1
|
Penning S, Hong Y, Cunliffe T, Hor L, Totsika M, Paxman JJ, Heras B. Unveiling the versatility of the thioredoxin framework: Insights from the structural examination of Francisella tularensis DsbA1. Comput Struct Biotechnol J 2024; 23:4324-4336. [PMID: 39697679 PMCID: PMC11653150 DOI: 10.1016/j.csbj.2024.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
In bacteria the formation of disulphide bonds is facilitated by a family of enzymes known as the disulphide bond forming (Dsb) proteins, which, despite low sequence homology, belong to the thioredoxin (TRX) superfamily. Among these enzymes is the disulphide bond-forming protein A (DsbA); a periplasmic thiol oxidase responsible for catalysing the oxidative folding of numerous cell envelope and secreted proteins. Pathogenic bacteria often contain diverse Dsb proteins with distinct functionalities commonly associated with pathogenesis. Here we investigate FtDsbA1, a DsbA homologue from the Gram-negative bacterium Francisella tularensis. Our study shows that FtDsbA1 shares a conserved TRX-like fold bridged by an alpha helical bundle showcased by all DsbA-like proteins. However, FtDsbA1 displays a highly unique variation on this structure, containing an extended and flexible N-terminus and secondary structural elements inserted within the core of the TRX fold itself, which together twist the overall DsbA-like architecture. Additionally, FtDsbA1 exhibits variations to the well conserved active site with an unusual dipeptide in the catalytic CXXC redox centre (CGKC), and a trans configuration for the conserved cis-proline loop, known for governing DsbA-substrate interactions. FtDsbA1's redox properties are comparable to other DsbA enzymes, however, consistent with its atypical structure, functional analysis reveals FtDsbA1 has a high degree of substrate specificity suggesting a specialised role within F. tularensis' oxidative folding pathway. Overall, this work underscores the remarkable malleability of the TRX catalytic core, a ubiquitous and ancestral protein fold. This not only contributes to broadening the structural and functional diversity seen within proteins utilising this core fold but will also enhance the accuracy of AI-driven protein structural prediction tools.
Collapse
Affiliation(s)
- Stephanie Penning
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| | - Yaoqin Hong
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Taylor Cunliffe
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| | - Lilian Hor
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Jason J. Paxman
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| | - Begoña Heras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| |
Collapse
|
2
|
Moradkasani S, Maurin M, Farrokhi AS, Esmaeili S. Development, Strategies, and Challenges for Tularemia Vaccine. Curr Microbiol 2024; 81:126. [PMID: 38564047 DOI: 10.1007/s00284-024-03658-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
Francisella tularensis is a facultative intracellular bacterial pathogen that affects both humans and animals. It was developed into a biological warfare weapon as a result. In this article, the current status of tularemia vaccine development is presented. A live-attenuated vaccine that was designed over 50 years ago using the less virulent F. tularensis subspecies holarctica is the only prophylactic currently available, but it has not been approved for use in humans or animals. Other promising live, killed, and subunit vaccine candidates have recently been developed and tested in animal models. This study will investigate some possible vaccines and the challenges they face during development.
Collapse
Affiliation(s)
- Safoura Moradkasani
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, KabudarAhang, Hamadan, Iran
- Department of Epidemiology and Biostatics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Max Maurin
- CNRS, Grenoble INP, CHU Grenoble Alpes, TIMC-IMAG, Universite Grenoble Alpes, 38000, Grenoble, France
| | | | - Saber Esmaeili
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, KabudarAhang, Hamadan, Iran.
- Department of Epidemiology and Biostatics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
3
|
Mlynek KD, Cline CR, Biryukov SS, Toothman RG, Bachert BA, Klimko CP, Shoe JL, Hunter M, Hedrick ZM, Dankmeyer JL, Mou S, Fetterer DP, Qiu J, Lee ED, Cote CK, Jia Q, Horwitz MA, Bozue JA. The rLVS Δ capB/ iglABC vaccine provides potent protection in Fischer rats against inhalational tularemia caused by various virulent Francisella tularensis strains. Hum Vaccin Immunother 2023; 19:2277083. [PMID: 37975637 PMCID: PMC10760400 DOI: 10.1080/21645515.2023.2277083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
Francisella tularensis is one of the several biothreat agents for which a licensed vaccine is needed. To ensure vaccine protection is achieved across a range of virulent F. tularensis strains, we assembled and characterized a panel of F. tularensis isolates to be utilized as challenge strains. A promising tularemia vaccine candidate is rLVS ΔcapB/iglABC (rLVS), in which the vector is the LVS strain with a deletion in the capB gene and which additionally expresses a fusion protein comprising immunodominant epitopes of proteins IglA, IglB, and IglC. Fischer rats were immunized subcutaneously 1-3 times at 3-week intervals with rLVS at various doses. The rats were exposed to a high dose of aerosolized Type A strain Schu S4 (FRAN244), a Type B strain (FRAN255), or a tick derived Type A strain (FRAN254) and monitored for survival. All rLVS vaccination regimens including a single dose of 107 CFU rLVS provided 100% protection against both Type A strains. Against the Type B strain, two doses of 107 CFU rLVS provided 100% protection, and a single dose of 107 CFU provided 87.5% protection. In contrast, all unvaccinated rats succumbed to aerosol challenge with all of the F. tularensis strains. A robust Th1-biased antibody response was induced in all vaccinated rats against all F. tularensis strains. These results demonstrate that rLVS ΔcapB/iglABC provides potent protection against inhalational challenge with either Type A or Type B F. tularensis strains and should be considered for further analysis as a future tularemia vaccine.
Collapse
Affiliation(s)
- Kevin D. Mlynek
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Curtis R. Cline
- Pathology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Sergei S. Biryukov
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Ronald G. Toothman
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Beth A. Bachert
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Christopher P. Klimko
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Jennifer L. Shoe
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Melissa Hunter
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Zander M. Hedrick
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Jennifer L. Dankmeyer
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Sherry Mou
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - David P. Fetterer
- Regulated Research Administration Division, U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, USA
| | - Ju Qiu
- Regulated Research Administration Division, U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, USA
| | - Eric D. Lee
- Pathology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Christopher K. Cote
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Qingmei Jia
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, CA, USA
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, CA, USA
| | - Joel A. Bozue
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| |
Collapse
|
4
|
Benziger PT, Kopping EJ, McLaughlin PA, Thanassi DG. Francisella tularensis disrupts TLR2-MYD88-p38 signaling early during infection to delay apoptosis of macrophages and promote virulence in the host. mBio 2023; 14:e0113623. [PMID: 37404047 PMCID: PMC10470500 DOI: 10.1128/mbio.01136-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023] Open
Abstract
Francisella tularensis is a zoonotic pathogen and the causative agent of tularemia. F. tularensis replicates to high levels within the cytosol of macrophages and other host cells while subverting the host response to infection. Critical to the success of F. tularensis is its ability to delay macrophage apoptosis to maintain its intracellular replicative niche. However, the host-signaling pathway(s) modulated by F. tularensis to delay apoptosis are poorly characterized. The outer membrane channel protein TolC is required for F. tularensis virulence and its ability to suppress apoptosis and cytokine expression during infection of macrophages. We took advantage of the F. tularensis ∆tolC mutant phenotype to identify host pathways that are important for activating macrophage apoptosis and that are disrupted by the bacteria. Comparison of macrophages infected with wild-type or ∆tolC F. tularensis revealed that the bacteria interfere with TLR2-MYD88-p38 signaling at early times post infection to delay apoptosis, dampen innate host responses, and preserve the intracellular replicative niche. Experiments using the mouse pneumonic tularemia model confirmed the in vivo relevance of these findings, revealing contributions of TLR2 and MYD88 signaling to the protective host response to F. tularensis, which is modulated by the bacteria to promote virulence. IMPORTANCE Francisella tularensis is a Gram-negative intracellular bacterial pathogen and the causative agent of the zoonotic disease tularemia. F. tularensis, like other intracellular pathogens, modulates host-programmed cell death pathways to ensure its replication and survival. We previously identified the outer membrane channel protein TolC as required for the ability of F. tularensis to delay host cell death. However, the mechanism by which F. tularensis delays cell death pathways during intracellular replication is unclear despite being critical to pathogenesis. In the present study, we address this gap in knowledge by taking advantage of ∆tolC mutants of F. tularensis to uncover signaling pathways governing host apoptotic responses to F. tularensis and which are modulated by the bacteria during infection to promote virulence. These findings reveal mechanisms by which intracellular pathogens subvert host responses and enhance our understanding of the pathogenesis of tularemia.
Collapse
Affiliation(s)
- P. Todd Benziger
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Erik J. Kopping
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Patrick A. McLaughlin
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - David G. Thanassi
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
5
|
Guo Y, Mao R, Xie Q, Cheng X, Xu T, Wang X, Du Y, Qi X. Francisella novicida Mutant XWK4 Triggers Robust Inflammasome Activation Favoring Infection. Front Cell Dev Biol 2021; 9:743335. [PMID: 34869331 PMCID: PMC8637620 DOI: 10.3389/fcell.2021.743335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
Bacterial infection tendentiously triggers inflammasome activation, whereas the roles of inflammasome activation in host defense against diverse infections remain unclear. Here, we identified that an ASC-dependent inflammasome activation played opposite roles in host defense against Francisella novicida wild-type (WT) U112 and mutant strain XWK4. Comparing with U112, XWK4 infection induced robust cytokine production, ASC-dependent inflammasome activation, and pyroptosis. Both AIM2 and NLRP3 were involved and played independent roles in XWK4-induced inflammasome activation. Type II interferon was partially required for XWK4-triggered inflammasome activation, which was different from type I interferon dependency in U112-induced inflammasome activation. Distinct from F. novicida U112 and Acinetobacter baumannii infection, Asc-/- mice were more resistant than WT mice response to XWK4 infection by limiting bacterial burden in vivo. The excessive inflammasome activation triggered by XWK4 infection caused dramatical cell death and pathological damage. Our study offers novel insights into mechanisms of inflammasome activation in host defense and provides potential therapeutic approach against bacterial infections and inflammatory diseases.
Collapse
Affiliation(s)
- Yu Guo
- School of Life Sciences, University of Science and Technology of China, Hefei, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Rudi Mao
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qingqing Xie
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaojie Cheng
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiaoyuan Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yan Du
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Xiaopeng Qi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
6
|
Slater SL, Mavridou DAI. Harnessing the potential of bacterial oxidative folding to aid protein production. Mol Microbiol 2021; 116:16-28. [PMID: 33576091 DOI: 10.1111/mmi.14700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/09/2021] [Indexed: 11/30/2022]
Abstract
Protein folding is central to both biological function and recombinant protein production. In bacterial expression systems, which are easy to use and offer high protein yields, production of the protein of interest in its native fold can be hampered by the limitations of endogenous posttranslational modification systems. Disulfide bond formation, entailing the covalent linkage of proximal cysteine amino acids, is a fundamental posttranslational modification reaction that often underpins protein stability, especially in extracytoplasmic environments. When these bonds are not formed correctly, the yield and activity of the resultant protein are dramatically decreased. Although the mechanism of oxidative protein folding is well understood, unwanted or incorrect disulfide bond formation often presents a stumbling block for the expression of cysteine-containing proteins in bacteria. It is therefore important to consider the biochemistry of prokaryotic disulfide bond formation systems in the context of protein production, in order to take advantage of the full potential of such pathways in biotechnology applications. Here, we provide a critical overview of the use of bacterial oxidative folding in protein production so far, and propose a practical decision-making workflow for exploiting disulfide bond formation for the expression of any given protein of interest.
