1
|
Ozanic M, Marecic V, Knezevic M, Kelava I, Stojková P, Lindgren L, Bröms JE, Sjöstedt A, Abu Kwaik Y, Santic M. The type IV pili component PilO is a virulence determinant of Francisella novicida. PLoS One 2022; 17:e0261938. [PMID: 35077486 PMCID: PMC8789160 DOI: 10.1371/journal.pone.0261938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Francisella tularensis is a highly pathogenic intracellular bacterium that causes the disease tularemia. While its ability to replicate within cells has been studied in much detail, the bacterium also encodes a less characterised type 4 pili (T4P) system. T4Ps are dynamic adhesive organelles identified as major virulence determinants in many human pathogens. In F. tularensis, the T4P is required for adherence to the host cell, as well as for protein secretion. Several components, including pilins, a pili peptidase, a secretin pore and two ATPases, are required to assemble a functional T4P, and these are encoded within distinct clusters on the Francisella chromosome. While some of these components have been functionally characterised, the role of PilO, if any, still is unknown. Here, we examined the role of PilO in the pathogenesis of F. novicida. Our results show that the PilO is essential for pilus assembly on the bacterial surface. In addition, PilO is important for adherence of F. novicida to human monocyte-derived macrophages, secretion of effector proteins and intracellular replication. Importantly, the pilO mutant is attenuated for virulence in BALB/c mice regardless of the route of infection. Following intratracheal and intradermal infection, the mutant caused no histopathology changes, and demonstrated impaired phagosomal escape and replication within lung liver as well as spleen. Thus, PilO is an essential virulence determinant of F. novicida.
Collapse
Affiliation(s)
- Mateja Ozanic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Valentina Marecic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Masa Knezevic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Ina Kelava
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Pavla Stojková
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Lena Lindgren
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Jeanette E. Bröms
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Anders Sjöstedt
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology and Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Marina Santic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
2
|
Matz LM, Petrosino JF. A study of innate immune kinetics reveals a role for a chloride transporter in a virulent Francisella tularensis type B strain. Microbiologyopen 2021; 10:e1170. [PMID: 33970545 PMCID: PMC8483402 DOI: 10.1002/mbo3.1170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 11/26/2022] Open
Abstract
Tularemia is a zoonotic disease of global proportions. Francisella tularensis subspecies tularensis (type A) and holarctica (type B) cause disease in healthy humans, with type A infections resulting in higher mortality. Repeated passage of a type B strain in the mid-20th century generated the Live Vaccine Strain (LVS). LVS remains unlicensed, does not protect against high inhalational doses of type A, and its exact mechanisms of attenuation are poorly understood. Recent data suggest that live attenuated vaccines derived from type B may cross-protect against type A. However, there is a dearth of knowledge regarding virulent type B pathogenesis and its capacity to stimulate the host's innate immune response. We therefore sought to increase our understanding of virulent type B in vitro characteristics using strain OR96-0246 as a model. Adding to our knowledge of innate immune kinetics in macrophages following infection with virulent type B, we observed robust replication of strain OR96-0246 in murine and human macrophages, reduced expression of pro-inflammatory cytokine genes from "wild type" type B-infected macrophages compared to LVS, and delayed macrophage cell death suggesting that virulent type B may suppress macrophage activation. One disruption in LVS is in the gene encoding the chloride transporter ClcA. We investigated the role of ClcA in macrophage infection and observed a replication delay in a clcA mutant. Here, we propose its role in acid tolerance. A greater understanding of LVS attenuation may reveal new mechanisms of pathogenesis and inform strategies toward the development of an improved vaccine against tularemia.
Collapse
Affiliation(s)
- Lisa M. Matz
- The Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTXUSA
- The Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTXUSA
- Baylor College of MedicineHoustonTXUSA
| | - Joseph F. Petrosino
- The Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTXUSA
- The Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTXUSA
- Baylor College of MedicineHoustonTXUSA
| |
Collapse
|
3
|
The Comparative Virulence of Francisella tularensis Subsp. mediasiatica for Vaccinated Laboratory Animals. Microorganisms 2020; 8:microorganisms8091403. [PMID: 32932593 PMCID: PMC7564995 DOI: 10.3390/microorganisms8091403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 11/17/2022] Open
Abstract
Tularemia is a severe infectious disease caused by the Gram-negative bacteria Fracisella tularensis. There are four subspecies of F.tularensis: holarctica, tularensis, mediasiatica, and novicida, which differ in their virulence and geographic distribution. One of them, subsp. mediasiatica remains extremely poorly studied, primarily due to the fact that it is found only in the sparsely populated regions of Central Asia and Russia. In particular there is little information in the literature on the virulence and pathogenicity of subsp. mediasiatica. In the present article, we evaluated the comparative virulence of subsp. mediasiatica in vaccinated laboratory animals which we infected with virulent strains: subsp. mediasiatica 678, subsp. holarctica 503, and subsp. tularensis SCHU within 60 to 180 days after vaccination. We found that subsp. mediasiatica is comparable in pathogenicity in mice with subsp. tularensis and in guinea pigs with subsp. holarctica. We also found that the live vaccine does not fully protect mice from subsp. mediasiatica but completely protects guinea pigs for at least six months. In general, our data suggest that subsp. mediasiatica occupies an intermediate position in virulence between spp. tularensis and holarctica.
Collapse
|
4
|
Soucy AM, Hurteau GJ, Metzger DW. Live Vaccination Generates Both Disease Tolerance and Host Resistance During Chronic Pulmonary Infection With Highly Virulent Francisella tularensis SchuS4. J Infect Dis 2019; 218:1802-1812. [PMID: 29931113 DOI: 10.1093/infdis/jiy379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/18/2018] [Indexed: 11/13/2022] Open
Abstract
Disease tolerance can preserve host homeostasis and limit the negative impact of infections. We report that vaccinated mice survived pulmonary challenge with the extremely virulent SchuS4 strain of Francisella tularensis for at least 100 days, despite the persistence of large numbers (~104) of organisms. Transfer of 100 of these resident bacteria to naive animals caused 100% lethality, demonstrating that virulence was maintained. Tissue damage in the lung was limited over the course of infection and was associated with increased levels of amphiregulin. Mice depleted of CD4+ cells had reduced amphiregulin and succumbed to infection. In addition, neutralization of interferon-γ or depletion of CD8+ cells resulted in increased pathogen loads, bacteremia, and death of the host. Conversely, depletion of Ly6G+ neutrophils had no effect on survival and actually resulted in reduced bacterial levels. Understanding the interplay between host resistance and disease tolerance will provide new insights into the understanding of chronic infectious diseases.
Collapse
Affiliation(s)
- Alicia M Soucy
- Department of Immunology and Microbial Disease, Albany Medical College, New York
| | - Gregory J Hurteau
- Department of Immunology and Microbial Disease, Albany Medical College, New York
| | - Dennis W Metzger
- Department of Immunology and Microbial Disease, Albany Medical College, New York
| |
Collapse
|
5
|
Sunagar R, Kumar S, Namjoshi P, Rosa SJ, Hazlett KRO, Gosselin EJ. Evaluation of an outbred mouse model for Francisella tularensis vaccine development and testing. PLoS One 2018; 13:e0207587. [PMID: 30533047 PMCID: PMC6289435 DOI: 10.1371/journal.pone.0207587] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/03/2018] [Indexed: 01/06/2023] Open
Abstract
Francisella tularensis (Ft) is a biothreat agent for which there is no FDA-approved human vaccine. Currently, there are substantial efforts underway to develop both vaccines and the tools to assess these vaccines. Tularemia laboratory research has historically relied primarily upon a small number of inbred mouse strains, but the utility of such findings to outbred animals may be limited. Specifically, C57BL/6 mice are more susceptible than BALB/c mice to Ft infection and less easily protected against challenge with highly virulent type A Ft. Thus, depending on the inbred mouse strain used, one could be misled as to which immunogen(s)/vaccine will ultimately be effective in an outbred human population. Accordingly, we evaluated an outbred Swiss Webster (SW) mouse model in direct comparison to a well-established, inbred C57BL/6 mouse model. Mucosal vaccination with the live, attenuated Ft LVS superoxide dismutase (sodB) mutant demonstrated significantly higher protection in outbred SW mice compared to inbred C57BL/6 mice against Ft SchuS4 respiratory challenge. The protection observed in vaccinated outbred mice correlated with lower bacterial density, reduced tissue inflammation, and reduced levels of pro-inflammatory cytokine production. This protection was CD4+ and CD8+ T cell-dependent and characterized by lower titers of serum antibody (Ab) that qualitatively differed from vaccinated inbred mice. Enhanced protection of vaccinated outbred mice correlated with early and robust production of IFN-γ and IL-17A. Neutralizing Ab administered at the time of challenge revealed that IFN-γ was central to this protection, while IL-17A neutralization did not alter bacterial burden or survival. The present study demonstrates the utility of the outbred mouse as an alternative vaccination model for testing tularemia vaccines. Given the limited MHC repertoire in inbred mice, this outbred model is more analogous to the human in terms of immunological diversity.
