeutic use
- Antibodies, Monoclonal/classification
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antigens, Bacterial/immunology
- Bacterial Vaccines/administration & dosage
- Cell Line, Tumor
- Cross Reactions
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay
- Female
- Francisella tularensis/immunology
- Francisella tularensis/pathogenicity
- Humans
- Hybridomas/immunology
- Hybridomas/microbiology
- Immunization/methods
- Lipopolysaccharides/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Protein Array Analysis
- Tularemia/immunology
- Tularemia/microbiology
- Tularemia/therapy
Collapse
Affiliation(s)
- Zhaohua Lu
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Marly I. Roche
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Julia H. Hui
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Berkay Unal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine School of Medicine, Irvine, CA 92697, USA
| | - Philip L. Felgner
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine School of Medicine, Irvine, CA 92697, USA
| | - Sunita Gulati
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical, Center, Worcester, MA 01605, USA
| | - Guillermo Madico
- Department of Medicine, Boston Medical Center, Boston, MA 02118, USA
| | - Jacqueline Sharon
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- * Corresponding author. Tel.: +1 617 638 4652; fax: +1 617 638 4079. E-mail address: (J. Sharon)
| |
Collapse
|