Collapse
Affiliation(s)
- Sabrina L Slater
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Despoina A I Mavridou
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
- John Ring LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
7
|
Cunningham AL, Mann BJ, Qin A, Santiago AE, Grassel C, Lipsky M, Vogel SN, Barry EM. Characterization of Schu S4 aro mutants as live attenuated tularemia vaccine candidates. Virulence 2021; 11:283-294. [PMID: 32241221 PMCID: PMC7161688 DOI: 10.1080/21505594.2020.1746557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
There is a need for development of an effective vaccine against Francisella tularensis, as this potential bioweapon has a high mortality rate and low infectious dose when delivered via the aerosol route. Moreover, this Tier 1 agent has a history of weaponization. We engineered targeted mutations in the Type A strain F. tularensis subspecies tularensis Schu S4 in aro genes encoding critical enzymes in aromatic amino acid biosynthesis. F. tularensis Schu S4ΔaroC, Schu S4ΔaroD, and Schu S4ΔaroCΔaroD mutant strains were attenuated for intracellular growth in vitro and for virulence in vivo and, conferred protection against pulmonary wild-type (WT) F. tularensis Schu S4 challenge in the C57BL/6 mouse model. F. tularensis Schu S4ΔaroD was identified as the most promising vaccine candidate, demonstrating protection against high-dose intranasal challenge; it protected against 1,000 CFU Schu S4, the highest level of protection tested to date. It also provided complete protection against challenge with 92 CFU of a F. tularensis subspecies holarctica strain (Type B). Mice responded to vaccination with Schu S4ΔaroD with systemic IgM and IgG2c, as well as the production of a functional T cell response as measured in the splenocyte-macrophage co-culture assay. This vaccine was further characterized for dissemination, histopathology, and cytokine/chemokine gene induction at defined time points following intranasal vaccination which confirmed its attenuation compared to WT Schu S4. Cytokine, chemokine, and antibody induction patterns compared to wild-type Schu S4 distinguish protective vs. pathogenic responses to F. tularensis and elucidate correlates of protection associated with vaccination against this agent.
Collapse
Affiliation(s)
| | - Barbara J Mann
- Department of Medicine, Division of Infectious Diseases and International Heath, University of Virginia, Charlottesville, VA, USA
| | - Aiping Qin
- Department of Medicine, Division of Infectious Diseases and International Heath, University of Virginia, Charlottesville, VA, USA
| | - Araceli E Santiago
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Christen Grassel
- Center for Vaccine Development, University of Maryland Baltimore, Baltimore, MD, USA
| | - Michael Lipsky
- Department of Pathology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Eileen M Barry
- Center for Vaccine Development, University of Maryland Baltimore, Baltimore, MD, USA
| |
Collapse
|
8
|
Zellner B, Mengin-Lecreulx D, Tully B, Gunning WT, Booth R, Huntley JF. A Francisella tularensis L,D-carboxypeptidase plays important roles in cell morphology, envelope integrity, and virulence. Mol Microbiol 2021; 115:1357-1378. [PMID: 33469978 DOI: 10.1111/mmi.14685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/15/2022]
Abstract
Francisella tularensis is a Gram-negative, intracellular bacterium that causes the zoonotic disease tularemia. Intracellular pathogens, including F. tularensis, have evolved mechanisms to survive in the harsh environment of macrophages and neutrophils, where they are exposed to cell envelope-damaging molecules. The bacterial cell wall, primarily composed of peptidoglycan (PG), maintains cell morphology, structure, and membrane integrity. Intracellular Gram-negative bacteria protect themselves from macrophage and neutrophil killing by recycling and repairing damaged PG--a process that involves over 50 different PG synthesis and recycling enzymes. Here, we identified a PG recycling enzyme, L,D-carboxypeptidase A (LdcA), of F. tularensis that is responsible for converting PG tetrapeptide stems to tripeptide stems. Unlike E. coli LdcA and most other orthologs, F. tularensis LdcA does not localize to the cytoplasm and also exhibits L,D-endopeptidase activity, converting PG pentapeptide stems to tripeptide stems. Loss of F. tularensis LdcA led to altered cell morphology and membrane integrity, as well as attenuation in a mouse pulmonary infection model and in primary and immortalized macrophages. Finally, an F. tularensis ldcA mutant protected mice against virulent Type A F. tularensis SchuS4 pulmonary challenge.
Collapse
Affiliation(s)
- Briana Zellner
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH, USA
| | - Dominique Mengin-Lecreulx
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Brenden Tully
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH, USA
| | | | - Robert Booth
- Department of Pathology, University of Toledo, Toledo, OH, USA
| | - Jason F Huntley
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH, USA
| |
Collapse
|
9
|
Bachert BA, Biryukov SS, Chua J, Rodriguez SA, Toothman RG, Cote CK, Klimko CP, Hunter M, Shoe JL, Williams JA, Kuehl KA, Biot FV, Bozue JA. A Francisella novicida Mutant, Lacking the Soluble Lytic Transglycosylase Slt, Exhibits Defects in Both Growth and Virulence. Front Microbiol 2019; 10:1343. [PMID: 31258523 PMCID: PMC6587636 DOI: 10.3389/fmicb.2019.01343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/29/2019] [Indexed: 01/28/2023] Open
Abstract
Francisella tularensis is the causative agent of tularemia and has gained recent interest as it poses a significant biothreat risk. F. novicida is commonly used as a laboratory surrogate for tularemia research due to genetic similarity and susceptibility of mice to infection. Currently, there is no FDA-approved tularemia vaccine, and identifying therapeutic targets remains a critical gap in strategies for combating this pathogen. Here, we investigate the soluble lytic transglycosylase or Slt in F. novicida, which belongs to a class of peptidoglycan-modifying enzymes known to be involved in cell division. We assess the role of Slt in biology and virulence of the organism as well as the vaccine potential of the slt mutant. We show that the F. novicida slt mutant has a significant growth defect in acidic pH conditions. Further microscopic analysis revealed significantly altered cell morphology compared to wild-type, including larger cell size, extensive membrane protrusions, and cell clumping and fusion, which was partially restored by growth in neutral pH or genetic complementation. Viability of the mutant was also significantly decreased during growth in acidic medium, but not at neutral pH. Furthermore, the slt mutant exhibited significant attenuation in a murine model of intranasal infection and virulence could be restored by genetic complementation. Moreover, we could protect mice using the slt mutant as a live vaccine strain against challenge with the parent strain; however, we were not able to protect against challenge with the fully virulent F. tularensis Schu S4 strain. These studies demonstrate a critical role for the Slt enzyme in maintaining proper cell division and morphology in acidic conditions, as well as replication and virulence in vivo. Our results suggest that although the current vaccination strategy with F. novicida slt mutant would not protect against Schu S4 challenges, the Slt enzyme could be an ideal target for future therapeutic development.
Collapse
Affiliation(s)
- Beth A Bachert
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Sergei S Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Jennifer Chua
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Sabrina A Rodriguez
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Ronald G Toothman
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Janice A Williams
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Kathleen A Kuehl
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Fabrice V Biot
- Unité de Bactériologie/UMR_MD1, Département de Biologie des Agents Transmissibles, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Joel A Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| |
Collapse
|
10
|
Wu CQ, Zhang T, Zhang W, Shi M, Tu F, Yu A, Li M, Yang M. Two DsbA Proteins Are Important for Vibrio parahaemolyticus Pathogenesis. Front Microbiol 2019; 10:1103. [PMID: 31156607 PMCID: PMC6531988 DOI: 10.3389/fmicb.2019.01103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/30/2019] [Indexed: 01/05/2023] Open
Abstract
Bacterial pathogens maintain disulfide bonds for protein stability and functions that are required for pathogenesis. Vibrio parahaemolyticus is a Gram-negative pathogen that causes food-borne gastroenteritis and is also an important opportunistic pathogen of aquatic animals. Two genes encoding the disulfide bond formation protein A, DsbA, are predicted to be encoded in the V. parahaemolyticus genome. DsbA plays an important role in Vibrio cholerae virulence but its role in V. parahaemolyticus is largely unknown. In this study, the activities and functions of the two V. parahaemolyticus DsbA proteins were characterized. The DsbAs affected virulence factor expression at the post-translational level. The protein levels of adhesion factor VpadF (VP1767) and the thermostable direct hemolysin (TDH) were significantly reduced in the dsbA deletion mutants. V. parahaemolyticus lacking dsbA also showed reduced attachment to Caco-2 cells, decreased β-hemolytic activity, and less toxicity to both zebrafish and HeLa cells. Our findings demonstrate that DsbAs contribute to V. parahaemolyticus pathogenesis.
Collapse
Affiliation(s)
- Chun-Qin Wu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou, China.,Department of Animal Science, Wenzhou Vocational College of Science and Technology, Wenzhou, China
| | - Ting Zhang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou, China
| | - Wenwen Zhang
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Mengting Shi
- College of Life Science, Nanjing Agricultural University, Nanjing, China
| | - Fei Tu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou, China
| | - Ai Yu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou, China
| | - Manman Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou, China
| | - Menghua Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou, China
| |
Collapse
|
11
|
Global Analysis of Genes Essential for Francisella tularensis Schu S4 Growth In Vitro and for Fitness during Competitive Infection of Fischer 344 Rats. J Bacteriol 2019; 201:JB.00630-18. [PMID: 30642993 PMCID: PMC6416918 DOI: 10.1128/jb.00630-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/02/2019] [Indexed: 01/02/2023] Open
Abstract
The highly virulent intracellular pathogen Francisella tularensis is a Gram-negative bacterium that has a wide host range, including humans, and is the causative agent of tularemia. To identify new therapeutic drug targets and vaccine candidates and investigate the genetic basis of Francisella virulence in the Fischer 344 rat, we have constructed an F. tularensis Schu S4 transposon library. This library consists of more than 300,000 unique transposon mutants and represents a transposon insertion for every 6 bp of the genome. A transposon-directed insertion site sequencing (TraDIS) approach was used to identify 453 genes essential for growth in vitro Many of these essential genes were mapped to key metabolic pathways, including glycolysis/gluconeogenesis, peptidoglycan synthesis, fatty acid biosynthesis, and the tricarboxylic acid (TCA) cycle. Additionally, 163 genes were identified as required for fitness during colonization of the Fischer 344 rat spleen. This in vivo selection screen was validated through the generation of marked deletion mutants that were individually assessed within a competitive index study against the wild-type F. tularensis Schu S4 strain.IMPORTANCE The intracellular bacterial pathogen Francisella tularensis causes a disease in humans characterized by the rapid onset of nonspecific symptoms such as swollen lymph glands, fever, and headaches. F. tularensis is one of the most infectious bacteria known and following pulmonary exposure can have a mortality rate exceeding 50% if left untreated. The low infectious dose of this organism and concerns surrounding its potential as a biological weapon have heightened the need for effective and safe therapies. To expand the repertoire of targets for therapeutic development, we initiated a genome-wide analysis. This study has identified genes that are important for F. tularensis under in vitro and in vivo conditions, providing candidates that can be evaluated for vaccine or antibacterial development.
Collapse
|
12
|
O’Malley KJ, Bowling JL, Stinson E, Cole KS, Mann BJ, Namjoshi P, Hazlett KRO, Barry EM, Reed DS. Aerosol prime-boost vaccination provides strong protection in outbred rabbits against virulent type A Francisella tularensis. PLoS One 2018; 13:e0205928. [PMID: 30346998 PMCID: PMC6197691 DOI: 10.1371/journal.pone.0205928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
Tularemia, also known as rabbit fever, is a severe zoonotic disease in humans caused by the gram-negative bacterium Francisella tularensis (Ft). While there have been a number of attempts to develop a vaccine for Ft, few candidates have advanced beyond experiments in inbred mice. We report here that a prime-boost strategy with aerosol delivery of recombinant live attenuated candidate Ft S4ΔaroD offers significant protection (83% survival) in an outbred animal model, New Zealand White rabbits, against aerosol challenge with 248 cfu (11 LD50) of virulent type A Ft SCHU S4. Surviving rabbits given two doses of the attenuated strains by aerosol did not exhibit substantial post-challenge fevers, changes in erythrocyte sedimentation rate or in complete blood counts. At a higher challenge dose (3,186 cfu; 139 LD50), protection was still good with 66% of S4ΔaroD-vaccinated rabbits surviving while 50% of S4ΔguaBA vaccinated rabbits also survived challenge. Pre-challenge plasma IgG titers against Ft SCHU S4 corresponded with survival time after challenge. Western blot analysis found that plasma antibody shifted from predominantly targeting Ft O-antigen after the prime vaccination to other antigens after the boost. These results demonstrate the superior protection conferred by a live attenuated derivative of virulent F. tularensis, particularly when given in an aerosol prime-boost regimen.