Collapse
Affiliation(s)
- Raju Sunagar
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, United States of America
| | - Sudeep Kumar
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, United States of America
| | - Prachi Namjoshi
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, United States of America
| | - Sarah J. Rosa
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, United States of America
| | - Karsten R. O. Hazlett
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, United States of America
| | - Edmund J. Gosselin
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, United States of America
- * E-mail:
| |
Collapse
|
6
|
Sunagar R, Kumar S, Rosa SJ, Hazlett KRO, Gosselin EJ. Differential In Vitro Cultivation of Francisella tularensis Influences Live Vaccine Protective Efficacy by Altering the Immune Response. Front Immunol 2018; 9:1594. [PMID: 30042767 PMCID: PMC6048226 DOI: 10.3389/fimmu.2018.01594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/27/2018] [Indexed: 11/13/2022] Open
Abstract
Francisella tularensis (Ft) is a biothreat agent for which there is no FDA-approved human vaccine. Currently, there are substantial efforts underway to develop both vaccines and improved tools to assess these vaccines. Ft expresses distinct sets of antigens (Ags) in vivo as compared to those expressed in vitro. Importantly, Ft grown in brain-heart infusion medium (BHIM) more closely mimics the antigenic profile of macrophage-grown Ft when compared to Mueller-Hinton medium (MHM)-grown Ft. Thus, we predicted that when used as a live vaccine BHIM-grown Ft (BHIM-Ft) would provide better protection, as compared to MHM-Ft. We first determined if there was a difference in growth kinetics between BHIM and MHM-Ft. We found that BHIM-Ft exhibited an initial growth advantage ex vivo that manifests as slightly hastened intracellular replication as compared to MHM-Ft. We also observed that BHIM-Ft exhibited an initial growth advantage in vivo represented by rapid bacterial expansion and systemic dissemination associated with a slightly shorter mean survival time of naive animals. Next, using two distinct strains of Ft LVS (WT and sodB), we observed that mice vaccinated with live BHIM-Ft LVS exhibited significantly better protection against Ft SchuS4 respiratory challenge compared to MHM-Ft-immunized mice. This enhanced protection correlated with lower bacterial burden, reduced tissue inflammation, and reduced pro-inflammatory cytokine production late in infection. Splenocytes from BHIM-Ft sodB-immunized mice contained more CD4+, effector, memory T-cells, and were more effective at limiting intracellular replication of Ft LVS in vitro. Concurrent with enhanced killing of Ft LVS, BHIM-Ft sodB-immune splenocytes produced significantly higher levels of IFN-γ and IL-17A cytokines than their MHM-Ft sodB-immunized counterparts indicating development of a more effective T cell memory response when immunizing mice with BHIM-Ft.
Collapse
Affiliation(s)
- Raju Sunagar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Sarah J Rosa
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Karsten R O Hazlett
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Edmund J Gosselin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
Collapse
|
7
|
Roberts LM, Powell DA, Frelinger JA. Adaptive Immunity to Francisella tularensis and Considerations for Vaccine Development. Front Cell Infect Microbiol 2018; 8:115. [PMID: 29682484 PMCID: PMC5898179 DOI: 10.3389/fcimb.2018.00115] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/23/2018] [Indexed: 11/13/2022] Open
Abstract
Francisella tularensis is an intracellular bacterium that causes the disease tularemia. There are several subspecies of F. tularensis whose ability to cause disease varies in humans. The most virulent subspecies, tularensis, is a Tier One Select Agent and a potential bioweapon. Although considerable effort has made to generate efficacious tularemia vaccines, to date none have been licensed for use in the United States. Despite the lack of a tularemia vaccine, we have learned a great deal about the adaptive immune response the underlies protective immunity. Herein, we detail the animal models commonly used to study tularemia and their recapitulation of human disease, the field's current understanding of vaccine-mediated protection, and discuss the challenges associated with new vaccine development.
Collapse
Affiliation(s)
- Lydia M Roberts
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, MT, United States
| | - Daniel A Powell
- Department of Immunobiology and Valley Fever Center for Excellence, University of Arizona, Tucson, AZ, United States
| | - Jeffrey A Frelinger
- Department of Immunobiology and Valley Fever Center for Excellence, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
8
|
Powell DA, Frelinger JA. Efficacy of Resistance to Francisella Imparted by ITY/NRAMP/SLC11A1 Depends on Route of Infection. Front Immunol 2017; 8:206. [PMID: 28360906 PMCID: PMC5350118 DOI: 10.3389/fimmu.2017.00206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/15/2017] [Indexed: 11/13/2022] Open
Abstract
Natural resistance-associated macrophage protein (NRAMP) encoded by the Slc11a1 gene is a membrane-associated transporter of divalent metal ions. Murine Slc11a1 has two known alleles, a functional Slc11a1Gly169, which is found in DBA2/J, NOD/LtJ, and 129p3/J and related mouse strains, and a non-functional Slc11a1Asp169, that is found in C56Bl/6J (B6) and BALB/cJ mice. B6 mice congenic for Slc11a1Gly169 (B6-Slc11a1G169) are markedly resistant to the intracellular pathogens Salmonella, Leishmania, and Mycobacterium tuberculosis. We examined the host cell response and replication of Francisella in B6-Slc11a1G169 mice. Bone marrow-derived macrophages from either B6-Slc11a1G169 or B6 mice were both effectively invaded by Francisella live vaccine strain (LVS). However, at 16 hours post-infection (hpi), the number of LVS bacteria recovered from B6 macrophages had increased roughly 100-fold, while in B6-Slc11a1G169 mice the number decreased 10-fold. When the mice were challenged intranasally (i.n.) B6 mice lost significant amounts (~15%) of weight, where as B6-Slc11a1G169 mice lost no weight. Three days after infection in B6-Slc11a1G169 mice, we failed to recover viable Francisella from the lungs, livers, or spleens. By contrast, B6 mice had bacterial burdens approaching 1 × 106 CFU/organ in all three organs. To further examine the degree of resistance imparted by Slc11a1Gly169 expression, we challenged mice deficient in TLR2, TLR4, and TLR9, but expressing the functional Slc11a1 (B6-Slc11a1G169Tlr2/4/9−/−). Surprisingly, B6-Slc11a1G169Tlr2/4/9−/− mice had no notable weight loss. Eighty percent of B6-Slc11a1G169Tlr2/4/9−/− mice yielded no detectable Francisella in any organ tested. Additionally, Slc11a1G169 produced little detectable cytokine either in the lung or serum compared to B6 mice. Mice expressing Slc11a1Gly169 survived even high doses (~80 LD50) of LVS inoculation. These data taken together serve to highlight that functional Slc11a1Gly169 can compensate the lack of TLR2/4/9. Thus Slc11a1 is a critical player in murine resistance to pulmonary Francisella infection, but not footpad infection.
Collapse
Affiliation(s)
- Daniel A Powell
- Department of Immunobiology, University of Arizona , Tucson, AZ , USA
| | | |
Collapse
|
9
|
Hutt JA, Lovchik JA, Dekonenko A, Hahn AC, Wu TH. The Natural History of Pneumonic Tularemia in Female Fischer 344 Rats after Inhalational Exposure to Aerosolized Francisella tularensis Subspecies tularensis Strain SCHU S4. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:252-267. [PMID: 27939130 DOI: 10.1016/j.ajpath.2016.09.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/02/2016] [Accepted: 09/22/2016] [Indexed: 12/11/2022]
Abstract
The inbred Fischer 344 rat is being evaluated for testing novel vaccines and therapeutics against pneumonic tularemia. Although primary pneumonic tularemia in humans typically occurs by inhalation of aerosolized bacteria, the rat model has relied on intratracheal inoculation of organisms because of safety and equipment issues. We now report the natural history of pneumonic tularemia in female Fischer 344 rats after nose-only inhalational exposure to lethal doses of aerosolized Francisella tularensis subspecies tularensis, strain SCHU S4. Our results are consistent with initial uptake of aerosolized SCHU S4 from the nasal cavity, lungs, and possibly the gastrointestinal tract. Bacteremia with hematogenous dissemination was first detected 2 days after exposure. Shortly thereafter, the infected rats exhibited fever, tachypnea, and hypertension that persisted for 24 to 36 hours and then rapidly decreased as animals succumbed to infection between days 5 and 8 after exposure. Tachycardia was observed briefly, but only after the core body temperature and blood pressure began to decrease as the animals were near death. Initial neutrophilic and histiocytic inflammation in affected tissues became progressively more fibrinous and necrotizing over time. At death, as many as 1010 colony-forming units were found in the lungs, spleen, and liver. Death was attributed to sepsis and disseminated intravascular coagulation. Overall, the pathogenesis of pneumonic tularemia in the female F344 rat model appears to replicate the disease in humans.
Collapse
Affiliation(s)
- Julie A Hutt
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico.
| | - Julie A Lovchik
- Center for Infectious Disease and Immunity, The University of New Mexico Health Science Center, Albuquerque, New Mexico; Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, New Mexico
| | - Alexander Dekonenko
- Center for Infectious Disease and Immunity, The University of New Mexico Health Science Center, Albuquerque, New Mexico
| | - Andrew C Hahn
- Center for Infectious Disease and Immunity, The University of New Mexico Health Science Center, Albuquerque, New Mexico
| | - Terry H Wu
- Center for Infectious Disease and Immunity, The University of New Mexico Health Science Center, Albuquerque, New Mexico; Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, New Mexico
| |
Collapse
|
10
|
Ozanic M, Gobin I, Brezovec M, Marecic V, Trobonjaca Z, Abu Kwaik Y, Santic M. F. novicida-Infected A. castellanii Does Not Enhance Bacterial Virulence in Mice. Front Cell Infect Microbiol 2016; 6:56. [PMID: 27242974 PMCID: PMC4870235 DOI: 10.3389/fcimb.2016.00056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/03/2016] [Indexed: 01/02/2023] Open
Abstract
Francisella tularensis is a facultative intracellular bacterium that causes tularemia in humans and animals. Epidemiology of tularemia worldwide is often associated with water-borne transmission, which includes mosquitoes and amoebae as the potential host reservoirs of the bacteria in water environment. In vitro studies showed intracellular replication of F. tularensis within Acanthamoeba castellanii and Hartmanella vermiformis cells. While infection of amoeba by Legionella pneumophila has been shown to enhance infectivity of L. pneumophila the role of F. tularensis-infected protozoa in the pathogenesis of tularemia is not known. We used 6 h coculture of A. castellanii and F. novicida for investigation of the effect of inhaled amoeba on the pathogenesis of tularemia on in vivo model. Balb/c mice were infected intratracheally with F. novicida or with F. novicida-infected A. castellanii. Surprisingly, infection with F. novicida-infected A. castellanii did not lead to bronchopneumonia in Balb/c mice, and Francisella did not disseminate into the liver and spleen. Upon inhalation, F. novicida infects a variety of host cells, though neutrophils are the predominant cells early during infection in the lung infiltrates of pulmonary tularemia. The numbers of neutrophils in the lungs of Balb/c mice were significantly lower in the infection of mice with F. novicida-infected A. castellanii in comparison to group of mice infected only with F. novicida. These results demonstrate that following inoculation of mice with F. novicida-infected A. castellanii, mice did not develop tularemia.
