1
|
Jang J, Kwon DH, Jang JH, Lee DG, Chang SH, Jeon MY, Jeong YS, Song DH, Min JK, Park JG, Lee MS, Han BS, Yang W, Lee NK, Lee J. Development of a novel sandwich immunoassay based on targeting recombinant Francisella outer membrane protein A for the diagnosis of tularemia. Front Cell Infect Microbiol 2024; 14:1455259. [PMID: 39228894 PMCID: PMC11368854 DOI: 10.3389/fcimb.2024.1455259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Introduction Tularemia, caused by the bacterium Francisella tularensis, poses health risks to humans and can spread through a variety of routes. It has also been classified as a Tier 1 Select agent by the CDC, highlighting its potential as a bioterrorism agent. Moreover, it is difficult to diagnose in a timely fashion, owing to the non-specific nature of tularemia infections. Rapid, sensitive, and accurate detection methods are required to reduce mortality rates. We aimed to develop antibodies directed against the outer membrane protein A of F. tularensis (FopA) for rapid and accurate diagnosis of tularemia. Methods We used a baculovirus insect cell expression vector system to produce the FopA antigen and generate anti-FopA antibodies through immunization of BALB/c mice. We then employed hybridoma and phage display technologies to screen for antibodies that could recognize unique epitopes on FopA. Result Two monoclonal antibodies, 6B12 and 3C1, identified through phage display screening specifically bound to recombinant FopA in a dose-dependent manner. The binding affinity of the anti-FopA 6B12 and 3C1 antibodies was observed to have an equilibrium dissociation constant of 1.76 × 10-10 M and 1.32 × 10-9 M, respectively. These antibodies were used to develop a sandwich ELISA system for the diagnosis of tularemia. This assay was found to be highly specific and sensitive, with detection limits ranging from 0.062 ng/mL in PBS to 0.064 ng/mL in skim milk matrices. Discussion Our findings demonstrate the feasibility of a novel diagnostic approach for detecting F. tularensis based on targeting FopA, as opposed to existing tests that target the bacterial lipopolysaccharide.
Collapse
Affiliation(s)
- Jieun Jang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Do Hyung Kwon
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Ju-Hong Jang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Dong-Gwang Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seo-Hyuk Chang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Min-Young Jeon
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Young-Su Jeong
- Chem-Bio Technology Center, Agency for Defense Development, Daejeon, Republic of Korea
| | - Dong-Hyun Song
- Chem-Bio Technology Center, Agency for Defense Development, Daejeon, Republic of Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Baek-Soo Han
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Wonjun Yang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Nam-Kyung Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
2
|
Biselli R, Nisini R, Lista F, Autore A, Lastilla M, De Lorenzo G, Peragallo MS, Stroffolini T, D’Amelio R. A Historical Review of Military Medical Strategies for Fighting Infectious Diseases: From Battlefields to Global Health. Biomedicines 2022; 10:2050. [PMID: 36009598 PMCID: PMC9405556 DOI: 10.3390/biomedicines10082050] [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: 07/27/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022] Open
Abstract
The environmental conditions generated by war and characterized by poverty, undernutrition, stress, difficult access to safe water and food as well as lack of environmental and personal hygiene favor the spread of many infectious diseases. Epidemic typhus, plague, malaria, cholera, typhoid fever, hepatitis, tetanus, and smallpox have nearly constantly accompanied wars, frequently deeply conditioning the outcome of battles/wars more than weapons and military strategy. At the end of the nineteenth century, with the birth of bacteriology, military medical researchers in Germany, the United Kingdom, and France were active in discovering the etiological agents of some diseases and in developing preventive vaccines. Emil von Behring, Ronald Ross and Charles Laveran, who were or served as military physicians, won the first, the second, and the seventh Nobel Prize for Physiology or Medicine for discovering passive anti-diphtheria/tetanus immunotherapy and for identifying mosquito Anopheline as a malaria vector and plasmodium as its etiological agent, respectively. Meanwhile, Major Walter Reed in the United States of America discovered the mosquito vector of yellow fever, thus paving the way for its prevention by vector control. In this work, the military relevance of some vaccine-preventable and non-vaccine-preventable infectious diseases, as well as of biological weapons, and the military contributions to their control will be described. Currently, the civil-military medical collaboration is getting closer and becoming interdependent, from research and development for the prevention of infectious diseases to disasters and emergencies management, as recently demonstrated in Ebola and Zika outbreaks and the COVID-19 pandemic, even with the high biocontainment aeromedical evacuation, in a sort of global health diplomacy.