Collapse
Affiliation(s)
- Katherine J. O’Malley
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jennifer L. Bowling
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Elizabeth Stinson
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kelly S. Cole
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Barbara J. Mann
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, United States of America
| | - Prachi Namjoshi
- Department for Immunology & Microbial Diseases, Albany Medical College, Albany, NY, United States of America
| | - Karsten R. O. Hazlett
- Department for Immunology & Microbial Diseases, Albany Medical College, Albany, NY, United States of America
| | - Eileen M. Barry
- Center for Vaccine Development, University of Maryland Baltimore, Baltimore, MD, United States of America
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
13
|
Champion AE, Bandara AB, Mohapatra N, Fulton KM, Twine SM, Inzana TJ. Further Characterization of the Capsule-Like Complex (CLC) Produced by Francisella tularensis Subspecies tularensis: Protective Efficacy and Similarity to Outer Membrane Vesicles. Front Cell Infect Microbiol 2018; 8:182. [PMID: 29963499 PMCID: PMC6013578 DOI: 10.3389/fcimb.2018.00182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/09/2018] [Indexed: 11/17/2022] Open
Abstract
Francisella tularensis is the etiologic agent of tularemia, and subspecies tularensis (type A) is the most virulent subspecies. The live vaccine strain (LVS) of subspecies holarctica produces a capsule-like complex (CLC) that consists of a large variety of glycoproteins. Expression of the CLC is greatly enhanced when the bacteria are subcultured in and grown on chemically defined medium. Deletion of two genes responsible for CLC glycosylation in LVS results in an attenuated mutant that is protective against respiratory tularemia in a mouse model. We sought to further characterize the CLC composition and to determine if a type A CLC glycosylation mutant would be attenuated in mice. The CLCs isolated from LVS extracted with 0.5% phenol or 1 M urea were similar, as determined by gel electrophoresis and Western blotting, but the CLC extracted with urea was more water-soluble. The CLC extracted with either 0.5% phenol or 1 M urea from type A strains was also similar to the CLC of LVS in antigenic properties, electrophoretic profile, and by transmission electron microscopy (TEM). The solubility of the CLC could be further enhanced by fractionation with Triton X-114 followed by N-Lauroylsarcosine detergents; the largest (>250 kDa) molecular size component appeared to be an aggregate of smaller components. Outer membrane vesicles/tubules (OMV/T) isolated by differential centrifugation and micro-filtration appeared similar to the CLC by TEM, and many of the proteins present in the OMV/T were also identified in soluble and insoluble fractions of the CLC. Further investigation is warranted to assess the relationship between OMV/T and the CLC. The CLC conjugated to keyhole limpet hemocyanin or flagellin was highly protective against high-dose LVS intradermal challenge and partially protective against intranasal challenge. A protective response was associated with a significant rise in cytokines IL-12, IL-10, and IFN-γ. However, a type A CLC glycosylation mutant remained virulent in BALB/c mice, and immunization with the CLC did not protect mice against high dose respiratory challenge with type A strain SCHU S4.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Capsules/genetics
- Bacterial Capsules/immunology
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Flagellin/genetics
- Flagellin/immunology
- Francisella tularensis/genetics
- Francisella tularensis/metabolism
- Francisella tularensis/pathogenicity
- Genes, Bacterial/genetics
- Glycoproteins/genetics
- Glycoproteins/immunology
- Glycoproteins/isolation & purification
- Hemocyanins/genetics
- Hemocyanins/immunology
- Interferon-gamma/metabolism
- Interleukin-10/metabolism
- Interleukin-12/metabolism
- Mice, Inbred BALB C
- Mutagenesis
- Sequence Deletion
- Tularemia/immunology
- Tularemia/prevention & control
- Vaccination
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Conjugate/genetics
- Vaccines, Conjugate/immunology
- Virulence Factors/genetics
- Virulence Factors/immunology
Collapse
Affiliation(s)
- Anna E. Champion
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Aloka B. Bandara
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Nrusingh Mohapatra
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Kelly M. Fulton
- Institute for Biological Sciences, National Research Council Canada, Ottawa, ON, Canada
| | - Susan M. Twine
- Institute for Biological Sciences, National Research Council Canada, Ottawa, ON, Canada
| | - Thomas J. Inzana
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|
14
|
Jia Q, Horwitz MA. Live Attenuated Tularemia Vaccines for Protection Against Respiratory Challenge With Virulent F. tularensis subsp. tularensis. Front Cell Infect Microbiol 2018; 8:154. [PMID: 29868510 PMCID: PMC5963219 DOI: 10.3389/fcimb.2018.00154] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/24/2018] [Indexed: 12/11/2022] Open
Abstract
Francisella tularensis is the causative agent of tularemia and a Tier I bioterrorism agent. In the 1900s, several vaccines were developed against tularemia including the killed "Foshay" vaccine, subunit vaccines comprising F. tularensis protein(s) or lipoproteins(s) in an adjuvant formulation, and the F. tularensis Live Vaccine Strain (LVS); none were licensed in the U.S.A. or European Union. The LVS vaccine retains toxicity in humans and animals-especially mice-but has demonstrated efficacy in humans, and thus serves as the current gold standard for vaccine efficacy studies. The U.S.A. 2001 anthrax bioterrorism attack spawned renewed interest in vaccines against potential biowarfare agents including F. tularensis. Since live attenuated-but not killed or subunit-vaccines have shown promising efficacy and since vaccine efficacy against respiratory challenge with less virulent subspecies holarctica or F. novicida, or against non-respiratory challenge with virulent subsp. tularensis (Type A) does not reliably predict vaccine efficacy against respiratory challenge with virulent subsp. tularensis, the route of transmission and species of greatest concern in a bioterrorist attack, in this review, we focus on live attenuated tularemia vaccine candidates tested against respiratory challenge with virulent Type A strains, including homologous vaccines derived from mutants of subsp. holarctica, F. novicida, and subsp. tularensis, and heterologous vaccines developed using viral or bacterial vectors to express F. tularensis immunoprotective antigens. We compare the virulence and efficacy of these vaccine candidates with that of LVS and discuss factors that can significantly impact the development and evaluation of live attenuated tularemia vaccines. Several vaccines meet what we would consider the minimum criteria for vaccines to go forward into clinical development-safety greater than LVS and efficacy at least as great as LVS, and of these, several meet the higher standard of having efficacy ≥LVS in the demanding mouse model of tularemia. These latter include LVS with deletions in purMCD, sodBFt , capB or wzy; LVS ΔcapB that also overexpresses Type VI Secretion System (T6SS) proteins; FSC200 with a deletion in clpB; the single deletional purMCD mutant of F. tularensis SCHU S4, and a heterologous prime-boost vaccine comprising LVS ΔcapB and Listeria monocytogenes expressing T6SS proteins.
Collapse
Affiliation(s)
- Qingmei Jia
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, 37-121 Center for Health Sciences, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Virulence of the Melioidosis Pathogen Burkholderia pseudomallei Requires the Oxidoreductase Membrane Protein DsbB. Infect Immun 2018; 86:IAI.00938-17. [PMID: 29440370 PMCID: PMC5913862 DOI: 10.1128/iai.00938-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/04/2018] [Indexed: 12/26/2022] Open
Abstract
The naturally antibiotic-resistant bacterium Burkholderia pseudomallei is the causative agent of melioidosis, a disease with stubbornly high mortality and a complex, protracted treatment regimen. The worldwide incidence of melioidosis is likely grossly underreported, though it is known to be highly endemic in northern Australia and Southeast Asia. Bacterial disulfide bond (DSB) proteins catalyze the oxidative folding and isomerization of disulfide bonds in substrate proteins. In the present study, we demonstrate that B. pseudomallei membrane protein disulfide bond protein B (BpsDsbB) forms a functional redox relay with the previously characterized virulence mediator B. pseudomallei disulfide bond protein A (BpsDsbA). Genomic analysis of diverse B. pseudomallei clinical isolates demonstrated that dsbB is a highly conserved core gene. Critically, we show that DsbB is required for virulence in B. pseudomallei. A panel of B. pseudomalleidsbB deletion strains (K96243, 576, MSHR2511, MSHR0305b, and MSHR5858) were phenotypically diverse according to the results of in vitro assays that assess hallmarks of virulence. Irrespective of their in vitro virulence phenotypes, two deletion strains were attenuated in a BALB/c mouse model of infection. A crystal structure of a DsbB-derived peptide complexed with BpsDsbA provides the first molecular characterization of their interaction. This work contributes to our broader understanding of DSB redox biology and will support the design of antimicrobial drugs active against this important family of bacterial virulence targets.
Collapse
|
16
|
Landeta C, Boyd D, Beckwith J. Disulfide bond formation in prokaryotes. Nat Microbiol 2018; 3:270-280. [PMID: 29463925 DOI: 10.1038/s41564-017-0106-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 12/21/2017] [Indexed: 12/25/2022]
Abstract
Interest in protein disulfide bond formation has recently increased because of the prominent role of disulfide bonds in bacterial virulence and survival. The first discovered pathway that introduces disulfide bonds into cell envelope proteins consists of Escherichia coli enzymes DsbA and DsbB. Since its discovery, variations on the DsbAB pathway have been found in bacteria and archaea, probably reflecting specific requirements for survival in their ecological niches. One variation found amongst Actinobacteria and Cyanobacteria is the replacement of DsbB by a homologue of human vitamin K epoxide reductase. Many Gram-positive bacteria express enzymes involved in disulfide bond formation that are similar, but non-homologous, to DsbAB. While bacterial pathways promote disulfide bond formation in the bacterial cell envelope, some archaeal extremophiles express proteins with disulfide bonds both in the cytoplasm and in the extra-cytoplasmic space, possibly to stabilize proteins in the face of extreme conditions, such as growth at high temperatures. Here, we summarize the diversity of disulfide-bond-catalysing systems across prokaryotic lineages, discuss examples for understanding the biological basis of such systems, and present perspectives on how such systems are enabling advances in biomedical engineering and drug development.
Collapse
Affiliation(s)
- Cristina Landeta
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Dana Boyd
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Jon Beckwith
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Zinc Acquisition Mechanisms Differ between Environmental and Virulent Francisella Species. J Bacteriol 2018; 200:JB.00587-17. [PMID: 29109188 PMCID: PMC5786701 DOI: 10.1128/jb.00587-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/27/2017] [Indexed: 12/29/2022] Open
Abstract
Zinc is an essential nutrient for bacterial growth. Because host cells can restrict pathogen access to zinc as an antimicrobial defense mechanism, intracellular pathogens such as Francisella must sense their environment and acquire zinc in response. In many bacteria, the conserved transcription factor Zur is a key regulator of zinc acquisition. To identify mechanisms of zinc uptake in Francisella novicida U112, transcriptome sequencing of wild-type and putative zur mutant bacteria was performed. Only three genes were confirmed as directly regulated by Zur and zinc limitation by quantitative reverse transcription-PCR. One of these genes, FTN_0879, is predicted to encode a protein with similarity to the zupT family of zinc transporters, which are not typically regulated by Zur. While a putative znuACB operon encoding a high-affinity zinc transporter was identified in U112, expression of this operon was not controlled by Zur or zinc concentration. Disruption of zupT but not znuA in U112 impaired growth under zinc limitation, suggesting that ZupT is the primary mechanism of zinc acquisition under these conditions. In the virulent Francisella tularensis subsp. tularensis Schu S4 strain, zupT is a pseudogene, and attempts to delete znuA were unsuccessful, suggesting that it is essential in this strain. A reverse TetR repression system was used to knock down the expression of znuA in Schu S4, revealing that znuA is required for growth under zinc limitation and contributes to intracellular growth within macrophages. Overall, this work identifies genes necessary for adaptation to zinc limitation and highlights nutritional differences between environmental and virulent Francisella strains. IMPORTANCEFrancisella tularensis is a tier 1 select agent with a high potential for lethality and no approved vaccine. A better understanding of Francisella virulence factors is required for the development of therapeutics. While acquisition of zinc has been shown to be required for the virulence of numerous intracellular pathogens, zinc uptake has not been characterized in Francisella. This work characterizes the Zur regulon in F. novicida and identifies two transporters that contribute to bacterial growth under zinc limitation. In addition, these data identify differences in mechanisms of zinc uptake and tolerance to zinc limitation between F. tularensis and F. novicida, highlighting the role of znuA in the growth of Schu S4 under zinc limitation.