Collapse
Affiliation(s)
- Mateja Ozanic
- Department of Microbiology and Parasitology, Faculty of Medicine, University of RijekaRijeka, Croatia
| | - Ivana Gobin
- Department of Microbiology and Parasitology, Faculty of Medicine, University of RijekaRijeka, Croatia
| | - Martin Brezovec
- Department of Microbiology and Parasitology, Faculty of Medicine, University of RijekaRijeka, Croatia
| | - Valentina Marecic
- Department of Microbiology and Parasitology, Faculty of Medicine, University of RijekaRijeka, Croatia
| | - Zlatko Trobonjaca
- Department of Physiology and Immunology, Faculty of Medicine, University of RijekaRijeka, Croatia
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology and Center for Predictive Medicine, College of Medicine, University of LouisvilleLouisville, KY, USA
| | - Marina Santic
- Department of Microbiology and Parasitology, Faculty of Medicine, University of RijekaRijeka, Croatia
| |
Collapse
|
11
|
Successful protection against tularemia in C57BL/6 mice is correlated with expansion of Francisella tularensis-specific effector T cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 22:119-28. [PMID: 25410207 DOI: 10.1128/cvi.00648-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Francisella tularensis is an intracellular, Gram-negative bacterium that causes the fatal disease tularemia. Currently, there are no licensed vaccines for tularemia and the requirements for protection against infection are poorly defined. To identify correlates of vaccine-induced immunity against tularemia, we compared different strains of the live vaccine strain (LVS) for their relative levels of virulence and ability to protect C57BL/6 mice against challenge with virulent F. tularensis strain SchuS4. Successful vaccination, as defined by survival of C57BL/6 mice, was correlated with significantly greater numbers of effector T cells in the spleen and lung. Further, lung cells and splenocytes from fully protected animals were more effective than lung cells and splenocytes from vaccinated but nonimmune animals in limiting intracellular replication of SchuS4 in vitro. Together, our data provide a unique model to compare efficacious vaccines to nonefficacious vaccines, which will enable comprehensive identification of host and bacterial components required for immunization against tularemia.
Collapse
|
12
|
De Pascalis R, Chou AY, Ryden P, Kennett NJ, Sjöstedt A, Elkins KL. Models derived from in vitro analyses of spleen, liver, and lung leukocyte functions predict vaccine efficacy against the Francisella tularensis Live Vaccine Strain (LVS). mBio 2014; 5:e00936. [PMID: 24713322 PMCID: PMC3993856 DOI: 10.1128/mbio.00936-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 03/06/2014] [Indexed: 12/11/2022] Open
Abstract
Currently, there are no licensed vaccines and no correlates of protection against Francisella tularensis, which causes tularemia. We recently demonstrated that measuring in vitro control of intramacrophage bacterial growth by murine F. tularensis-immune splenocytes, as well as transcriptional analyses, discriminated Francisella vaccines of different efficacies. Further, we identified potential correlates of protection against systemic challenge. Here, we extended this approach by studying leukocytes derived from lungs and livers of mice immunized by parenteral and respiratory routes with F. tularensis vaccines. Liver and lung leukocytes derived from intradermally and intranasally vaccinated mice controlled in vitro Francisella Live Vaccine Strain (LVS) intramacrophage replication in patterns similar to those of splenocytes. Gene expression analyses of potential correlates also revealed similar patterns in liver cells and splenocytes. In some cases (e.g., tumor necrosis factor alpha [TNF-α], interleukin 22 [IL-22], and granulocyte-macrophage colony-stimulating factor [GM-CSF]), liver cells exhibited even higher relative gene expression, whereas fewer genes exhibited differential expression in lung cells. In contrast with their strong ability to control LVS replication, splenocytes from intranasally vaccinated mice expressed few genes with a hierarchy of expression similar to that of splenocytes from intradermally vaccinated mice. Thus, the relative levels of gene expression vary between cell types from different organs and by vaccination route. Most importantly, because studies comparing cell sources and routes of vaccination supported the predictive validity of this coculture and gene quantification approach, we combined in vitro LVS replication with gene expression data to develop analytical models that discriminated between vaccine groups and successfully predicted the degree of vaccine efficacy. Thus, this strategy remains a promising means of identifying and quantifying correlative T cell responses. IMPORTANCE Identifying and quantifying correlates of protection is especially challenging for intracellular bacteria, including Francisella tularensis. F. tularensis is classified as a category A bioterrorism agent, and no vaccines have been licensed in the United States, but tularemia is a rare disease. Therefore, clinical trials to test promising vaccines are impractical. In this report, we further evaluated a novel approach to developing correlates by assessing T cell immune responses in lungs and livers of differentially vaccinated mice; these nonprofessional immune tissues are colonized by Francisella. The relative degree of vaccine efficacy against systemic challenge was reflected by the ability of immune T cells, particularly liver T cells, to control the intramacrophage replication of bacteria in vitro and by relative gene expression of several immunological mediators. We therefore developed analytical models that combined bacterial replication data and gene expression data. Several resulting models provided excellent discrimination between vaccines of different efficacies.
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, Bethesda, Maryland, USA
| | - 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, Bethesda, Maryland, USA
| | - Patrik Ryden
- Department of Mathematics and Mathematical Statistics, Umeå University, Umeå, Sweden
| | - Nikki J. Kennett
- 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, Bethesda, Maryland, USA
| | - Anders Sjöstedt
- Department of Clinical Microbiology, Clinical Bacteriology, and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - 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, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Cyclic di-GMP stimulates biofilm formation and inhibits virulence of Francisella novicida. Infect Immun 2012; 80:4239-47. [PMID: 22988021 DOI: 10.1128/iai.00702-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis is a gram-negative bacterium that is highly virulent in humans, causing the disease tularemia. F. novicida is closely related to F. tularensis and exhibits high virulence in mice, but it is avirulent in healthy humans. An F. novicida-specific gene cluster (FTN0451 to FTN0456) encodes two proteins with diguanylate cyclase (DGC) and phosphodiesterase (PDE) domains that modulate the synthesis and degradation of cyclic di-GMP (cdGMP). No DGC- or PDE-encoding protein genes are present in the F. tularensis genome. F. novicida strains lacking either the two DGC/PDE genes (cdgA and cdgB) or the entire gene cluster (strain KKF457) are defective for biofilm formation. In addition, expression of CdgB or a heterologous DGC in strain KKF457 stimulated F. novicida biofilms, even in a strain lacking the biofilm regulator QseB. Genetic evidence suggests that CdgA is predominantly a PDE, while CdgB is predominantly a DGC. The F. novicida qseB strain showed reduced cdgA and cdgB transcript levels, demonstrating an F. novicida biofilm signaling cascade that controls cdGMP levels. Interestingly, KKF457 with elevated cdGMP levels exhibited a decrease in intramacrophage replication and virulence in mice, as well as increased growth yields and biofilm formation in vitro. Microarray analyses revealed that cdGMP stimulated the transcription of a chitinase (ChiB) known to contribute to biofilm formation. Our results indicate that elevated cdGMP in F. novicida stimulates biofilm formation and inhibits virulence. We suggest that differences in human virulence between F. novicida and F. tularensis may be due in part to the absence of cdGMP signaling in F. tularensis.
Collapse
|
14
|
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: 3.6] [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
|
15
|
Protective immunity against tularemia provided by an adenovirus-vectored vaccine expressing Tul4 of Francisella tularensis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:359-64. [PMID: 22278325 DOI: 10.1128/cvi.05384-11] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Francisella tularensis, a category A bioterrorism agent, is a highly infectious organism that is passed on via skin contact and inhalation routes. A live attenuated vaccine strain (LVS) has been developed, but it has not been licensed for public use by the FDA due to safety concerns. Thus, there exists a need for a safer and improved vaccine. In this study, we have constructed a replication-incompetent adenovirus, Ad/opt-Tul4, carrying a codon-optimized gene for expression of a membrane protein, Tul4, of F. tularensis LVS. Its ability to protect against lethal challenge and its immunogenicity were evaluated in a murine model. An intramuscular injection of a single dose (1 × 10(7) PFU) of Ad/opt-Tul4 elicited a robust Tul4-specific antibody response. Assays suggest a Th1-driven response. A single dose elicited 20% protection against challenge with 100 × 50% lethal dose (LD(50)) F. tularensis LVS; two additional booster shots resulted in 60% protection. In comparison, three doses of 5 μg recombinant Tul4 protein did not elicit significant protection against challenge. Therefore, the Ad/opt-Tul4 vaccine was more effective than the protein vaccine, and protection was dose dependent. Compared to LVS, the protection rate is lower, but an adenovirus-vectored vaccine may be more attractive due to its enhanced safety profile and mucosal route of delivery. Furthermore, simple genetic modification of the vaccine may potentially produce antibodies protective against a fully virulent strain of F. tularensis. Our data support the development and further research of an adenovirus-vectored vaccine against Tul4 of F. tularensis LVS.
Collapse
|
16
|
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: 3.9] [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
|
17
|
Atianand MK, Duffy EB, Shah A, Kar S, Malik M, Harton JA. Francisella tularensis reveals a disparity between human and mouse NLRP3 inflammasome activation. J Biol Chem 2011; 286:39033-42. [PMID: 21930705 PMCID: PMC3234728 DOI: 10.1074/jbc.m111.244079] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 08/29/2011] [Indexed: 11/06/2022] Open
Abstract
Pathogen-triggered activation of the inflammasome complex leading to caspase-1 activation and IL-1β production involves similar sensor proteins between mouse and human. However, the specific sensors used may differ between infectious agents and host species. In mice, Francisella infection leads to seemingly exclusive activation of the Aim2 inflammasome with no apparent role for Nlrp3. Here we examine the IL-1β response of human cells to Francisella infection. Francisella strains exhibit differences in IL-1β production by influencing induction of IL-1β and ASC transcripts. Unexpectedly, our results demonstrate that Francisella activates the NLRP3 inflammasome in human cells. Francisella infection of THP-1 cells elicits IL-1β production, which is reduced by siRNA targeting of NLRP3. Moreover, in reconstituted 293T cells, Francisella triggers assembly of the NLRP3 inflammasome complex. In addition, inhibitors of reactive oxygen species, cathepsin B, and K(+) efflux pathways, known to specifically influence NLRP3, substantially but not completely impair the Francisella-elicited IL-1β response, suggesting the involvement of another inflammasome pathway. Finally, shRNA targeting of NLRP3 and AIM2 reveals that both pathways contribute to the inflammasome response. Together these results establish NLRP3 as a cytosolic sensor for Francisella in human cells, a role not observed in mouse.