Collapse
Affiliation(s)
- Roberto Biselli
- Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - Florigio Lista
- Dipartimento Scientifico, Policlinico Militare, Comando Logistico dell’Esercito, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Alberto Autore
- Osservatorio Epidemiologico della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Marco Lastilla
- Istituto di Medicina Aerospaziale, Comando Logistico dell’Aeronautica Militare, Viale Piero Gobetti 2, 00185 Roma, Italy
| | - Giuseppe De Lorenzo
- Comando Generale dell’Arma dei Carabinieri, Dipartimento per l’Organizzazione Sanitaria e Veterinaria, Viale Romania 45, 00197 Roma, Italy
| | - Mario Stefano Peragallo
- Centro Studi e Ricerche di Sanità e Veterinaria, Comando Logistico dell’Esercito, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Tommaso Stroffolini
- Dipartimento di Malattie Infettive e Tropicali, Policlinico Umberto I, 00161 Roma, Italy
| | - Raffaele D’Amelio
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Via di Grottarossa 1035-1039, 00189 Roma, Italy
| |
Collapse
|
3
|
Nagaratnam N, Martin-Garcia JM, Yang JH, Goode MR, Ketawala G, Craciunescu FM, Zook JD, Sonowal M, Williams D, Grant TD, Fromme R, Hansen DT, Fromme P. Structural and biophysical properties of FopA, a major outer membrane protein of Francisella tularensis. PLoS One 2022; 17:e0267370. [PMID: 35913965 PMCID: PMC9342783 DOI: 10.1371/journal.pone.0267370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis is an extremely infectious pathogen and a category A bioterrorism agent. It causes the highly contagious zoonosis, Tularemia. Currently, FDA approved vaccines against tularemia are unavailable. F. tularensis outer membrane protein A (FopA) is a well-studied virulence determinant and protective antigen against tularemia. It is a major outer membrane protein (Omp) of F. tularensis. However, FopA-based therapeutic intervention is hindered due to lack of complete structural information for membrane localized mature FopA. In our study, we established recombinant expression, monodisperse purification, crystallization and X-ray diffraction (~6.5 Å) of membrane localized mature FopA. Further, we performed bioinformatics and biophysical experiments to unveil its structural organization in the outer membrane. FopA consists of 393 amino acids and has less than 40% sequence identity to known bacterial Omps. Using comprehensive sequence alignments and structure predictions together with existing partial structural information, we propose a two-domain organization for FopA. Circular dichroism spectroscopy and heat modifiability assay confirmed FopA has a β-barrel domain consistent with alphafold2’s prediction of an eight stranded β-barrel at the N-terminus. Small angle X-ray scattering (SAXS) and native-polyacrylamide gel electrophoresis revealed FopA purified in detergent micelles is predominantly dimeric. Molecular density derived from SAXS at 31 Å shows putative dimeric N-terminal β-barrels surrounded by detergent corona and connected to C-terminal domains via flexible linker. Disorder analysis predicts N- and C-terminal domains are interspersed by a long intrinsically disordered region and alphafold2 predicts this region to be largely unstructured. Taken together, we propose a dimeric, two-domain organization of FopA in the outer membrane: the N-terminal β-barrel is membrane embedded, provides dimerization interface and tethers to membrane extrinsic C-terminal domain via long flexible linker. Structure determination of membrane localized mature FopA is essential to understand its role in pathogenesis and develop anti-tularemia therapeutics. Our results pave the way towards it.
Collapse
Affiliation(s)
- Nirupa Nagaratnam
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jose M. Martin-Garcia
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jay-How Yang
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Matthew R. Goode
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Gihan Ketawala
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Felicia M. Craciunescu
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - James D. Zook
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Manashi Sonowal
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Dewight Williams
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- Eyring Materials Center, Arizona State University, Tempe, Arizona, United States of America
| | - Thomas D. Grant
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, New York, New York, United States of America
| | - Raimund Fromme
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Debra T. Hansen
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- Center for Innovations in Medicine, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Petra Fromme
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
- * E-mail:
| |
Collapse
|
4
|
Roberts LM, Wehrly TD, Leighton I, Hanley P, Lovaglio J, Smith BJ, Bosio CM. Circulating T Cells Are Not Sufficient for Protective Immunity against Virulent Francisella tularensis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1180-1188. [PMID: 35149529 PMCID: PMC8881340 DOI: 10.4049/jimmunol.2100915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/23/2021] [Indexed: 11/19/2022]
Abstract
Pulmonary infections elicit a combination of tissue-resident and circulating T cell responses. Understanding the contribution of these anatomically distinct cellular pools in protective immune responses is critical for vaccine development. Francisella tularensis is a highly virulent bacterium capable of causing lethal systemic disease following pulmonary infection for which there is no currently licensed vaccine. Although T cells are required for survival of F. tularensis infection, the relative contribution of tissue-resident and circulating T cells is not completely understood, hampering design of effective, long-lasting vaccines directed against this bacterium. We have previously shown that resident T cells were not sufficient to protect against F. tularensis, suggesting circulating cells may serve a critical role in host defense. To elucidate the role of circulating T cells, we used a model of vaccination and challenge of parabiotic mice. Intranasally infected naive mice conjoined to immune animals had increased numbers of circulating memory T cells and similar splenic bacterial burdens as vaccinated-vaccinated pairs. However, bacterial loads in the lungs of naive parabionts were significantly greater than those observed in vaccinated-vaccinated pairs, but despite early control of F. tularensis replication, all naive-vaccinated pairs succumbed to infection. Together, these data define the specific roles of circulating and resident T cells in defense against infection that is initiated in the pulmonary compartment but ultimately causes disseminated disease. These data also provide evidence for employing vaccination strategies that elicit both pools of T cells for immunity against F. tularensis and may be a common theme for other disseminating bacterial infections.