Collapse
|
18
|
Fabrik I, Link M, Putzova D, Plzakova L, Lubovska Z, Philimonenko V, Pavkova I, Rehulka P, Krocova Z, Hozak P, Santic M, Stulik J. The Early Dendritic Cell Signaling Induced by Virulent Francisella tularensis Strain Occurs in Phases and Involves the Activation of Extracellular Signal-Regulated Kinases (ERKs) and p38 In the Later Stage. Mol Cell Proteomics 2017; 17:81-94. [PMID: 29046388 DOI: 10.1074/mcp.ra117.000160] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/22/2017] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) infected by Francisella tularensis are poorly activated and do not undergo classical maturation process. Although reasons of such unresponsiveness are not fully understood, their impact on the priming of immunity is well appreciated. Previous attempts to explain the behavior of Francisella-infected DCs were hypothesis-driven and focused on events at later stages of infection. Here, we took an alternative unbiased approach by applying methods of global phosphoproteomics to analyze the dynamics of cell signaling in primary DCs during the first hour of infection by Francisella tularensis Presented results show that the early response of DCs to Francisella occurs in phases and that ERK and p38 signaling modules induced at the later stage are differentially regulated by virulent and attenuated ΔdsbA strain. These findings imply that the temporal orchestration of host proinflammatory pathways represents the integral part of Francisella life-cycle inside hijacked DCs.
Collapse
Affiliation(s)
- Ivo Fabrik
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Marek Link
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Daniela Putzova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Lenka Plzakova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Lubovska
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic
| | - Vlada Philimonenko
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic.,¶Institute of Molecular Genetics ASCR v.v.i., Department of Biology of the Cell Nucleus, 142 20 Prague 4, Czech Republic
| | - Ivona Pavkova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Pavel Rehulka
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Krocova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Pavel Hozak
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic.,¶Institute of Molecular Genetics ASCR v.v.i., Department of Biology of the Cell Nucleus, 142 20 Prague 4, Czech Republic
| | - Marina Santic
- ‖Department of Microbiology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Jiri Stulik
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic;
| |
Collapse
|
19
|
Kolappan S, Lo KY, Shen CLJ, Guttman JA, Craig L. Structure of the conserved Francisella virulence protein FvfA. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2017; 73:814-821. [PMID: 28994410 DOI: 10.1107/s205979831701333x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/18/2017] [Indexed: 11/10/2022]
Abstract
Francisella tularensis is a potent human pathogen that invades and survives in macrophage and epithelial cells. Two identical proteins, FTT_0924 from F. tularensis subsp. tularensis and FTL_1286 from F. tularensis subsp. holarctica LVS, have previously been identified as playing a role in protection of the bacteria from osmotic shock and its survival in macrophages. FTT_0924 has been shown to localize to the inner membrane, with its C-terminus exposed to the periplasm. Here, crystal structures of the F. novicida homologue FTN_0802, which we call FvfA, in two crystal forms are reported at 1.8 Å resolution. FvfA differs from FTT_0924 and FTL_1286 by a single amino acid. FvfA has a DUF1471 fold that closely resembles the Escherichia coli outer membrane lipoprotein RscF, a component of a phosphorelay pathway involved in protecting bacteria from outer membrane perturbation. The structural and functional similarities and differences between these proteins and their implications for F. tularensis pathogenesis are discussed.
Collapse
Affiliation(s)
- Subramania Kolappan
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Karen Y Lo
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Chiao Ling Jennifer Shen
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Julian A Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Lisa Craig
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
20
|
Putzova D, Panda S, Härtlova A, Stulík J, Gekara NO. Subversion of innate immune responses by Francisella involves the disruption of TRAF3 and TRAF6 signalling complexes. Cell Microbiol 2017; 19. [PMID: 28745813 DOI: 10.1111/cmi.12769] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 07/20/2017] [Accepted: 07/21/2017] [Indexed: 12/13/2022]
Abstract
The success of pathogens depends on their ability to circumvent immune defences. Francisella tularensis is one of the most infectious bacteria known. The remarkable virulence of Francisella is believed to be due to its capacity to evade or subvert the immune system, but how remains obscure. Here, we show that Francisella triggers but concomitantly inhibits the Toll-like receptor, RIG-I-like receptor, and cytoplasmic DNA pathways. Francisella subverts these pathways at least in part by inhibiting K63-linked polyubiquitination and assembly of TRAF6 and TRAF3 complexes that control the transcriptional responses of pattern recognition receptors. We show that this mode of inhibition requires a functional type VI secretion system and/or the presence of live bacteria in the cytoplasm. The ability of Francisella to enter the cytosol while simultaneously inhibiting multiple pattern recognition receptor pathways may account for the notable capacity of this bacterium to invade and proliferate in the host without evoking a self-limiting innate immune response.
Collapse
Affiliation(s)
- Daniela Putzova
- Department of Molecular Biology, The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden.,Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Hradec Králové, Czech Republic
| | - Swarupa Panda
- Department of Molecular Biology, The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Anetta Härtlova
- Department of Molecular Biology, The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Jiří Stulík
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Hradec Králové, Czech Republic
| | - Nelson O Gekara
- Department of Molecular Biology, The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| |
Collapse
|
21
|
Putzova D, Senitkova I, Stulik J. Tularemia vaccines. Folia Microbiol (Praha) 2016; 61:495-504. [PMID: 27194547 DOI: 10.1007/s12223-016-0461-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/01/2016] [Indexed: 10/21/2022]
Abstract
Francisella tularensis is the causative agent of the potentially lethal disease tularemia. Due to a low infectious dose and ease of airborne transmission, Francisella is classified as a category A biological agent. Despite the possible risk to public health, there is no safe and fully licensed vaccine. A potential vaccine candidate, an attenuated live vaccine strain, does not fulfil the criteria for general use. In this review, we will summarize existing and new candidates for live attenuated and subunit vaccines.
Collapse
Affiliation(s)
- Daniela Putzova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Iva Senitkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jiri Stulik
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic.
| |
Collapse
|
22
|
Xue Y, Tu F, Shi M, Wu CQ, Ren G, Wang X, Fang W, Song H, Yang M. Redox pathway sensing bile salts activates virulence gene expression in Vibrio cholerae. Mol Microbiol 2016; 102:909-924. [PMID: 27610607 DOI: 10.1111/mmi.13497] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2016] [Indexed: 01/05/2023]
Abstract
Vibrio cholerae, the causative agent of the severe diarrheal disease cholera, has evolved signal transduction systems to control co-ordinately the expression of virulence determinants. It was previously shown that the presence of the bile salts glycocholate and taurocholate in the small intestine causes dimerization of the transmembrane transcription factor TcpP by inducing intermolecular disulphide bonds in the TcpP periplasmic domain. In this study, they further investigated the mechanism of how taurocholate affects V. cholerae virulence determinants. In vitro assay of TcpP oxidation by VcDsbA showed that VcDsbA induced TcpP dimerization in the presence of taurocholate. Taurocholate bound to VcDsbA with a KD of 40 ± 2.5 μM, and also bound other Dsb proteins, including EcDsbA, EcDsbC and VcDsbC. Taurocholate inhibited VcDsbA reductase activity without affecting VcDsbA secondary structure or thermostability. VcDsbA and its substrates were more extensively reduced in the presence of taurocholate, as compared with their redox state in the absence of taurocholate. The data presented here not only provide new insights into the mechanism by which bile salts induce V. cholerae virulence but also suggest a means by which to develop inhibitors against DsbA.
Collapse
Affiliation(s)
- Yuanyuan Xue
- College of Animal Science & Technology, China-Australia Joint-Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang A&F University, Lin'an 311300, Zhejiang Province, China
| | - Fei Tu
- College of Animal Science & Technology, China-Australia Joint-Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang A&F University, Lin'an 311300, Zhejiang Province, China
| | - Mengting Shi
- College of Animal Science & Technology, China-Australia Joint-Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang A&F University, Lin'an 311300, Zhejiang Province, China
| | - Chun-Qin Wu
- College of Animal Science & Technology, China-Australia Joint-Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang A&F University, Lin'an 311300, Zhejiang Province, China.,Department of Animal Science, Wenzhou Vocational College of Science and Technology, Wenzhou, Zhejiang, China
| | - Guoping Ren
- New England Biolabs Inc, Ipswich, Massachusetts, USA
| | - Xiaojie Wang
- College of Animal Science & Technology, China-Australia Joint-Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang A&F University, Lin'an 311300, Zhejiang Province, China
| | - Weihuan Fang
- Institute of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Houhui Song
- College of Animal Science & Technology, China-Australia Joint-Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang A&F University, Lin'an 311300, Zhejiang Province, China
| | - Menghua Yang
- College of Animal Science & Technology, China-Australia Joint-Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang A&F University, Lin'an 311300, Zhejiang Province, China
| |
Collapse
|
23
|
Abstract
Francisella tularensis (Ft) is a gram-negative intercellular pathogen and category A biothreat agent. However, despite 15 years of strong government investment and intense research focused on the development of a US Food and Drug Administration-approved vaccine against Ft, the primary goal remains elusive. This article reviews research efforts focused on developing an Ft vaccine, as well as a number of important factors, some only recently recognized as such, which can significantly impact the development and evaluation of Ft vaccine efficacy. Finally, an assessment is provided as to whether a US Food and Drug Administration-approved Ft vaccine is likely to be forthcoming and the potential means by which this might be achieved.
Collapse
Affiliation(s)
- Raju Sunagar
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Sudeep Kumar
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Brian J Franz
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Edmund J Gosselin
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| |
Collapse
|
24
|
Qin A, Zhang Y, Clark ME, Moore EA, Rabideau MM, Moreau GB, Mann BJ. Components of the type six secretion system are substrates of Francisella tularensis Schu S4 DsbA-like FipB protein. Virulence 2016; 7:882-894. [PMID: 27028889 PMCID: PMC5160417 DOI: 10.1080/21505594.2016.1168550] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
FipB, an essential virulence factor in the highly virulent Schu S4 strain of F. tularensis subsp. tularensis, shares sequence similarity with Disulfide Bond formation (Dsb) proteins, which can have oxidoreductase, isomerase, or chaperone activity. To further explore FipB's role in virulence potential substrates were identified by co-purification and 2D gel electrophoresis, followed by protein sequencing using mass spectrometry. A total of 119 potential substrates were identified. Proteins with predicted enzymatic activity were prevalent, and there were 19 proteins that had been previously identified as impacting virulence. Among the potential substrates were IglC, IglB, and PdpB, three components of the Francisella Type Six Secretion System (T6SS), which is also essential for virulence. T6SS are widespread in Gram-negative pathogens, but have not been reported to be dependent on Dsb-like proteins for assembly or function. The presented results suggest that FipB affects IglB and IglC substrates differently. In a fipB mutant there were differences in free sulfhydryl accessibility of IglC, but not IglB, when compared to wild-type bacteria. However, for both proteins FipB appears to act as a chaperone that facilitates proper folding and conformation. Understanding the role FipB plays the assembly and structure in this T6SS may reveal critical aspects of assembly that are common and novel among this widely distributed class of secretion systems.
Collapse
Affiliation(s)
- Aiping Qin
- a Department of Medicine , Division of Infectious Diseases, University of Virginia , Charlottesville , VA , USA
| | - Yan Zhang
- a Department of Medicine , Division of Infectious Diseases, University of Virginia , Charlottesville , VA , USA
| | - Melinda E Clark
- a Department of Medicine , Division of Infectious Diseases, University of Virginia , Charlottesville , VA , USA
| | - Emily A Moore
- a Department of Medicine , Division of Infectious Diseases, University of Virginia , Charlottesville , VA , USA
| | - Meaghan M Rabideau
- a Department of Medicine , Division of Infectious Diseases, University of Virginia , Charlottesville , VA , USA
| | - G Brett Moreau
- a Department of Medicine , Division of Infectious Diseases, University of Virginia , Charlottesville , VA , USA
| | - Barbara J Mann
- a Department of Medicine , Division of Infectious Diseases, University of Virginia , Charlottesville , VA , USA
| |
Collapse
|
25
|
Lo KY, Visram S, Vogl AW, Shen CLJ, Guttman JA. Morphological analysis of Francisella novicida epithelial cell infections in the absence of functional FipA. Cell Tissue Res 2016; 363:449-59. [PMID: 26239909 DOI: 10.1007/s00441-015-2246-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 06/22/2015] [Indexed: 12/16/2022]
Abstract
Francisella novicida is a surrogate pathogen commonly used to study infections by the potential bioterrorism agent, Francisella tularensis. One of the primary sites of Francisella infections is the liver where >90% of infected cells are hepatocytes. It is known that once Francisella enter cells it occupies a membrane-bound compartment, the Francisella-containing vacuole (FCV), from which it rapidly escapes to replicate in the cytosol. Recent work examining the Francisella disulfide bond formation (Dsb) proteins, FipA and FipB, have demonstrated that these proteins are important during the Francisella infection process; however, details as to how the infections are altered in epithelial cells have remained elusive. To identify the stage of the infections where these Dsbs might act during epithelial infections, we exploited a hepatocyte F. novicida infection model that we recently developed. We found that F. novicida ΔfipA-infected hepatocytes contained bacteria clustered within lysosome-associated membrane protein 1-positive FCVs, suggesting that FipA is involved in the escape of F. novicida from its vacuole. Our morphological evidence provides a tangible link as to how Dsb FipA can influence Francisella infections.