Collapse
Affiliation(s)
- Maninjay K. Atianand
- From the Center for Immunology and Microbial Disease, Albany Medical College and
| | - Ellen B. Duffy
- From the Center for Immunology and Microbial Disease, Albany Medical College and
| | - Aaloki Shah
- From the Center for Immunology and Microbial Disease, Albany Medical College and
| | - Supriya Kar
- From the Center for Immunology and Microbial Disease, Albany Medical College and
| | - Meenakshi Malik
- From the Center for Immunology and Microbial Disease, Albany Medical College and
- the Department of Arts and Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York 12208
| | - Jonathan A. Harton
- From the Center for Immunology and Microbial Disease, Albany Medical College and
| |
Collapse
|
18
|
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: 4.3] [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
|
19
|
Conlan JW, Chen W, Bosio CM, Cowley SC, Elkins KL. Infection of mice with Francisella as an immunological model. CURRENT PROTOCOLS IN IMMUNOLOGY 2011; Chapter 19:Unit 19.14. [PMID: 21462168 PMCID: PMC3405980 DOI: 10.1002/0471142735.im1914s93] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This unit describes the utility of various mouse models of infection for studying pathogenesis and adaptive immune responses to the facultative intracellular bacteria pathogen Francisella tularensis. By judicious use of different combinations of mouse and bacterial strains, as well as different routes of infection, murine tularemia models may be used to explore a complete picture of F. tularensis infection and immunity. Moreover, studies using Francisella, particularly the Live Vaccine Strain (LVS), serve as a convenient and tractable model system that appears to be representative of mammalian host responses to intracellular pathogens in general.
Collapse
Affiliation(s)
- J Wayne Conlan
- National Research Council of Canada, Institute for Biological Sciences, Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
20
|
Directed screen of Francisella novicida virulence determinants using Drosophila melanogaster. Infect Immun 2010; 78:3118-28. [PMID: 20479082 DOI: 10.1128/iai.00146-10] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Francisella tularensis is a highly virulent, facultative intracellular human pathogen whose virulence mechanisms are not well understood. Occasional outbreaks of tularemia and the potential use of F. tularensis as a bioterrorist agent warrant better knowledge about the pathogenicity of this bacterium. Thus far, genome-wide in vivo screens for virulence factors have been performed in mice, all however restricted by the necessity to apply competition-based, negative-selection assays. We wanted to individually evaluate putative virulence determinants suggested by such assays and performed directed screening of 249 F. novicida transposon insertion mutants by using survival of infected fruit flies as a measure of bacterial virulence. Some 20% of the genes tested were required for normal virulence in flies; most of these had not previously been investigated in detail in vitro or in vivo. We further characterized their involvement in bacterial proliferation and pathogenicity in flies and in mouse macrophages. Hierarchical cluster analysis of mutant phenotypes indicated a functional linkage between clustered genes. One cluster grouped all but four genes of the Francisella pathogenicity island and other loci required for intracellular survival. We also identified genes involved in adaptation to oxidative stress and genes which might induce host energy wasting. Several genes related to type IV pilus formation demonstrated hypervirulent mutant phenotypes. Collectively, the data demonstrate that the bacteria in part use similar virulence mechanisms in mammals as in Drosophila melanogaster but that a considerable proportion of the virulence factors active in mammals are dispensable for pathogenicity in the insect model.
Collapse
|
21
|
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: 24] [Impact Index Per Article: 1.7] [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
|
22
|
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
|
23
|
Conlan JW, Shen H, Golovliov I, Zingmark C, Oyston PCF, Chen W, House RV, Sjöstedt A. Differential ability of novel attenuated targeted deletion mutants of Francisella tularensis subspecies tularensis strain SCHU S4 to protect mice against aerosol challenge with virulent bacteria: effects of host background and route of immunization. Vaccine 2009; 28:1824-31. [PMID: 20018266 DOI: 10.1016/j.vaccine.2009.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 11/19/2009] [Accepted: 12/01/2009] [Indexed: 12/25/2022]
Abstract
Francisella tularensis subspecies tularensis is a highly virulent facultative intracellular pathogen of humans and a potential biological weapon. A live vaccine strain, F. tularensis LVS, was developed more than 50 years ago by pragmatic attenuation of a strain of the less virulent holarctica subspecies. LVS was demonstrated to be highly effective in human volunteers who were exposed to intradermal challenge with fully virulent subsp. tularensis, but was less effective against aerosol exposure. LVS faces regulatory hurdles that to date have prevented its licensure for general use. Therefore, a better defined and more effective vaccine is being sought. To this end we have created gene deletion mutants in the virulent subsp. tularensis strain and tested them for their ability to elicit a protective immune response against systemic or aerosol challenge with the highly virulent wild-type subsp. tularensis strain, SCHU S4. Both oral and intradermal (ID) primary vaccination routes were assessed in BALB/c and C3H/HeN mice as was oral boosting. One SCHU S4 mutant missing the heat shock gene, clpB, was significantly more attenuated than LVS whereas a double deletion mutant missing genes FTT0918 and capB was as attenuated as LVS. In general mice immunized with SCHU S4DeltaclpB were significantly better protected against aerosol challenge than mice immunized with LVS. A single ID immunization of BALB/c mice with SCHU S4DeltaclpB was at least as effective as any other regimen examined. Mice immunized with SCHU S4Delta0918DeltacapB were generally protected to a similar degree as mice immunized with LVS. A preliminary examination of immune responses to vaccination with LVS, SCHU S4DeltaclpB, or SCHU S4Delta0918DeltacapB provided no obvious correlate to their relative efficacies.
Collapse
Affiliation(s)
- J Wayne Conlan
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ontario K1A 0R6, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Vaccination with a defined Francisella tularensis subsp. novicida pathogenicity island mutant (DeltaiglB) induces protective immunity against homotypic and heterotypic challenge. Vaccine 2009; 27:5554-61. [PMID: 19651173 DOI: 10.1016/j.vaccine.2009.07.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 06/25/2009] [Accepted: 07/14/2009] [Indexed: 11/21/2022]
Abstract
Francisella tularensis, an intracellular Gram-negative bacterium, is the causative agent of tularemia and a potential bioweapon. Currently, there is no licensed vaccine against this organism. We have characterized the efficacy of a defined F. tularensis subsp. novicida mutant (DeltaiglB) as a live attenuated vaccine against pneumonic tularemia. Replication of the iglB mutant (KKF235) in murine macrophages was significantly lower than the wild type novicida strain U112, and exhibited an LD(50) greater than 10(6)-fold (>10(7)CFU vs <10CFU) in an intranasal challenge model. Mice immunized with KKF235 intranasally or orally induced robust antigen-specific splenic IFN-gamma recall responses, as well as the production of systemic and mucosal antibodies. Intranasal vaccination with KKF235 protected mice from subsequent homotypic challenge with U112 as well as heterotypic challenge with F. tularensis subsp. holarctica (LVS). Moreover, protected animals also exhibited minimal pathological changes compared with mock-vaccinated and challenged animals. The protection conferred by KKF235 vaccination was shown to be highly dependent on endogenous IFN-gamma production. Most significantly, oral immunization with KKF235 protected mice from a highly lethal subsp. tularensis (SCHU S4) pulmonary challenge. Collectively, these results further suggest the feasibility of using defined pathogenicity island mutants as live vaccine candidates against pneumonic tularemia.
Collapse
|
25
|
Wu TH, Zsemlye JL, Statom GL, Hutt JA, Schrader RM, Scrymgeour AA, Lyons CR. Vaccination of Fischer 344 rats against pulmonary infections by Francisella tularensis type A strains. Vaccine 2009; 27:4684-93. [PMID: 19520198 DOI: 10.1016/j.vaccine.2009.05.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 05/07/2009] [Accepted: 05/21/2009] [Indexed: 12/31/2022]
Abstract
Pneumonic tularemia caused by inhalation of the type A strains of Francisella tularensis is associated with high morbidity and mortality in humans. The only vaccine known to protect humans against this disease is the attenuated live vaccine strain (LVS), but it is not currently registered for human use. To develop a new generation of vaccines, multiple animal models are needed that reproduce the human response to F. tularensis infection and vaccination. We examined the potential use of Fischer 344 rat as such a model. Fischer 344 rats were very sensitive to intratracheal infection with the virulent type A strain SCHU S4 and generally succumbed less than 2 weeks after infection. Similar to humans and non-human primates, Fischer 344 rats vaccinated with LVS by subcutaneous or intradermal routes were protected against a greater range of respiratory SCHU S4 challenge doses than has been reported for LVS vaccinated mice. Intratracheal LVS vaccination also induced effective immunity, but it was less protective when the challenge dose exceeded 10(5) SCHU S4. LVS vaccination did not prevent SCHU S4 infection but rather controlled bacterial growth and pathology, leading to the eventual clearance of the bacteria. Our results suggest that the Fischer 344 rat may be a good model for studying pneumonic tularemia and evaluating potential vaccine candidates.
Collapse
Affiliation(s)
- Terry H Wu
- Center for Infectious Disease & Immunity, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Identification of genes contributing to the virulence of Francisella tularensis SCHU S4 in a mouse intradermal infection model. PLoS One 2009; 4:e5463. [PMID: 19424499 PMCID: PMC2675058 DOI: 10.1371/journal.pone.0005463] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 04/15/2009] [Indexed: 12/14/2022] Open
Abstract
Background Francisella tularensis is a highly virulent human pathogen. The most virulent strains belong to subspecies tularensis and these strains cause a sometimes fatal disease. Despite an intense recent research effort, there is very limited information available that explains the unique features of subspecies tularensis strains that distinguish them from other F. tularensis strains and that explain their high virulence. Here we report the use of targeted mutagenesis to investigate the roles of various genes or pathways for the virulence of strain SCHU S4, the type strain of subspecies tularensis. Methodology/Principal Findings The virulence of SCHU S4 mutants was assessed by following the outcome of infection after intradermal administration of graded doses of bacteria. By this route, the LD50 of the SCHU S4 strain is one CFU. The virulence of 20 in-frame deletion mutants and 37 transposon mutants was assessed. A majority of the mutants did not show increased prolonged time to death, among them notably ΔpyrB and ΔrecA. Of the remaining, mutations in six unique targets, tolC, rep, FTT0609, FTT1149c, ahpC, and hfq resulted in significantly prolonged time to death and mutations in nine targets, rplA, wbtI, iglB, iglD, purL, purF, ggt, kdtA, and glpX, led to marked attenuation with an LD50 of >103 CFU. In fact, the latter seven mutants showed very marked attenuation with an LD50 of ≥107 CFU. Conclusions/Significance The results demonstrate that the characterization of targeted mutants yielded important information about essential virulence determinants that will help to identify the so far little understood extreme virulence of F. tularensis subspecies tularensis.
Collapse
|
27
|
Oral live vaccine strain-induced protective immunity against pulmonary Francisella tularensis challenge is mediated by CD4+ T cells and antibodies, including immunoglobulin A. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:444-52. [PMID: 19211773 DOI: 10.1128/cvi.00405-08] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Francisella tularensis is an intracellular gram-negative bacterium and the etiological agent of pulmonary tularemia. Given the high degrees of infectivity in the host and of dissemination of bacteria following respiratory infection, immunization strategies that target mucosal surfaces are critical for the development of effective vaccines against this organism. In this study, we have characterized the efficacy of protective immunity against pneumonic tularemia following oral vaccination with F. tularensis LVS (live vaccine strain). Mice vaccinated orally with LVS displayed colocalization of LVS with intestinal M cells, with subsequent enhanced production of splenic antigen-specific gamma interferon and of systemic and mucosal antibodies, including immunoglobulin A (IgA). LVS-vaccinated BALB/c mice were highly protected against intranasal (i.n.) SCHU S4 challenge and exhibited significantly less bacterial replication in the lungs, liver, and spleen than mock-immunized animals. Depletion of CD4(+) T cells significantly abrogated the protective immunity, and mice deficient in B cells or IgA displayed partial protection against SCHU S4 challenge. These results suggest that oral vaccination with LVS induces protective immunity against i.n. challenge with F. tularensis SCHU S4 by a process mediated cooperatively by CD4(+) T cells and antibodies, including IgA.
Collapse
|
28
|
Raymond CR, Conlan JW. Differential susceptibility of Sprague-Dawley and Fischer 344 rats to infection by Francisella tularensis. Microb Pathog 2009; 46:231-4. [PMID: 19490832 DOI: 10.1016/j.micpath.2009.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 01/08/2009] [Accepted: 01/14/2009] [Indexed: 10/21/2022]
Abstract
The type A and B subspecies of Francisella tularensis cause severe disease, tularemia, in humans. However, only the former can be lethal especially if inhaled. It is likely that non-lethal infection is due at least in part to the ability of innate host defenses to control pathogen growth whilst acquired immunity develops. Most common small laboratory animals rapidly succumb to clinical strains of F. tularensis and are, therefore, poor models with which to study innate immunity. In an attempt to improve upon this situation in the present study, Sprague-Dawley and Fischer 344 rats were examined for their ability to combat challenge with type A and B strains of the pathogen. Sprague-Dawley rats were significantly more resistant than Fischer rats to infection with either subspecies. This correlated with the ability of Sprague-Dawley rats to arrest the growth of the pathogen at both the site of challenge and at sites of disseminated infection. The rapidity with which F. tularensis kills susceptible rats and the early onset of control of infection in resistant rats suggests that differences in innate immunity account for these disparate outcomes. Thus, the rat might be a more useful model for studying innate immunity to virulent F. tularensis than other small mammals.
Collapse
Affiliation(s)
- Claudine R Raymond
- National Research Council Canada, Institute for Biological Sciences, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | | |
Collapse
|
29
|
Roth KM, Gunn JS, Lafuse W, Satoskar AR. Francisella inhibits STAT1-mediated signaling in macrophages and prevents activation of antigen-specific T cells. Int Immunol 2009; 21:19-28. [PMID: 19001470 PMCID: PMC2638840 DOI: 10.1093/intimm/dxn119] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Accepted: 10/04/2008] [Indexed: 12/26/2022] Open
Abstract
Signal transducer and activator of transcription-1 (STAT1) signaling mediate most biological functions of IFNalpha, IFNbeta and IFNgamma although recent studies indicate that IFNgamma can alter expression of several genes via a STAT1-independent pathway. STAT1 is critical for immunity against a variety of intracellular pathogens and some studies show that pathogens evade host immunity by interfering with STAT1 signaling. Here, we have investigated the role of STAT1 in host defense against pulmonary Francisella novicida infection using STAT1-/- mice. In addition, we examined the effect of F. novicida on STAT1 signaling in macrophages and on their ability to activate antigen-specific T cells. Both wild-type (WT) and STAT1-/- BALB/c mice were susceptible to aerosol challenge with 10(3) F. novicida and displayed 100% mortality. However, STAT1-/- mice developed more severe pneumonia, liver pathology and succumbed to infection faster than WT mice. The lungs, liver and hearts from F. novicida-infected STAT1-/- mice also contained more bacteria than WT mice at the time of death. In vitro studies showed that F. novicida suppressed IFNgammaRalpha (alpha subunit of IFNgamma receptor) and MHC class II expression, down-regulated IFNgamma-induced STAT1 activation and reduced nuclear binding of STAT1 in RAW264.7 macrophages. Furthermore, F. novicida-infected BMDM loaded with ovalbumin (OVA) were less efficient in activating OVA-specific CD4+ T cells in vitro. These findings demonstrate that STAT1-mediated signaling participates in the host defense against pulmonary F. novicida infection but it is not sufficient to prevent mortality associated with this infection. Moreover, our results show that F. novicida attenuates STAT1-mediated IFNgamma signaling in macrophages and impairs their ability to activate antigen-specific CD4+ T cells.
Collapse
Affiliation(s)
- Kimberly M Roth
- Department of Microbiology, Ohio State University, Columbus, OH, USA
| | | | | | | |
Collapse
|
30
|
Levofloxacin rescues mice from lethal intra-nasal infections with virulent Francisella tularensis and induces immunity and production of protective antibody. Vaccine 2008; 26:6874-82. [PMID: 18930100 DOI: 10.1016/j.vaccine.2008.09.077] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 09/09/2008] [Accepted: 09/26/2008] [Indexed: 10/21/2022]
Abstract
The ability to protect mice against respiratory infections with virulent Francisella tularensis has been problematic and the role of antibody-versus-cell-mediated immunity controversial. In this study, we tested the hypothesis that protective immunity can develop in mice that were given antibiotic therapy following infection via the respiratory tract with F. tularensis SCHU S4. We show that mice infected with a lethal dose of SCHU S4, via an intra-nasal challenge, could be protected with levofloxacin treatment. This protection was evident even when levofloxacin treatment was delayed 72h post-infection. At early time points after levofloxacin treatment, significant numbers of bacteria could be recovered from the lungs and spleens of mice, which was followed by a dramatic disappearance of bacteria from these tissues. Mice successfully treated with levofloxacin were later shown to be almost completely resistant to re-challenge with SCHU S4 by the intra-nasal route. Serum antibody appeared to play an important role in this immunity. Normal mice, when given sera from animals protected by levofloxacin treatment, were solidly protected from a lethal intra-nasal challenge with SCHU S4. The protective antiserum contained high titers of SCHU S4-specific IgG2a, indicating that a strong Th1 response was induced following levofloxacin treatment. Thus, this study describes a potentially valuable animal model for furthering our understanding of respiratory tularemia and provides suggestive evidence that antibody can protect against respiratory infections with virulent F. tularensis.
Collapse
|
31
|
Bakshi CS, Malik M, Mahawar M, Kirimanjeswara GS, Hazlett KRO, Palmer LE, Furie MB, Singh R, Melendez JA, Sellati TJ, Metzger DW. An improved vaccine for prevention of respiratory tularemia caused by Francisella tularensis SchuS4 strain. Vaccine 2008; 26:5276-88. [PMID: 18692537 DOI: 10.1016/j.vaccine.2008.07.051] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 07/14/2008] [Accepted: 07/15/2008] [Indexed: 11/29/2022]
Abstract
Vaccination of mice with Francisella tularensis live vaccine strain (LVS) mutants described so far have failed to induce protection in C57BL/6 mice against challenge with the virulent strain F. tularensis SchuS4. We have previously reported that a mutant of F. tularensis LVS deficient in iron superoxide dismutase (sodB(Ft)) is hypersensitive to oxidative stress and attenuated for virulence in mice. Herein, we evaluated the efficacy of this mutant as a vaccine candidate against respiratory tularemia caused by F. tularensis SchuS4. C57BL/6 mice were vaccinated intranasally (i.n.) with the sodB(Ft) mutant and challenged i.n. with lethal doses of F. tularensis SchuS4. The level of protection against SchuS4 challenge was higher in sodB(Ft) vaccinated group as compared to the LVS vaccinated mice. sodB(Ft) vaccinated mice following SchuS4 challenge exhibited significantly reduced bacterial burden in lungs, liver and spleen, regulated production of pro-inflammatory cytokines and less severe histopathological lesions compared to the LVS vaccinated mice. The sodB(Ft) vaccination induced a potent humoral immune response and protection against SchuS4 required both CD4 and CD8 T cells in the vaccinated mice. sodB(Ft) mutants revealed upregulated levels of chaperonine proteins DnaK, GroEL and Bfr that have been shown to be important for generation of a potent immune response against Francisella infection. Collectively, this study describes an improved live vaccine candidate against respiratory tularemia that has an attenuated virulence and enhanced protective efficacy than the LVS.