Collapse
Affiliation(s)
- Karen Y Lo
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| | - Shyanne Visram
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| | - A Wayne Vogl
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, British Columbia, Canada
| | - Chiao Ling Jennifer Shen
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| | - Julian A Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada.
| |
Collapse
|
26
|
Dankova V, Balonova L, Link M, Straskova A, Sheshko V, Stulik J. Inactivation of Francisella tularensis Gene Encoding Putative ABC Transporter Has a Pleiotropic Effect upon Production of Various Glycoconjugates. J Proteome Res 2016; 15:510-24. [DOI: 10.1021/acs.jproteome.5b00864] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Vera Dankova
- Department
of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove 500 01, Czech Republic
| | - Lucie Balonova
- Department
of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove 500 01, Czech Republic
| | - Marek Link
- Department
of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove 500 01, Czech Republic
| | - Adela Straskova
- Department
of Phototrophic Microorganisms, Institute of Microbiology, The Academy of Sciences of The Czech Republic, Trebon 379 81, Czech Republic
| | - Valeria Sheshko
- Department
of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove 500 01, Czech Republic
| | - Jiri Stulik
- Department
of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove 500 01, Czech Republic
| |
Collapse
|
27
|
Rowe HM, Huntley JF. From the Outside-In: The Francisella tularensis Envelope and Virulence. Front Cell Infect Microbiol 2015; 5:94. [PMID: 26779445 PMCID: PMC4688374 DOI: 10.3389/fcimb.2015.00094] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022] Open
Abstract
Francisella tularensis is a highly-infectious bacterium that causes the rapid, and often lethal disease, tularemia. Many studies have been performed to identify and characterize the virulence factors that F. tularensis uses to infect a wide variety of hosts and host cell types, evade immune defenses, and induce severe disease and death. This review focuses on the virulence factors that are present in the F. tularensis envelope, including capsule, LPS, outer membrane, periplasm, inner membrane, secretion systems, and various molecules in each of aforementioned sub-compartments. Whereas, no single bacterial molecule or molecular complex single-handedly controls F. tularensis virulence, we review here how diverse bacterial systems work in conjunction to subvert the immune system, attach to and invade host cells, alter phagosome/lysosome maturation pathways, replicate in host cells without being detected, inhibit apoptosis, and induce host cell death for bacterial release and infection of adjacent cells. Given that the F. tularensis envelope is the outermost layer of the bacterium, we highlight herein how many of these molecules directly interact with the host to promote infection and disease. These and future envelope studies are important to advance our collective understanding of F. tularensis virulence mechanisms and offer targets for future vaccine development efforts.
Collapse
Affiliation(s)
- Hannah M Rowe
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Jason F Huntley
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| |
Collapse
|
28
|
Brissac T, Ziveri J, Ramond E, Tros F, Kock S, Dupuis M, Brillet M, Barel M, Peyriga L, Cahoreau E, Charbit A. Gluconeogenesis, an essential metabolic pathway for pathogenic Francisella. Mol Microbiol 2015; 98:518-34. [PMID: 26192619 DOI: 10.1111/mmi.13139] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2015] [Indexed: 01/23/2023]
Abstract
Intracellular multiplication and dissemination of the infectious bacterial pathogen Francisella tularensis implies the utilization of multiple host-derived nutrients. Here, we demonstrate that gluconeogenesis constitutes an essential metabolic pathway in Francisella pathogenesis. Indeed, inactivation of gene glpX, encoding the unique fructose 1,6-bisphosphatase of Francisella, severely impaired bacterial intracellular multiplication when cells were supplemented by gluconeogenic substrates such as glycerol or pyruvate. The ΔglpX mutant also showed a severe virulence defect in the mouse model, confirming the importance of this pathway during the in vivo life cycle of the pathogen. Isotopic profiling revealed the major role of the Embden-Meyerhof (glycolysis) pathway in glucose catabolism in Francisella and confirmed the importance of glpX in gluconeogenesis. Altogether, the data presented suggest that gluconeogenesis allows Francisella to cope with the limiting glucose availability it encounters during its infectious cycle by relying on host amino acids. Hence, targeting the gluconeogenic pathway might constitute an interesting therapeutic approach against this pathogen.
Collapse
Affiliation(s)
- Terry Brissac
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Jason Ziveri
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Elodie Ramond
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Fabiola Tros
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Stephanie Kock
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Marion Dupuis
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Magali Brillet
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Monique Barel
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| | - Lindsay Peyriga
- Université de Toulouse, INSA, UPS, INP, LISBP, 135 Avenue de Rangueil, Toulouse, 31077, France.,INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, Toulouse, 31400, France.,CNRS, UMR5504, Toulouse, 31400, France
| | - Edern Cahoreau
- Université de Toulouse, INSA, UPS, INP, LISBP, 135 Avenue de Rangueil, Toulouse, 31077, France.,INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, Toulouse, 31400, France.,CNRS, UMR5504, Toulouse, 31400, France
| | - Alain Charbit
- Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades, Equipe 11: Pathogénie des Infections Systémiques, Paris, France
| |
Collapse
|
29
|
Straskova A, Spidlova P, Mou S, Worsham P, Putzova D, Pavkova I, Stulik J. Francisella tularensis type B ΔdsbA mutant protects against type A strain and induces strong inflammatory cytokine and Th1-like antibody response in vivo. Pathog Dis 2015; 73:ftv058. [PMID: 26253078 DOI: 10.1093/femspd/ftv058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2015] [Indexed: 11/13/2022] Open
Abstract
Francisella tularensis subspecies tularensis is a highly virulent intracellular bacterial pathogen, causing the disease tularemia. However, a safe and effective vaccine for routine application against F. tularensis has not yet been developed. We have recently constructed the deletion mutants for the DsbA homolog protein (ΔdsbA/FSC200) and a hypothetical protein IglH (ΔiglH/FSC200) in the type B F. tularensis subsp. holarctica FSC200 strain, which exerted different protection capacity against parental virulent strain. In this study, we further investigated the immunological correlates for these different levels of protection provided by ΔdsbA/FSC200 and ΔiglH/FSC200 mutants. Our results show that ΔdsbA/FSC200 mutant, but not ΔiglH/FSC200 mutant, induces an early innate inflammatory response leading to strong Th1-like antibody response. Furthermore, vaccination with ΔdsbA/FSC200 mutant, but not with ΔiglH/FSC200, elicited protection against the subsequent challenge with type A SCHU S4 strain in mice. An immunoproteomic approach was used to map a spectrum of antigens targeted by Th1-like specific antibodies, and more than 80 bacterial antigens, including novel ones, were identified. Comparison of tularemic antigens recognized by the ΔdsbA/FSC200 post-vaccination and the SCHU S4 post-challenge sera then revealed the existence of 22 novel SCHU S4 specific antibody clones.
Collapse
Affiliation(s)
- Adela Straskova
- Department of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Trebesska 1575, Hradec Kralove 500 01, Czech Republic
| | - Petra Spidlova
- Department of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Trebesska 1575, Hradec Kralove 500 01, Czech Republic
| | - Sherry Mou
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702-5011, USA
| | - Patricia Worsham
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702-5011, USA
| | - Daniela Putzova
- Department of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Trebesska 1575, Hradec Kralove 500 01, Czech Republic
| | - Ivona Pavkova
- Department of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Trebesska 1575, Hradec Kralove 500 01, Czech Republic
| | - Jiri Stulik
- Department of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Trebesska 1575, Hradec Kralove 500 01, Czech Republic
| |
Collapse
|
30
|
Plzakova L, Krocova Z, Kubelkova K, Macela A. Entry of Francisella tularensis into Murine B Cells: The Role of B Cell Receptors and Complement Receptors. PLoS One 2015; 10:e0132571. [PMID: 26161475 PMCID: PMC4498600 DOI: 10.1371/journal.pone.0132571] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/16/2015] [Indexed: 01/06/2023] Open
Abstract
Francisella tularensis, the etiological agent of tularemia, is an intracellular pathogen that dominantly infects and proliferates inside phagocytic cells but can be seen also in non-phagocytic cells, including B cells. Although protective immunity is known to be almost exclusively associated with the type 1 pathway of cellular immunity, a significant role of B cells in immune responses already has been demonstrated. Whether their role is associated with antibody-dependent or antibody-independent B cell functions is not yet fully understood. The character of early events during B cell–pathogen interaction may determine the type of B cell response regulating the induction of adaptive immunity. We used fluorescence microscopy and flow cytometry to identify the basic requirements for the entry of F. tularensis into B cells within in vivo and in vitro infection models. Here, we present data showing that Francisella tularensis subsp. holarctica strain LVS significantly infects individual subsets of murine peritoneal B cells early after infection. Depending on a given B cell subset, uptake of Francisella into B cells is mediated by B cell receptors (BCRs) with or without complement receptor CR1/2. However, F. tularensis strain FSC200 ΔiglC and ΔftdsbA deletion mutants are defective in the ability to enter B cells. Once internalized into B cells, F. tularensis LVS intracellular trafficking occurs along the endosomal pathway, albeit without significant multiplication. The results strongly suggest that BCRs alone within the B-1a subset can ensure the internalization process while the BCRs on B-1b and B-2 cells need co-signaling from the co receptor containing CR1/2 to initiate F. tularensis engulfment. In this case, fluidity of the surface cell membrane is a prerequisite for the bacteria’s internalization. The results substantially underline the functional heterogeneity of B cell subsets in relation to F. tularensis.
Collapse
Affiliation(s)
- Lenka Plzakova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Zuzana Krocova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
- * E-mail:
| | - Ales Macela
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| |
Collapse
|
31
|
Santiago AE, Mann BJ, Qin A, Cunningham AL, Cole LE, Grassel C, Vogel SN, Levine MM, Barry EM. Characterization of Francisella tularensis Schu S4 defined mutants as live-attenuated vaccine candidates. Pathog Dis 2015; 73:ftv036. [PMID: 25986219 PMCID: PMC4462183 DOI: 10.1093/femspd/ftv036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2015] [Indexed: 01/11/2023] Open
Abstract
Francisella tularensis (Ft), the etiological agent of tularemia and a Tier 1 select agent, has been previously weaponized and remains a high priority for vaccine development. Ft tularensis (type A) and Ft holarctica (type B) cause most human disease. We selected six attenuating genes from the live vaccine strain (LVS; type B), F. novicida and other intracellular bacteria: FTT0507, FTT0584, FTT0742, FTT1019c (guaA), FTT1043 (mip) and FTT1317c (guaB) and created unmarked deletion mutants of each in the highly human virulent Ft strain Schu S4 (Type A) background. FTT0507, FTT0584, FTT0742 and FTT1043 Schu S4 mutants were not attenuated for virulence in vitro or in vivo. In contrast, Schu S4 gua mutants were unable to replicate in murine macrophages and were attenuated in vivo, with an i.n. LD50 > 105 CFU in C57BL/6 mice. However, the gua mutants failed to protect mice against lethal challenge with WT Schu S4, despite demonstrating partial protection in rabbits in a previous study. These results contrast with the highly protective capacity of LVS gua mutants against a lethal LVS challenge in mice, and underscore differences between these strains and the animal models in which they are evaluated, and therefore have important implications for vaccine development. Mutations in guanine biosynthesis genes, but not in four other hypothetical virulence factors in highly virulent Francisella tularensis strain Schu S4 resulted in attenuation in macrophage replication and mouse virulence.