Collapse
Affiliation(s)
- Chandra Shekhar Bakshi
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Native outer membrane proteins protect mice against pulmonary challenge with virulent type A Francisella tularensis. Infect Immun 2008; 76:3664-71. [PMID: 18505805 DOI: 10.1128/iai.00374-08] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Francisella tularensis is a gram-negative intracellular bacterium and the causative agent of the zoonotic disease tularemia. F. tularensis is a category A select agent and thus a potential agent of bioterrorism. Whereas an F. tularensis live, attenuated vaccine strain (LVS) is the basis of an investigational vaccine, this vaccine is not licensed for human use because of efficacy and safety concerns. In the present study, we immunized mice with isolated native outer membrane proteins (OMPs), ethanol-inactivated LVS (iLVS), or purified LVS lipopolysaccharide (LPS) and assessed the ability of each vaccine preparation to protect mice against pulmonary challenge with the virulent type A F. tularensis strain SchuS4. Antibody isotyping indicated that both Th1 and Th2 antibody responses were generated in mice after immunization with OMPs or iLVS, whereas LPS immunization resulted in only immunoglobulin A production. In survival studies, OMP immunization provided the greatest level of protection (50% survival at 20 days after infection with SchuS4), and there were associated 3-log reductions in the spleen and liver bacterial burdens (compared to nonvaccinated mice). Cytokine quantitation for the sera of SchuS4-challenged mice indicated that OMP and iLVS immunizations induced high levels of tumor necrosis factor alpha and interleukin-2 (IL-2) production, whereas only OMP immunization induced high levels of IL-10 production. By comparison, high levels of proinflammatory cytokines, including RANTES, granulocyte colony-stimulating factor, IL-6, IL-1alpha, IL-12p40, and KC, in nonvaccinated mice indicated that these cytokines may facilitate disease progression. Taken together, the results of this study demonstrate the potential utility of an OMP subunit (acellular) vaccine for protecting mammals against type A F. tularensis.
Collapse
|
33
|
Roth KM, Oghumu S, Satoskar AA, Gunn JS, van Rooijen N, Satoskar AR. Respiratory infection with Francisella novicida induces rapid dystrophic cardiac calcinosis (DCC). ACTA ACUST UNITED AC 2008; 53:72-8. [PMID: 18400010 DOI: 10.1111/j.1574-695x.2008.00395.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Francisella tularensis causes pulmonary tularemia and death in humans when left untreated. Here, using a novel aerosol infection model, we show that acute pulmonary Francisella novicida infection not only causes pneumonia and liver damage, but also induces dystrophic cardiac calcinosis (DCC) in BALB/c mice. C57BL/6 mice also develop pneumonia and hepatic damage, but fail to develop DCC. Development of DCC in BALB/c mice is associated with significant induction of RANKL but not osteopontin in their organs. Depletion of lung macrophages prior to infection markedly reduces pericarditis and calcification in BALB/c mice but does not increase their susceptibility to infection.
Collapse
Affiliation(s)
- Kimberly M Roth
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
34
|
Conlan JW, Zhao X, Harris G, Shen H, Bolanowski M, Rietz C, Sjostedt A, Chen W. Molecular immunology of experimental primary tularemia in mice infected by respiratory or intradermal routes with type A Francisella tularensis. Mol Immunol 2008; 45:2962-9. [PMID: 18321578 DOI: 10.1016/j.molimm.2008.01.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 01/15/2008] [Accepted: 01/19/2008] [Indexed: 11/18/2022]
Abstract
The type A subspecies of Francisella tularensis is a highly virulent facultative intracellular bacterial pathogen, and a potential biological weapon. Recently, there has been renewed interest in developing new vaccines and therapeutics against this bacterium. Natural cases of disease, tularemia, caused by the type A subspecies are very rare. Therefore, the United States Food and Drug Administration will rely on the so-called Animal Rule for efficacy testing of anti-Francisella medicines. This requires the human disease to be modeled in one or more animal species in which the pathogenicity of the agent is reasonably well understood. Mice are natural hosts for F. tularensis, and might be able to satisfy this requirement. Tularemia pathogenesis appears to be primarily due to the host inflammatory response which is poorly understood at the molecular level. Additionally, the extent to which this response varies depending on host and pathogen genetic background, or by pathogen challenge route or dose is unknown. Therefore, the present study examined sera and infected tissues from C57BL/6 and BALB/c mice challenged by natural intradermal (ID) and respiratory routes with one of two distinct type A strains of the pathogen for cytokine and chemokine responses that might help to explain the morbidity associated with tularemia. The results show that the molecular immune response was mostly similar regardless of the variables examined. For instance, mRNA for the proinflammatory cytokine IL-6, and chemokines KC, and IP-10 was consistently upregulated at all sites of infection. Upregulation of mRNA for several other cytokines and chemokines occurred in a more tissue restricted manner. For instance, IFN-gamma was highly upregulated in the skin of BALB/c, but not C57BL/6 mice after ID inoculation of the pathogen, whilst IL-10 mRNA upregulation was only consistently seen in the skin and lungs.
Collapse
Affiliation(s)
- J Wayne Conlan
- Institute for Biological Sciences, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Li J, Ryder C, Mandal M, Ahmed F, Azadi P, Snyder DS, Pechous RD, Zahrt T, Inzana TJ. Attenuation and protective efficacy of an O-antigen-deficient mutant of Francisella tularensis LVS. MICROBIOLOGY-SGM 2007; 153:3141-3153. [PMID: 17768257 DOI: 10.1099/mic.0.2007/006460-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Francisella tularensis is a zoonotic, Gram-negative coccobacillus that causes tularemia in humans and animals. F. tularensis subspecies tularensis (type A) and F. tularensis subspecies holarctica (type B) are antigenically similar and more virulent than Francisella novicida in humans. The genetic locus that encodes the LPS O antigen was found to be substantially different between the type B live vaccine strain (LVS) and F. novicida. One LVS-specific gene with homology to a galactosyl transferase was selected for allelic replacement using a sacB-chloramphenicol expression suicide plasmid, and recombinants were screened for colony morphology on Congo red agar that matched that of F. novicida. Two mutants (WbtI(S187Y) and WbtI(G191V)) were isolated that contained substitutions in conserved motifs in the sugar transamine/perosamine synthetase (WbtI) of the O-antigen locus, and the latter mutant was extensively tested and characterized. WbtI(G191V) grew at the same rate as the parent strain in Chamberlain's defined medium, completely lacked O antigen, was serum-sensitive but could grow in a mouse macrophage cell line, had increased resistance to sodium deoxycholate, and was highly attenuated in mice. Complementation of WbtI(G191V) with the wild-type wbtI gene in trans restored normal LPS synthesis, phenotypic properties similar to the parent, and virulence in mice. Immunization with WbtI(G191V) protected mice against a relatively low-dose intraperitoneal challenge with LVS, but was less protective against a high-dose challenge. These results indicate that complete loss of O antigen alters the surface phenotype and abrogates virulence in F. tularensis, but also compromises the induction of full protective immunity against F. tularensis infection in mice.
Collapse
Affiliation(s)
- Jiaxin Li
- Center for Molecular Medicine and Infectious Diseases, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Cheryl Ryder
- Center for Molecular Medicine and Infectious Diseases, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Manas Mandal
- Center for Molecular Medicine and Infectious Diseases, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Farzana Ahmed
- Center for Molecular Medicine and Infectious Diseases, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - D Scott Snyder
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Roger D Pechous
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thomas Zahrt
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thomas J Inzana
- Center for Molecular Medicine and Infectious Diseases, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
36
|
Identification of immunologic and pathologic parameters of death versus survival in respiratory tularemia. Infect Immun 2007; 76:486-96. [PMID: 18025095 DOI: 10.1128/iai.00862-07] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis can cause severe disseminated disease after respiratory infection. The identification of factors involved in mortality or recovery following induction of tularemia in the mouse will improve our understanding of the natural history of this disease and facilitate future evaluation of vaccine candidate preparations. BALB/c mice were infected intranasally with the live vaccine strain (LVS) of F. tularensis subsp. holarctica and euthanized at different stages of disease to analyze the induction of immune molecules, gross anatomical features of organs, bacterial burdens, and progression of the histopathological changes in lung and spleen. Tissue-specific interleukin-6 (IL-6), macrophage inflammatory protein 2, and monocyte chemotactic protein 1 were immune markers of mortality, while anti-LVS immunoglobulin M and IL-1beta were associated with survival. Moribund mice had enlarged spleens and lungs, while surviving mice had even more prominent splenomegaly and normal-appearing lungs. Histopathology of the spleens of severely ill mice was characterized by disrupted lymphoid follicles and fragmented nuclei, while the spleens of survivors appeared healthy but with increased numbers of megakaryocytes and erythrocytes. Histopathology of the lungs of severely ill mice indicated severe pneumonia. Lungs of survivors at early time points showed increased inflammation, while at late times they appeared healthy with peribronchial lymphoid aggregates. Our results suggest that host immune factors are able to affect bacterial dissemination after respiratory tularemia, provide new insights regarding the pathological characteristics of pulmonary tularemia leading to systemic disease, and potentially identify immune markers associated with recovery from the disease.
Collapse
|
37
|
Eyles JE, Hartley MG, Laws TR, Oyston PCF, Griffin KF, Titball RW. Protection afforded against aerosol challenge by systemic immunisation with inactivated Francisella tularensis live vaccine strain (LVS). Microb Pathog 2007; 44:164-8. [PMID: 17904793 DOI: 10.1016/j.micpath.2007.08.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Accepted: 08/10/2007] [Indexed: 11/20/2022]
Abstract
BALB/c mice were immunised with inactivated Francisella tularensis live vaccine strain (LVS) and the level of protection afforded against aerosol challenge with virulent strains of F. tularensis ascertained. Intramuscular (IM) injection of inactivated LVS with an aluminium-hydroxide-based adjuvant-stimulated IgG1-biased LVS-specific antibody responses and afforded no protection against aerosol challenge with subspecies holarctica (strain HN63). Conversely, IM injection of inactivated LVS adjuvanted with preformed immune-stimulating complexes (ISCOMS) admixed with immunostimulatory CpG oligonucleotides afforded robust protection against aerosol-initiated infection with HN63. However, despite a significantly extended time-to-death relative to naïve controls, the majority of mice immunised with the most potent vaccine formulation were not protected against a low-dose aerosol challenge with subspecies tularensis (strain Schu S4). These data indicate that parenterally administered non-living vaccines can be used for effective immunisation against aerosol challenges with subspecies holarctica, although not high virulence strains of F. tularensis.
Collapse
Affiliation(s)
- J E Eyles
- Defence Science and Technology Laboratory, Porton Down, Salisbury, UK.
| | | | | | | | | | | |
Collapse
|
38
|
Kirimanjeswara GS, Golden JM, Bakshi CS, Metzger DW. Prophylactic and Therapeutic Use of Antibodies for Protection against Respiratory Infection withFrancisella tularensis. THE JOURNAL OF IMMUNOLOGY 2007; 179:532-9. [PMID: 17579074 DOI: 10.4049/jimmunol.179.1.532] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The role of Abs in protection against respiratory infection with the intracellular bacterium Francisella tularensis is not clear. To investigate the ability of Abs to clear bacteria from the lungs and prevent systemic spread, immune serum was passively administered i.p. to naive mice before intranasal F. tularensis live vaccine strain infection. It was found that immune serum treatment provided 100% protection against lethal challenge while normal serum or Ig-depleted immune serum provided no protection. Protective efficacy was correlated with increased clearance of bacteria from the lung and required expression of FcgammaR on phagocytes, including macrophages and neutrophils. However, complement was not required for protection. In vitro experiments demonstrated that macrophages were more readily infected by Ab-opsonized bacteria but became highly efficient in killing upon activation by IFN-gamma. Consistent with this finding, in vivo Ab-mediated protection was found to be dependent upon IFN-gamma. SCID mice were not protected by passive Ab transfer, suggesting that T cells but not NK cells serve as the primary source for IFN-gamma. These data suggest that a critical interaction of humoral and cellular immune responses is necessary to provide sterilizing immunity against F. tularensis. Of considerable interest was the finding that serum Abs were capable of conferring protection against lethal respiratory tularemia when given 24-48 h postexposure. Thus, this study provides the first evidence for the therapeutic use of Abs in Francisella-infected individuals.
Collapse
Affiliation(s)
- Girish S Kirimanjeswara
- Center for Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Avenue Albany, NY 12208, USA
| | | | | | | |
Collapse
|
39
|
Malik M, Bakshi CS, McCabe K, Catlett SV, Shah A, Singh R, Jackson PL, Gaggar A, Metzger DW, Melendez JA, Blalock JE, Sellati TJ. Matrix metalloproteinase 9 activity enhances host susceptibility to pulmonary infection with type A and B strains of Francisella tularensis. THE JOURNAL OF IMMUNOLOGY 2007; 178:1013-20. [PMID: 17202364 DOI: 10.4049/jimmunol.178.2.1013] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A striking feature of pulmonary infection with the Gram-negative intracellular bacterium Francisella tularensis, a category A biological threat agent, is an intense accumulation of inflammatory cells, particularly neutrophils and macrophages, at sites of bacterial replication. Given the essential role played by host matrix metalloproteinases (MMPs) in modulating leukocyte recruitment and the potentially indiscriminate destructive capacity of these cells, we investigated whether MMP-9, an important member of this protease family released by neutrophils and activated macrophages, plays a role in the pathogenesis of respiratory tularemia. We found that F. tularensis induced expression of MMP-9 in FVB/NJ mice and that the action of this protease is associated with higher bacterial burdens in pulmonary and extrapulmonary tissues, development of more extensive histopathology predominated by neutrophils, and increased morbidity and mortality compared with mice lacking MMP-9 (MMP-9(-/-)). Moreover, MMP-9(-/-) mice were able to resolve infection with either the virulence-attenuated type B (live vaccine strain) or the highly virulent type A (SchuS4) strain of F. tularensis. Disease resolution was accompanied by diminished leukocyte recruitment and reductions in both bacterial burden and proinflammatory cytokine production. Notably, neutrophilic infiltrates were significantly reduced in MMP-9(-/-) mice, owing perhaps to limited release of Pro-Gly-Pro, a potent neutrophil chemotactic tripeptide released from extracellular matrix through the action of MMP-9. Collectively, these results suggest that MMP-9 activity plays a central role in modulating the clinical course and severity of respiratory tularemia and identifies MMPs as novel targets for therapeutic intervention as a means of modulating neutrophil recruitment.
Collapse
Affiliation(s)
- Meenakshi Malik
- Center for Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Tempel R, Lai XH, Crosa L, Kozlowicz B, Heffron F. Attenuated Francisella novicida transposon mutants protect mice against wild-type challenge. Infect Immun 2006; 74:5095-105. [PMID: 16926401 PMCID: PMC1594869 DOI: 10.1128/iai.00598-06] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis is the bacterial pathogen that causes tularemia in humans and a number of animals. To date, there is no approved vaccine for this widespread and life-threatening disease. The goal of this study was to identify F. tularensis mutants that can be used in the development of a live attenuated vaccine. We screened F. novicida transposon mutants to identify mutants that exhibited reduced growth in mouse macrophages, as these cells are the preferred host cells of Francisella and an essential component of the innate immune system. This approach yielded 16 F. novicida mutants that were 100-fold more attenuated for virulence in a mouse model than the wild-type parental strain. These mutants were then tested to determine their abilities to protect mice against challenge with high doses of wild-type bacteria. Five of the 16 attenuated mutants (with mutations corresponding to dsbB, FTT0742, pdpB, fumA, and carB in the F. tularensis SCHU S4 strain) provided mice with protection against challenge with high doses (>8 x 10(5) CFU) of wild-type F. novicida. We believe that these findings will be of use in the design of a vaccine against tularemia.
Collapse
Affiliation(s)
- Rebecca Tempel
- 6543 Basic Sciences Addition/CROET Building, Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | | | | | | | | |
Collapse
|
41
|
Mohapatra NP, Balagopal A, Soni S, Schlesinger LS, Gunn JS. AcpA is a Francisella acid phosphatase that affects intramacrophage survival and virulence. Infect Immun 2006; 75:390-6. [PMID: 17060465 PMCID: PMC1828385 DOI: 10.1128/iai.01226-06] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AcpA of Francisella spp. is a respiratory-burst-inhibiting acid phosphatase that also exhibits phospholipase C activity. To better understand the molecular basis of AcpA in virulence, a deletion of acpA was constructed in Francisella novicida. The phosphatase and lipase activities were reduced 10-fold and 8-fold, respectively, in the acpA mutant compared to the wild type and were found mostly associated with the outer membrane. The acpA mutant was more susceptible to intracellular killing than the wild-type strain in the THP-1 human macrophage-like cell line. In addition, mice infected with the acpA mutant survived longer than the wild-type strain and were less fit than the wild-type strain in competition infection assays. Transmission electron microscopy showed that the acpA mutant was delayed in escape from macrophage phagosomes, as more than 75% of acpA mutant bacteria could still be found inside phagosomes after 12 h of infection in THP-1 cells and human monocyte-derived macrophages, whereas most of the wild-type bacteria had escaped from the phagosome by 6 h postinfection. Thus, AcpA affects intracellular trafficking and the fate of Francisella within host macrophages.
Collapse
Affiliation(s)
- Nrusingh P Mohapatra
- Center for Microbial Interface Biology, Department of Molecular Virology, Immunology, and Medical Genetics, and Department of Internal Medicine, Division of Infectious Diseases, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
42
|
Twine SM, Petit MD, Shen H, Mykytczuk NCS, Kelly JF, Conlan JW. Immunoproteomic analysis of the murine antibody response to successful and failed immunization with live anti-Francisella vaccines. Biochem Biophys Res Commun 2006; 346:999-1008. [PMID: 16781667 DOI: 10.1016/j.bbrc.2006.06.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Accepted: 06/02/2006] [Indexed: 11/26/2022]
Abstract
Francisella tularensis subspecies tularensis is one of the most virulent of bacterial pathogens for humans. Protective immunity against the pathogen can be induced in humans and some, but not all, mouse strains by vaccination with live, but not killed, vaccines. In mice, this protection is mediated predominantly by CD4+ and CD8+ T cells. This is thought to be the case too for humans. Nevertheless, it is possible that successful vaccination elicits antigen-specific antibodies that can serve as correlates of protection. To test this hypothesis we examined the repertoire of antibodies induced following successful immunization of BALB/c and CH3/HeN mice versus unsuccessful vaccination of C57BL/6 and DBA\2 mice with F. tularensis Live Vaccine Strain or following unsuccessful vaccination of BALB/c mice with highly related subspecies, F. novicida. The results showed that successful vaccination elicited antibodies to at least six proteins that were not recognized by antisera from vaccinated but unprotected mice.
Collapse
Affiliation(s)
- Susan M Twine
- Institute for Biological Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Ont., Canada K1A 0R6.
| | | | | | | | | | | |
Collapse
|
43
|
Katz J, Zhang P, Martin M, Vogel SN, Michalek SM. Toll-like receptor 2 is required for inflammatory responses to Francisella tularensis LVS. Infect Immun 2006; 74:2809-16. [PMID: 16622218 PMCID: PMC1459727 DOI: 10.1128/iai.74.5.2809-2816.2006] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Accepted: 02/20/2006] [Indexed: 01/19/2023] Open
Abstract
Francisella tularensis, a gram-negative bacterium, is the etiologic agent of tularemia and has recently been classified as a category A bioterrorism agent. Infections with F. tularensis result in an inflammatory response that plays an important role in the pathogenesis of the disease; however, the cellular mechanisms mediating this response have not been completely elucidated. In the present study, we determined the role of Toll-like receptors (TLRs) in mediating inflammatory responses to F. tularensis LVS, and the role of NF-kappaB in regulating these responses. Stimulation of bone marrow-derived dendritic cells from C57BL/6 wild-type (wt) and TLR4-/- but not TLR2-/- mice, with live F. tularensis LVS elicited a dose-dependent increase in the production of tumor necrosis factor alpha. F. tularensis LVS also induced in a dose-dependent manner an up-regulation in the expression of the costimulatory molecules CD80 and CD86 and of CD40 and the major histocompatibility complex class II molecules on dendritic cells from wt and TLR4-/- but not TLR2-/- mice. TLR6, not TLR1, was shown to be involved in mediating the inflammatory response to F. tularensis LVS, indicating that the functional heterodimer is TLR2/TLR6. Stimulation of dendritic cells with F. tularensis resulted in the activation of NF-kappaB, which resulted in a differential effect on the production of pro- and anti-inflammatory cytokines. Taken together, our results demonstrate the role of TLR2/TLR6 in the host's inflammatory response to F. tularensis LVS in vitro and the regulatory function of NF-kappaB in modulating the inflammatory response.