Collapse
Affiliation(s)
- Araceli E Santiago
- Departments of Pediatrics, University of Virginia Children's Hospital, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Barbara J Mann
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Aiping Qin
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Aimee L Cunningham
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Leah E Cole
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Christen Grassel
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Eileen M Barry
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
32
|
De Pascalis R, Mittereder L, Chou AY, Kennett NJ, Elkins KL. Francisella tularensis Vaccines Elicit Concurrent Protective T- and B-Cell Immune Responses in BALB/cByJ Mice. PLoS One 2015; 10:e0126570. [PMID: 25973794 PMCID: PMC4431730 DOI: 10.1371/journal.pone.0126570] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/06/2015] [Indexed: 12/28/2022] Open
Abstract
In the last decade several new vaccines against Francisella tularensis, which causes tularemia, have been characterized in animal models. Whereas many of these vaccine candidates showed promise, it remains critical to bridge the preclinical studies to human subjects, ideally by taking advantage of correlates of protection. By combining in vitro intramacrophage LVS replication with gene expression data through multivariate analysis, we previously identified and quantified correlative T cell immune responses that discriminate vaccines of different efficacy. Further, using C57BL/6J mice, we demonstrated that the relative levels of gene expression vary according to vaccination route and between cell types from different organs. Here, we extended our studies to the analysis of T cell functions of BALB/cByJ mice to evaluate whether our approach to identify correlates of protection also applies to a Th2 dominant mouse strain. BALB/cByJ mice had higher survival rates than C57BL/6J mice when they were immunized with suboptimal vaccines and challenged. However, splenocytes derived from differentially vaccinated BALB/cByJ mice controlled LVS intramacrophage replication in vitro in a pattern that reflected the hierarchy of protection observed in C57BL/6J mice. In addition, gene expression of selected potential correlates revealed similar patterns in splenocytes of BALB/cByJ and C57BL/6J mice. The different survival patterns were related to B cell functions, not necessarily to specific antibody production, which played an important protective role in BALB/cByJ mice when vaccinated with suboptimal vaccines. Our studies therefore demonstrate the range of mechanisms that operate in the most common mouse strains used for characterization of vaccines against F. tularensis, and illustrate the complexity necessary to define a comprehensive set of correlates.
Collapse
Affiliation(s)
- Roberto De Pascalis
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
- * E-mail:
| | - Lara Mittereder
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| | - Alicia Y. Chou
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| | - Nikki J. Kennett
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| | - Karen L. Elkins
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| |
Collapse
|
33
|
Suresh RV, Ma Z, Sunagar R, Bhatty V, Banik S, Catlett SV, Gosselin EJ, Malik M, Bakshi CS. Preclinical testing of a vaccine candidate against tularemia. PLoS One 2015; 10:e0124326. [PMID: 25897786 PMCID: PMC4405390 DOI: 10.1371/journal.pone.0124326] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/02/2015] [Indexed: 11/18/2022] Open
Abstract
Tularemia is caused by a gram-negative, intracellular bacterial pathogen, Francisella tularensis (Ft). The history weaponization of Ft in the past has elevated concerns that it could be used as a bioweapon or an agent of bioterrorism. Since the discovery of Ft, three broad approaches adopted for tularemia vaccine development have included inactivated, live attenuated, or subunit vaccines. Shortcomings in each of these approaches have hampered the development of a suitable vaccine for prevention of tularemia. Recently, we reported an oxidant sensitive mutant of Ft LVS in putative EmrA1 (FTL_0687) secretion protein. The emrA1 mutant is highly sensitive to oxidants, attenuated for intramacrophage growth and virulence in mice. We reported that EmrA1 contributes to oxidant resistance by affecting the secretion of antioxidant enzymes SodB and KatG. This study investigated the vaccine potential of the emrA1 mutant in prevention of respiratory tularemia caused by Ft LVS and the virulent SchuS4 strain in C57BL/6 mice. We report that emrA1 mutant is safe and can be used at an intranasal (i. n.) immunization dose as high as 1x106 CFU without causing any adverse effects in immunized mice. The emrA1 mutant is cleared by vaccinated mice by day 14-21 post-immunization, induces minimal histopathological lesions in lungs, liver and spleen and a strong humoral immune response. The emrA1 mutant vaccinated mice are protected against 1000-10,000LD100 doses of i.n. Ft LVS challenge. Such a high degree of protection has not been reported earlier against respiratory challenge with Ft LVS using a single immunization dose with an attenuated mutant generated on Ft LVS background. The emrA1 mutant also provides partial protection against i.n. challenge with virulent Ft SchuS4 strain in vaccinated C57BL/6 mice. Collectively, our results further support the notion that antioxidants of Ft may serve as potential targets for development of effective vaccines for prevention of tularemia.
Collapse
Affiliation(s)
| | - Zhuo Ma
- Albany College of Pharmacy and Health Sciences, Albany, United States of America
| | - Raju Sunagar
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| | - Vivek Bhatty
- Department of Microbiology and Immunology, New York Medical College, Valhalla, United States of America
| | - Sukalyani Banik
- Department of Microbiology and Immunology, New York Medical College, Valhalla, United States of America
| | - Sally V. Catlett
- Albany College of Pharmacy and Health Sciences, Albany, United States of America
| | - Edmund J. Gosselin
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, United States of America
| | - Meenakshi Malik
- Albany College of Pharmacy and Health Sciences, Albany, United States of America
- * E-mail: (MM); (CSB)
| | - Chandra Shekhar Bakshi
- Department of Microbiology and Immunology, New York Medical College, Valhalla, United States of America
- * E-mail: (MM); (CSB)
| |
Collapse
|
34
|
Senitkova I, Spidlova P, Stulik J. Cooperation of both, the FKBP_N-like and the DSBA-like, domains is necessary for the correct function of FTS_1067 protein involved in Francisella tularensis virulence and pathogenesis. Pathog Dis 2015; 73:ftv030. [PMID: 25896829 DOI: 10.1093/femspd/ftv030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2015] [Indexed: 11/14/2022] Open
Abstract
Francisella tularensis the etiological agent of tularaemia is one of the most infectious human pathogen known. Our knowledge about its key virulence factors has increased recently but it still remains a lot to explore. One of the described essential virulence factors is membrane lipoprotein FTS_1067 (nomenclature of F. tularensis subsp. holarctica strain FSC200) with homology to the protein family of disulphide oxidoreductases DsbA. Lipoprotein consists of two different domains: the C-terminal DsbA_Com1-like domain (DSBA-like) and the N-terminal FKBP-type peptidyl-prolyl cis/trans isomerases (FKBP_N-like). To uncover the biological role of these domains, we created bacterial strain with deletion of the DSBA-like domain. This defect in gene coding for lipoprotein FTS_1067 led to high in vivo attenuation associated with the ability to induce host protective immunity. Analyses performed with the truncated recombinant protein showed that the absence of DSBA-like domain revealed the loss of thiol/disulphide oxidoreductase activity and, additionally, confirmed the role of the FKBP_N-like domain in the FTS_1067 oligomerization and chaperone-like function. Finally, we verified that only full-length form of FTS_1067 recombinant protein possesses the isomerase activity. Based on our results, we proposed that for the correct FTS_1067 protein function both domains are needed.
Collapse
Affiliation(s)
- Iva Senitkova
- Faculty of Military Health Sciences, University of Defence, Department of Molecular Pathology and Biology, Trebesska 1575, Hradec Kralove 50001, Czech Republic
| | - Petra Spidlova
- Faculty of Military Health Sciences, University of Defence, Department of Molecular Pathology and Biology, Trebesska 1575, Hradec Kralove 50001, Czech Republic
| | - Jiri Stulik
- Faculty of Military Health Sciences, University of Defence, Department of Molecular Pathology and Biology, Trebesska 1575, Hradec Kralove 50001, Czech Republic
| |
Collapse
|
35
|
Kubelkova K, Macela A. Putting the Jigsaw Together - A Brief Insight Into the Tularemia. Open Life Sci 2015. [DOI: 10.1515/biol-2015-0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AbstractTularemia is a debilitating febrile and potentially fatal zoonotic disease of humans and other vertebrates caused by the Gram-negative bacterium Francisella tularensis. The natural reservoirs are small rodents, hares, and possibly amoebas in water. The etiological agent, Francisella tularensis, is a non-spore forming, encapsulated, facultative intracellular bacterium, a member of the γ-Proteobacteria class of Gram-negative bacteria. Francisella tularensis is capable of invading and replicating within phagocytic as well as non-phagocytic cells and modulate inflammatory response. Infection by the pulmonary, dermal, or oral routes, respectively, results in pneumonic, ulceroglandular, or oropharyngeal tularemia. The highest mortality rates are associated with the pneumonic form of this disease. All members of Francisella tularensis species cause more or less severe disease Due to their abilities to be transmitted to humans via multiple routes and to be disseminated via biological aerosol that can cause the disease after inhalation of even an extremely low infectious dose, Francisella tularensis has been classified as a Category A bioterrorism agent. The current standard of care for tularemia is treatment with antibiotics, as this therapy is highly effective if used soon after infection, although it is not, however, absolutely effective in all cases.
Collapse
|
36
|
Ren G, Champion MM, Huntley JF. Identification of disulfide bond isomerase substrates reveals bacterial virulence factors. Mol Microbiol 2014; 94:926-44. [PMID: 25257164 DOI: 10.1111/mmi.12808] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2014] [Indexed: 01/22/2023]
Abstract
Bacterial pathogens are exposed to toxic molecules inside the host and require efficient systems to form and maintain correct disulfide bonds for protein stability and function. The intracellular pathogen Francisella tularensis encodes a disulfide bond formation protein ortholog, DsbA, which previously was reported to be required for infection of macrophages and mice. However, the molecular mechanisms by which F. tularensis DsbA contributes to virulence are unknown. Here, we demonstrate that F. tularensis DsbA is a bifunctional protein that oxidizes and, more importantly, isomerizes complex disulfide connectivity in substrates. A single amino acid in the conserved cis-proline loop of the DsbA thioredoxin domain was shown to modulate both isomerase activity and F. tularensis virulence. Trapping experiments in F. tularensis identified over 50 F. tularensis DsbA substrates, including outer membrane proteins, virulence factors, and many hypothetical proteins. Six of these hypothetical proteins were randomly selected and deleted, revealing two novel proteins, FTL_1548 and FTL_1709, which are required for F. tularensis virulence. We propose that the extreme virulence of F. tularensis is partially due to the bifunctional nature of DsbA, that many of the newly identified substrates are required for virulence, and that the development of future DsbA inhibitors could have broad anti-bacterial implications.
Collapse
Affiliation(s)
- Guoping Ren
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | | | | |
Collapse
|
37
|
Characterization of tetratricopeptide repeat-like proteins in Francisella tularensis and identification of a novel locus required for virulence. Infect Immun 2014; 82:5035-48. [PMID: 25245806 DOI: 10.1128/iai.01620-14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Francisella tularensis is a highly infectious bacterium that causes the potentially lethal disease tularemia. This extremely virulent bacterium is able to replicate in the cytosolic compartments of infected macrophages. To invade macrophages and to cope with their intracellular environment, Francisella requires multiple virulence factors, which are still being identified. Proteins containing tetratricopeptide repeat (TPR)-like domains seem to be promising targets to investigate, since these proteins have been reported to be directly involved in virulence-associated functions of bacterial pathogens. Here, we studied the role of the FTS_0201, FTS_0778, and FTS_1680 genes, which encode putative TPR-like proteins in Francisella tularensis subsp. holarctica FSC200. Mutants defective in protein expression were prepared by TargeTron insertion mutagenesis. We found that the locus FTS_1680 and its ortholog FTT_0166c in the highly virulent Francisella tularensis type A strain SchuS4 are required for proper intracellular replication, full virulence in mice, and heat stress tolerance. Additionally, the FTS_1680-encoded protein was identified as a membrane-associated protein required for full cytopathogenicity in macrophages. Our study thus identifies FTS_1680/FTT_0166c as a new virulence factor in Francisella tularensis.