Collapse
Affiliation(s)
- Jannet Katz
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
44
|
Twine SM, Shen H, Kelly JF, Chen W, Sjöstedt A, Conlan JW. Virulence comparison in mice of distinct isolates of type A Francisella tularensis. Microb Pathog 2006; 40:133-8. [PMID: 16448801 PMCID: PMC1868512 DOI: 10.1016/j.micpath.2005.12.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2005] [Revised: 12/08/2005] [Accepted: 12/12/2005] [Indexed: 10/25/2022]
Abstract
Francisella tularensis subspecies tularensis (type A F. tularensis) is considered to be one of the most virulent of all bacterial pathogens. Mice are extremely susceptible to infection with this subspecies (LD100 via various inoculation routes is <10 cfu). However, it has not been established whether overt virulence differences exist amongst type A strains of F. tularensis. To this end, the present study compared the virulence of two distinct type A strains, FSC033 and SCHU S4, for naïve mice and mice immunized with the live vaccine strain of the pathogen, F. tularensis LVS. One nominal isolate of SCHU S4 was found to be completely avirulent. Another isolate was highly virulent, but all examined cases appeared somewhat less virulent than FSC033. The implication of these findings for future infection and immunity studies is discussed.
Collapse
Affiliation(s)
- Susan M. Twine
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ont. K1A 0R6, Canada
| | - Hua Shen
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ont. K1A 0R6, Canada
| | - John F. Kelly
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ont. K1A 0R6, Canada
| | - Wangxue Chen
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ont. K1A 0R6, Canada
| | - Anders Sjöstedt
- Department of Clinical Microbiology, Clinical Bacteriology, Umeå University, SE-90185 Umeå, Sweden
| | - J. Wayne Conlan
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ont. K1A 0R6, Canada
- * Corresponding author. Tel.: +1613 993 8829; fax: +1613 952 9092. E-mail address: (J.W. Conlan)
| |
Collapse
|
45
|
Twine S, Byström M, Chen W, Forsman M, Golovliov I, Johansson A, Kelly J, Lindgren H, Svensson K, Zingmark C, Conlan W, Sjöstedt A. A mutant of Francisella tularensis strain SCHU S4 lacking the ability to express a 58-kilodalton protein is attenuated for virulence and is an effective live vaccine. Infect Immun 2006; 73:8345-52. [PMID: 16299332 PMCID: PMC1307091 DOI: 10.1128/iai.73.12.8345-8352.2005] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis subsp. tularensis (type A) strain SCHU S4 is a prototypic strain of the pathogen that is highly virulent for humans and other mammals. Its intradermal (i.d.) 50% lethal dose (LD50) for mice is <10 CFU. We discovered a spontaneous mutant, designated FSC043, of SCHU S4 with an i.d. LD50 of >10(8) CFU. FSC043 effectively vaccinated mice against challenge with a highly virulent type A strain, and the protective efficacy was at least as good as that of F. tularensis LVS, an empirically attenuated strain which has been used as an efficacious human vaccine. Comparative proteomics was used to identify two proteins of unknown function that were identified as defective in LVS and FSC043, and deletion mutants of SCHU S4 were created for each of the two encoding genes. One mutant, the DeltaFTT0918 strain, failed to express a 58-kDa protein, had an i.d. LD50 of approximately 10(5) CFU, and was found to be less capable than SCHU S4 of growing in peritoneal mouse macrophages. Mice that recovered from sublethal infection with the DeltaFTT0918 mutant survived when challenged 2 months later with >100 LD50s of the highly virulent type A strain FSC033. This is the first report of the generation of defined mutants of F. tularensis subsp. tularensis and their use as live vaccines.
Collapse
Affiliation(s)
- Susan Twine
- Department of Clinical Microbiology, Clinical Bacteriology, Umeå University, SE-901 85 Umeå, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Oyston PCF, Quarry JE. Tularemia vaccine: past, present and future. Antonie van Leeuwenhoek 2005; 87:277-81. [PMID: 15928980 DOI: 10.1007/s10482-004-6251-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Accepted: 11/15/2004] [Indexed: 11/28/2022]
Abstract
Francisella tularensis is a Gram negative intracellular pathogen that causes the highly debilitating or fatal disease tularemia. F. tularensis can infect a wide range of animals and can be transmitted to humans in a variety of ways, the most common being by the bite of an infected insect or arthropod vector. The attenuated F. tularensis live vaccine strain (LVS) has been used previously under investigational new drug status to vaccinate at-risk individuals. However the history of the strain and lack of knowledge regarding the basis of attenuation has so far prevented its licensing. Therefore the focus of current research is on producing a new vaccine against tularemia that would be suitable for licensing.
Collapse
Affiliation(s)
- Petra C F Oyston
- Defence Science and Technology Laboratories, Porton Down, Salisbury, Wiltshire, SP4 0JQ.
| | | |
Collapse
|
47
|
Ulrich RL, Amemiya K, Waag DM, Roy CJ, DeShazer D. Aerogenic vaccination with a Burkholderia mallei auxotroph protects against aerosol-initiated glanders in mice. Vaccine 2005; 23:1986-92. [PMID: 15734072 DOI: 10.1016/j.vaccine.2004.10.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2004] [Revised: 09/27/2004] [Accepted: 10/05/2004] [Indexed: 11/24/2022]
Abstract
Burkholderia mallei is an obligate mammalian pathogen that causes the zoonotic disease glanders. Two live attenuated B. mallei strains, a capsule mutant and a branched-chain amino acid auxotroph, were evaluated for use as vaccines against aerosol-initiated glanders in mice. Animals were aerogenically vaccinated and serum samples were obtained before aerosol challenge with a high-dose (>300 times the LD50) of B. mallei ATCC 23344. Mice vaccinated with the capsule mutant developed a Th2-like Ig subclass antibody response and none survived beyond 5 days. In comparison, the auxotrophic mutant elicited a Th1-like Ig subclass antibody response and 25% of the animals survived for 1 month postchallenge. After a low-dose (5 times the LD50) aerosol challenge, the survival rates of auxotroph-vaccinated and unvaccinated animals were 50 and 0%, respectively. Thus, live attenuated strains that promote a Th1-like Ig response may serve as promising vaccine candidates against aerosol infection with B. mallei.
Collapse
Affiliation(s)
- Ricky L Ulrich
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD 21702, USA
| | | | | | | | | |
Collapse
|
48
|
Wayne Conlan J, Shen H, Kuolee R, Zhao X, Chen W. Aerosol-, but not intradermal-immunization with the live vaccine strain of Francisella tularensis protects mice against subsequent aerosol challenge with a highly virulent type A strain of the pathogen by an αβ T cell- and interferon gamma- dependent mechanism. Vaccine 2005; 23:2477-85. [PMID: 15752834 DOI: 10.1016/j.vaccine.2004.10.034] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Revised: 10/18/2004] [Accepted: 10/23/2004] [Indexed: 11/29/2022]
Abstract
Francisella tularensis is an extremely virulent facultative intracellular bacterial pathogen of many mammalian species including mice and humans in which it causes a spectrum of disease collectively called tularemia. In humans, intradermal or inhaled inocula of 10cfu or less of the most virulent strains of the pathogen are sufficient to cause severe infection and possible death; in mice similar inocula are routinely lethal. An attenuated live vaccine strain, F. tularensis LVS, was developed almost 50 years ago, and remains the sole prophylactic against virulent strains of the pathogen. Using F. tularensis LVS as a model vaccine, we recently showed that it was possible to systemically immunize various mouse strains and protect them against subsequent massive (2000 cfu) intradermal (i.d.) challenge, but not against low dose (approximately 10 cfu) aerosol challenge, with virulent strains of the pathogen. This is troubling because the latter route is considered an important means of deliberately disseminating F. tularensis in a bioterrorist attack. Others have previously shown that administering LVS to humans, guinea pigs and monkeys as an aerosol enhanced protection against subsequent aerosol challenge with virulent F. tularensis. In the present study, we show the same phenomenon in BALB/c and C3H/HeN mice. In this model, interferon gamma (IFNgamma) and CD4+ and CD8+ T cells are essential for the expression of anti-Francisella immunity in the lungs. Combined this immune response operates by limiting dissemination of the pathogen to susceptible internal organs. Further, understanding of how inhaled LVS elicits local cell-mediated protective immunity will be critical for devising improved vaccines against pulmonary tularemia.
Collapse
Affiliation(s)
- J Wayne Conlan
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ont., Canada K1A 0R6.
| | | | | | | | | |
Collapse
|
49
|
Shen H, Chen W, Conlan JW. Mice sublethally infected with Francisella novicida U112 develop only marginal protective immunity against systemic or aerosol challenge with virulent type A or B strains of F. tularensis. Microb Pathog 2004; 37:107-10. [PMID: 15312850 DOI: 10.1016/j.micpath.2004.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2004] [Accepted: 04/23/2004] [Indexed: 11/20/2022]
Abstract
The current study determined the ability of Francisella novicida to act as a live vaccine against the much more virulent, but closely related pathogen, Francisella tularensis. Live attenuated strains of the latter are effective vaccines against human tularemia. However, the molecular cause of their attenuation remains unknown, and this is a regulatory barrier for licensing such vaccines. Moreover, F. tularensis is exceptionally difficult to manipulate genetically. This is hampering the development of rationally attenuated vaccine strains. F. novicida shares a lot of genetic homology with F. tularensis and is more amenable to genetic manipulation. If the former naturally expresses the protective antigens of the latter, it could be used to develop a defined tularemia vaccine. However, the results presented herein show that wild-type F. novicida elicits almost no protection in mice against challenge with virulent F. tularensis.
Collapse
Affiliation(s)
- Hua Shen
- National Research Council Canada, Institute for Biological Sciences, 100 Sussex Drive, Room 3065 Ottawa, Ont., Canada K1A OR6
| | | | | |
Collapse
|