Collapse
|
38
|
FipB, an essential virulence factor of Francisella tularensis subsp. tularensis, has dual roles in disulfide bond formation. J Bacteriol 2014; 196:3571-81. [PMID: 25092026 DOI: 10.1128/jb.01359-13] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
FipB, an essential virulence factor of Francisella tularensis, is a lipoprotein with two conserved domains that have similarity to disulfide bond formation A (DsbA) proteins and the amino-terminal dimerization domain of macrophage infectivity potentiator (Mip) proteins, which are proteins with peptidyl-prolyl cis/trans isomerase activity. This combination of conserved domains is unusual, so we further characterized the enzymatic activity and the importance of the Mip domain and lipid modification in virulence. Unlike typical DsbA proteins, which are oxidases, FipB exhibited both oxidase and isomerase activities. FipA, which also shares similarity with Mip proteins, potentiated the isomerase activity of FipB in an in vitro assay and within the bacteria, as measured by increased copper sensitivity. To determine the importance of the Mip domain and lipid modification of FipB, mutants producing FipB proteins that lacked either the Mip domain or the critical cysteine necessary for lipid modification were constructed. Both strains replicated within host cells and retained virulence in mice, though there was some attenuation. FipB formed surface-exposed dimers that were sensitive to dithiothreitol (DTT), dependent on the Mip domain and on at least one cysteine in the active site of the DsbA-like domain. However, these dimers were not essential for virulence, because the Mip deletion mutant, which failed to form dimers, was still able to replicate intracellularly and retained virulence in mice. Thus, the Mip domains of FipB and FipA impart additional isomerase functionality to FipB, but only the DsbA-like domain and oxidase activity are essential for its critical virulence functions.
Collapse
|
39
|
Live attenuated mutants of Francisella tularensis protect rabbits against aerosol challenge with a virulent type A strain. Infect Immun 2014; 82:2098-105. [PMID: 24614653 DOI: 10.1128/iai.01498-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Francisella tularensis, a Gram-negative bacterium, is the causative agent of tularemia. No licensed vaccine is currently available for protection against tularemia, although an attenuated strain, dubbed the live vaccine strain (LVS), is given to at-risk laboratory personnel as an investigational new drug (IND). In an effort to develop a vaccine that offers better protection, recombinant attenuated derivatives of a virulent type A strain, SCHU S4, were evaluated in New Zealand White (NZW) rabbits. Rabbits vaccinated via scarification with the three attenuated derivatives (SCHU S4 ΔguaBA, ΔaroD, and ΔfipB strains) or with LVS developed a mild fever, but no weight loss was detected. Twenty-one days after vaccination, all vaccinated rabbits were seropositive for IgG to F. tularensis lipopolysaccharide (LPS). Thirty days after vaccination, all rabbits were challenged with aerosolized SCHU S4 at doses ranging from 50 to 500 50% lethal doses (LD50). All rabbits developed fevers and weight loss after challenge, but the severity was greater for mock-vaccinated rabbits. The ΔguaBA and ΔaroD SCHU S4 derivatives provided partial protection against death (27 to 36%) and a prolonged time to death compared to results for the mock-vaccinated group. In contrast, LVS and the ΔfipB strain both prolonged the time to death, but there were no survivors from the challenge. This is the first demonstration of vaccine efficacy against aerosol challenge with virulent type A F. tularensis in a species other than a rodent since the original work with LVS in the 1960s. The ΔguaBA and ΔaroD SCHU S4 derivatives warrant further evaluation and consideration as potential vaccines for tularemia and for identification of immunological correlates of protection.
Collapse
|
40
|
Uda A, Sekizuka T, Tanabayashi K, Fujita O, Kuroda M, Hotta A, Sugiura N, Sharma N, Morikawa S, Yamada A. Role of pathogenicity determinant protein C (PdpC) in determining the virulence of the Francisella tularensis subspecies tularensis SCHU. PLoS One 2014; 9:e89075. [PMID: 24558472 PMCID: PMC3928404 DOI: 10.1371/journal.pone.0089075] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 01/14/2014] [Indexed: 02/02/2023] Open
Abstract
Francisella tularensis subspecies tularensis, the etiological agent of tularemia, is highly pathogenic to humans and animals. However, the SCHU strain of F. tularensis SCHU P0 maintained by passaging in artificial media has been found to be attenuated. To better understand the molecular mechanisms behind the pathogenicity of F. tularensis SCHU, we attempted to isolate virulent bacteria by serial passages in mice. SCHU P5 obtained after 5th passages in mice remained avirulent, while SCHU P9 obtained after 9th passages was completely virulent in mice. Moreover, SCHU P9 grew more efficiently in J774.1 murine macrophages compared with that in the less pathogenic SCHU P0 and P5. Comparison of the nucleotide sequences of the whole genomes of SCHU P0, P5, and P9 revealed only 1 nucleotide difference among P0, P5 and P9 in 1 of the 2 copies of pathogenicity determinant protein C (pdpC) gene. An adenine residue deletion was observed in the pdpC1 gene of SCHU P0, P5, and P9 and in the pdpC2 gene of SCHU P0, and P5, while P9 was characterized by the wild type pdpC2 gene. Thus, SCHU P0 and P5 expressed only truncated forms of PdpC protein, while SCHU P9 expressed both wild type and truncated versions. To validate the pathogenicity of PdpC, both copies of the pdpC gene in SCHU P9 have been inactivated by Targetron mutagenesis. SCHU P9 mutants with inactivated pdpC gene showed low intracellular growth in J774.1 cells and did not induce severe disease in experimentally infected mice, while virulence of the mutants was restored by complementation with expression of the intact PdpC. These results demonstrate that PdpC is crucial in determining the virulence of F. tularensis SCHU.
Collapse
Affiliation(s)
- Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomics Center, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Kiyoshi Tanabayashi
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Osamu Fujita
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Makoto Kuroda
- Pathogen Genomics Center, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Akitoyo Hotta
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Naoko Sugiura
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
- United Graduate School of Veterinary Science, Gifu University, Gifu, Japan
| | - Neekun Sharma
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
- United Graduate School of Veterinary Science, Gifu University, Gifu, Japan
| | - Shigeru Morikawa
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Akio Yamada
- Laboratory of Veterinary Public Health, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| |
Collapse
|
41
|
Golovliov I, Twine SM, Shen H, Sjostedt A, Conlan W. A ΔclpB mutant of Francisella tularensis subspecies holarctica strain, FSC200, is a more effective live vaccine than F. tularensis LVS in a mouse respiratory challenge model of tularemia. PLoS One 2013; 8:e78671. [PMID: 24236032 PMCID: PMC3827231 DOI: 10.1371/journal.pone.0078671] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/13/2013] [Indexed: 12/20/2022] Open
Abstract
Francisella tularensis subsp. tularensis is a highly virulent pathogen for humans especially if inhaled. Consequently, it is considered to be a potential biothreat agent. An experimental vaccine, F. tularensis live vaccine strain, derived from the less virulent subsp. holarctica, was developed more than 50 years ago, but remains unlicensed. Previously, we developed a novel live vaccine strain, by deleting the chaperonin clpB gene from F. tularensis subsp. tularensis strain, SCHU S4. SCHU S4ΔclpB was less virulent for mice than LVS and a more effective vaccine against respiratory challenge with wild type SCHU S4. In the current study, we were interested to determine whether a similar mutant on the less virulent subsp. holarctica background would also outperform LVS in terms of safety and efficacy. To this end, clpB was deleted from clinical holarctica strain, FSC200. FSC200ΔclpB had a significantly higher intranasal LD50 than LVS for BALB/c mice, but replicated to higher numbers at foci of infection after dermal inoculation. Moreover, FSC200ΔclpB killed SCID mice more rapidly than LVS. However, dermal vaccination of BALB/c mice with the former versus the latter induced greater protection against respiratory challenge with SCHU S4. This increased efficacy was associated with enhanced production of pulmonary IL-17 after SCHU S4 challenge.
Collapse
Affiliation(s)
- Igor Golovliov
- Department of Clinical Microbiology, Clinical Bacteriology, Umeå University, Umeå, Sweden
| | - Susan M. Twine
- National Research Council Canada, Human Health and Therapeutics Portfolio, Ottawa, Ontario, Canada
| | - Hua Shen
- National Research Council Canada, Human Health and Therapeutics Portfolio, Ottawa, Ontario, Canada
| | - Anders Sjostedt
- Department of Clinical Microbiology, Clinical Bacteriology, Umeå University, Umeå, Sweden
| | - Wayne Conlan
- National Research Council Canada, Human Health and Therapeutics Portfolio, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
42
|
Schmidt M, Klimentova J, Rehulka P, Straskova A, Spidlova P, Szotakova B, Stulik J, Pavkova I. Francisella tularensis subsp. holarctica DsbA homologue: a thioredoxin-like protein with chaperone function. MICROBIOLOGY-SGM 2013; 159:2364-2374. [PMID: 24014665 DOI: 10.1099/mic.0.070516-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Francisella tularensis is a highly infectious facultative intracellular bacterium and aetiological agent of tularaemia. The conserved hypothetical lipoprotein with homology to thiol/disulphide oxidoreductase proteins (FtDsbA) is an essential virulence factor in F. tularensis. Its protein sequence has two different domains: the DsbA_Com1_like domain (DSBA), with the highly conserved catalytically active site CXXC and cis-proline residue; and the domain amino-terminal to FKBP-type peptidyl-prolyl isomerases (FKBP_N). To establish the role of both domains in tularaemia infection models, site-directed and deletion mutagenesis affecting the active site (AXXA), the cis-proline (P286T) and the FKBP_N domain (ΔFKBP_N) were performed. The generated mutations led to high attenuation with the ability to induce full or partial host protective immunity. Recombinant protein analysis revealed that the active site CXXC as well as the cis-proline residue and the FKBP_N domain are necessary for correct thiol/disulphide oxidoreductase activity. By contrast, only the DSBA domain (and not the FKBP_N domain) seems to be responsible for the in vitro chaperone activity of the FtDsbA protein.
Collapse
Affiliation(s)
- Monika Schmidt
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic.,Department of Biochemical Studies, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, 500 05 Hradec Kralove, Czech Republic
| | - Jana Klimentova
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Pavel Rehulka
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Adela Straskova
- Center of Advanced Studies, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Petra Spidlova
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Barbora Szotakova
- Department of Biochemical Studies, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, 500 05 Hradec Kralove, Czech Republic
| | - Jiri Stulik
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Ivona Pavkova
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| |
Collapse
|
43
|
Abstract
Francisella tularensis is a highly virulent bacterial pathogen that is easily aerosolized and has a low infectious dose. As an intracellular pathogen, entry of Francisella into host cells is critical for its survival and virulence. However, the initial steps of attachment and internalization of Francisella into host cells are not well characterized, and little is known about bacterial factors that promote these processes. This review highlights our current understanding of Francisella attachment and internalization into host cells. In particular, we emphasize the host cell types Francisella has been shown to interact with, as well as specific receptors and signaling processes involved in the internalization process. This review will shed light on gaps in our current understanding and future areas of investigation.
Collapse
Affiliation(s)
- G Brett Moreau
- Department of Microbiology, Immunology, and Cancer Biology; University of Virginia; Charlottesville, VA USA
| | - Barbara J Mann
- Department of Microbiology, Immunology, and Cancer Biology; University of Virginia; Charlottesville, VA USA; Department of Medicine; Division of Infectious Diseases and International Health; University of Virginia; Charlottesville, VA USA
| |
Collapse
|
44
|
Chong A, Child R, Wehrly TD, Rockx-Brouwer D, Qin A, Mann BJ, Celli J. Structure-Function Analysis of DipA, a Francisella tularensis Virulence Factor Required for Intracellular Replication. PLoS One 2013; 8:e67965. [PMID: 23840797 PMCID: PMC3694160 DOI: 10.1371/journal.pone.0067965] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/23/2013] [Indexed: 12/26/2022] Open
Abstract
Francisella tularensis is a highly infectious bacterium whose virulence relies on its ability to rapidly reach the macrophage cytosol and extensively replicate in this compartment. We previously identified a novel Francisella virulence factor, DipA (FTT0369c), which is required for intramacrophage proliferation and survival, and virulence in mice. DipA is a 353 amino acid protein with a Sec-dependent signal peptide, four Sel1-like repeats (SLR), and a C-terminal coiled-coil (CC) domain. Here, we determined through biochemical and localization studies that DipA is a membrane-associated protein exposed on the surface of the prototypical F. tularensis subsp. tularensis strain SchuS4 during macrophage infection. Deletion and substitution mutagenesis showed that the CC domain, but not the SLR motifs, of DipA is required for surface exposure on SchuS4. Complementation of the dipA mutant with either DipA CC or SLR domain mutants did not restore intracellular growth of Francisella, indicating that proper localization and the SLR domains are required for DipA function. Co-immunoprecipitation studies revealed interactions with the Francisella outer membrane protein FopA, suggesting that DipA is part of a membrane-associated complex. Altogether, our findings indicate that DipA is positioned at the host–pathogen interface to influence the intracellular fate of this pathogen.
Collapse
Affiliation(s)
- Audrey Chong
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| | - Robert Child
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Tara D. Wehrly
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Dedeke Rockx-Brouwer
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Aiping Qin
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Barbara J. Mann
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jean Celli
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| |
Collapse
|
45
|
Live attenuated tularemia vaccines: recent developments and future goals. Vaccine 2013; 31:3485-91. [PMID: 23764535 DOI: 10.1016/j.vaccine.2013.05.096] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/20/2013] [Accepted: 05/24/2013] [Indexed: 12/24/2022]
Abstract
In the aftermath of the 2001 anthrax attacks in the U.S., numerous efforts were made to increase the level of preparedness against a biological attack both in the US and worldwide. As a result, there has been an increase in research interest in the development of vaccines and other countermeasures against a number of agents with the potential to be used as biological weapons. One such agent, Francisella tularensis, has been the subject of a surge in the level of research being performed, leading to a substantial increase in knowledge of the pathogenic mechanisms of the organism and the induced immune responses. This information has facilitated the development of multiple new Francisella vaccine candidates. Herein we review the latest live attenuated F. tularensis vaccine efforts. Historically, live attenuated vaccines have demonstrated the greatest degree of success in protection against tularemia and the greatest promise in recent efforts to develop of a fully protective vaccine. This review summarizes recent live attenuated Francisella vaccine candidates and the lessons learned from those studies, with the goal of collating known characteristics associated with successful attenuation, immunogenicity, and protection.
Collapse
|
46
|
Walters KA, Olsufka R, Kuestner RE, Cho JH, Li H, Zornetzer GA, Wang K, Skerrett SJ, Ozinsky A. Francisella tularensis subsp. tularensis induces a unique pulmonary inflammatory response: role of bacterial gene expression in temporal regulation of host defense responses. PLoS One 2013; 8:e62412. [PMID: 23690939 PMCID: PMC3653966 DOI: 10.1371/journal.pone.0062412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/20/2013] [Indexed: 12/26/2022] Open
Abstract
Pulmonary exposure to Francisella tularensis is associated with severe lung pathology and a high mortality rate. The lack of induction of classical inflammatory mediators, including IL1-β and TNF-α, during early infection has led to the suggestion that F. tularensis evades detection by host innate immune surveillance and/or actively suppresses inflammation. To gain more insight into the host response to Francisella infection during the acute stage, transcriptomic analysis was performed on lung tissue from mice exposed to virulent (Francisella tularensis ssp tularensis SchuS4). Despite an extensive transcriptional response in the lungs of animals as early as 4 hrs post-exposure, Francisella tularensis was associated with an almost complete lack of induction of immune-related genes during the initial 24 hrs post-exposure. This broad subversion of innate immune responses was particularly evident when compared to the pulmonary inflammatory response induced by other lethal (Yersinia pestis) and non-lethal (Legionella pneumophila, Pseudomonas aeruginosa) pulmonary infections. However, the unique induction of a subset of inflammation-related genes suggests a role for dysregulation of lymphocyte function and anti-inflammatory pathways in the extreme virulence of Francisella. Subsequent activation of a classical inflammatory response 48 hrs post-exposure was associated with altered abundance of Francisella-specific transcripts, including those associated with bacterial surface components. In summary, virulent Francisella induces a unique pulmonary inflammatory response characterized by temporal regulation of innate immune pathways correlating with altered bacterial gene expression patterns. This study represents the first simultaneous measurement of both host and Francisella transcriptome changes that occur during in vivo infection and identifies potential bacterial virulence factors responsible for regulation of host inflammatory pathways.
Collapse
|
47
|
Identification of a live attenuated vaccine candidate for tularemia prophylaxis. PLoS One 2013; 8:e61539. [PMID: 23613871 PMCID: PMC3629233 DOI: 10.1371/journal.pone.0061539] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 03/11/2013] [Indexed: 12/25/2022] Open
Abstract
Francisella tularensis is the causative agent of a fatal human disease, tularemia. F. tularensis was used in bioweapon programs in the past and is now classified as a category A select agent owing to its possible use in bioterror attacks. Despite over a century since its discovery, an effective vaccine is yet to be developed. In this study four transposon insertion mutants of F. tularensis live vaccine strain (LVS) in Na/H antiporter (FTL_0304), aromatic amino acid transporter (FTL_0291), outer membrane protein A (OmpA)-like family protein (FTL_0325) and a conserved hypothetical membrane protein gene (FTL_0057) were evaluated for their attenuation and protective efficacy against F. tularensis SchuS4 strain. All four mutants were 100–1000 fold attenuated for virulence in mice than parental F. tularensis. Except for the FTL_0304, single intranasal immunization with the other three mutants provided 100% protection in BALB/c mice against intranasal challenge with virulent F. tularensis SchuS4. Differences in the protective ability of the FTL_0325 and FTL_0304 mutant which failed to provide protection against SchuS4 were investigated further. The results indicated that an early pro-inflammatory response and persistence in host tissues established a protective immunity against F. tularensis SchuS4 in the FTL_0325 immunized mice. No differences were observed in the levels of serum IgG antibodies amongst the two vaccinated groups. Recall response studies demonstrated that splenocytes from the FTL_0325 mutant immunized mice induced significantly higher levels of IFN-γ and IL-17 cytokines than the FTL_0304 immunized counterparts indicating development of an effective memory response. Collectively, this study demonstrates that persistence of the vaccine strain together with its ability to induce an early pro-inflammatory innate immune response and strong memory responses can discriminate between successful and failed vaccinations against tularemia. This study describes a live attenuated vaccine which may prove to be an ideal vaccine candidate for prevention of respiratory tularemia.
Collapse
|
48
|
Celli J, Zahrt TC. Mechanisms of Francisella tularensis intracellular pathogenesis. Cold Spring Harb Perspect Med 2013; 3:a010314. [PMID: 23545572 DOI: 10.1101/cshperspect.a010314] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Francisella tularensis is a zoonotic intracellular pathogen and the causative agent of the debilitating febrile illness tularemia. Although natural infections by F. tularensis are sporadic and generally localized, the low infectious dose, with the ability to be transmitted to humans via multiple routes and the potential to cause life-threatening infections, has led to concerns that this bacterium could be used as an agent of bioterror and released intentionally into the environment. Recent studies of F. tularensis and other closely related Francisella species have greatly increased our understanding of mechanisms used by this organism to infect and cause disease within the host. Here, we review the intracellular life cycle of Francisella and highlight key genetic determinants and/or pathways that contribute to the survival and proliferation of this bacterium within host cells.
Collapse
Affiliation(s)
- Jean Celli
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MO 59840, USA
| | | |
Collapse
|
49
|
Kdo hydrolase is required for Francisella tularensis virulence and evasion of TLR2-mediated innate immunity. mBio 2013; 4:e00638-12. [PMID: 23404403 PMCID: PMC3573668 DOI: 10.1128/mbio.00638-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED The highly virulent Francisella tularensis subsp. tularensis has been classified as a category A bioterrorism agent. A live vaccine strain (LVS) has been developed but remains unlicensed in the United States because of an incomplete understanding of its attenuation. Lipopolysaccharide (LPS) modification is a common strategy employed by bacterial pathogens to avoid innate immunity. A novel modification enzyme has recently been identified in F. tularensis and Helicobacter pylori. This enzyme, a two-component Kdo (3-deoxy-d-manno-octulosonic acid) hydrolase, catalyzes the removal of a side chain Kdo sugar from LPS precursors. The biological significance of this modification has not yet been studied. To address the role of the two-component Kdo hydrolase KdhAB in F. tularensis pathogenesis, a ΔkdhAB deletion mutant was constructed from the LVS strain. In intranasal infection of mice, the ΔkdhAB mutant strain had a 50% lethal dose (LD(50)) 2 log(10) units higher than that of the parental LVS strain. The levels of the proinflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β) in bronchoalveolar lavage fluid were significantly higher (2-fold) in mice infected with the ΔkdhAB mutant than in mice infected with LVS. In vitro stimulation of bone marrow-derived macrophages with the ΔkdhAB mutant induced higher levels of TNF-α and IL-1β in a TLR2-dependent manner. In addition, TLR2(-/-) mice were more susceptible than wild-type mice to ΔkdhAB bacterial infection. Finally, immunization of mice with ΔkdhAB bacteria elicited a high level of protection against the highly virulent F. tularensis subsp. tularensis strain Schu S4. These findings suggest an important role for the Francisella Kdo hydrolase system in virulence and offer a novel mutant as a candidate vaccine. IMPORTANCE The first line of defense against a bacterial pathogen is innate immunity, which slows the progress of infection and allows time for adaptive immunity to develop. Some bacterial pathogens, such as Francisella tularensis, suppress the early innate immune response, killing the host before adaptive immunity can mature. To avoid an innate immune response, F. tularensis enzymatically modifies its lipopolysaccharide (LPS). A novel LPS modification-Kdo (3-deoxy-d-manno-octulosonic acid) saccharide removal--has recently been reported in F. tularensis. We found that the kdhAB mutant was significantly attenuated in mice. Additionally, the mutant strain induced an early innate immune response in mice both in vitro and in vivo. Immunization of mice with this mutant provided protection against the highly virulent F. tularensis strain Schu S4. Thus, our study has identified a novel LPS modification important for microbial virulence. A mutant lacking this modification may be used as a live attenuated vaccine against tularemia.
Collapse
|
50
|
Signarovitz AL, Ray HJ, Yu JJ, Guentzel MN, Chambers JP, Klose KE, Arulanandam BP. Mucosal immunization with live attenuated Francisella novicida U112ΔiglB protects against pulmonary F. tularensis SCHU S4 in the Fischer 344 rat model. PLoS One 2012; 7:e47639. [PMID: 23118885 PMCID: PMC3484155 DOI: 10.1371/journal.pone.0047639] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 09/18/2012] [Indexed: 12/17/2022] Open
Abstract
The need for an efficacious vaccine against Francisella tularensis is a consequence of its low infectious dose and high mortality rate if left untreated. This study sought to characterize a live attenuated subspecies novicida-based vaccine strain (U112ΔiglB) in an established second rodent model of pulmonary tularemia, namely the Fischer 344 rat using two distinct routes of vaccination (intratracheal [i.t.] and oral). Attenuation was verified by comparing replication of U112ΔiglB with wild type parental strain U112 in F344 primary alveolar macrophages. U112ΔiglB exhibited an LD50>107 CFU compared to the wild type (LD50 = 5×106 CFU i.t.). Immunization with 107 CFU U112ΔiglB by i.t. and oral routes induced antigen-specific IFN-γ and potent humoral responses both systemically (IgG2a>IgG1 in serum) and at the site of mucosal vaccination (respiratory/intestinal compartment). Importantly, vaccination with U112ΔiglB by either i.t. or oral routes provided equivalent levels of protection (50% survival) in F344 rats against a subsequent pulmonary challenge with ∼25 LD50 (1.25×104 CFU) of the highly human virulent strain SCHU S4. Collectively, these results provide further evidence on the utility of a mucosal vaccination platform with a defined subsp. novicida U112ΔiglB vaccine strain in conferring protective immunity against pulmonary tularemia.
Collapse
Affiliation(s)
- Aimee L. Signarovitz
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Heather J. Ray
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - M. N. Guentzel
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - James P. Chambers
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Karl E. Klose
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|