Collapse
Affiliation(s)
- Lydia M Roberts
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Tara D Wehrly
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Ian Leighton
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Patrick Hanley
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Brian J Smith
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Catharine M Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT; and
| |
Collapse
|
5
|
Anti gC1qR/p32/HABP1 Antibody Therapy Decreases Tumor Growth in an Orthotopic Murine Xenotransplant Model of Triple Negative Breast Cancer. Antibodies (Basel) 2020; 9:antib9040051. [PMID: 33036212 PMCID: PMC7709104 DOI: 10.3390/antib9040051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/18/2020] [Accepted: 09/08/2020] [Indexed: 01/09/2023] Open
Abstract
gC1qR is highly expressed in breast cancer and plays a role in cancer cell proliferation. This study explored therapy with gC1qR monoclonal antibody 60.11, directed against the C1q binding domain of gC1qR, in a murine orthotopic xenotransplant model of triple negative breast cancer. MDA231 breast cancer cells were injected into the mammary fat pad of athymic nu/nu female mice. Mice were segregated into three groups (n = 5, each) and treated with the vehicle (group 1) or gC1qR antibody 60.11 (100 mg/kg) twice weekly, starting at day 3 post-implantation (group 2) or when the tumor volume reached 100 mm3 (group 3). At study termination (d = 35), the average tumor volume in the control group measured 895 ± 143 mm3, compared to 401 ± 48 mm3 and 701 ± 100 mm3 in groups 2 and 3, respectively (p < 0.05). Immunohistochemical staining of excised tumors revealed increased apoptosis (caspase 3 and TUNEL staining) in 60.11-treated mice compared to controls, and decreased angiogenesis (CD31 staining). Slightly decreased white blood cell counts were noted in 60.11-treated mice. Otherwise, no overt toxicities were observed. These data are the first to demonstrate an in vivo anti-tumor effect of 60.11 therapy in a mouse model of triple negative breast cancer.
Collapse
|
6
|
Inhibition of Francisella tularensis phagocytosis using a novel anti-LPS scFv antibody fragment. Sci Rep 2019; 9:11418. [PMID: 31388083 PMCID: PMC6684794 DOI: 10.1038/s41598-019-47931-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 07/25/2019] [Indexed: 12/23/2022] Open
Abstract
Francisella tularensis (Ft), the causative agent of lethal tularemia, is classified as a category A biological warfare threat agent. While Ft infection is treatable by antibiotics, many failed antibiotic treatments were reported, highlighting the need for effective new treatments. It has been demonstrated that binding of antibody-coated bacteria to the Fc receptor located on phagocytic cells is a key process needed for efficient protection against Ft. Yet, Ft utilizes the same receptor to enter the phagocytic cells in order to escape the immune system. To address the question whether an anti-Ft LPS antibody lacking the ability to bind the Fc receptor may inhibit the entry of Ft into host cells, a soluble scFv (TL1-scFv) was constructed from an anti Ft-LPS antibody (TL1) that was isolated from an immune single-chain (scFv) phage-display library. Bacterial uptake was assessed upon infection of macrophages with Ft live attenuated strain (LVS) in the presence of either TL1 or TL1-scFv. While incubation of LVS in the presence of TL1 greatly enhanced bacterial uptake, LVS uptake was significantly inhibited in the presence of TL1-scFv. These results prompt further experiments probing the therapeutic efficacy of TL1-scFv, alone or in combination with antibiotic treatment.
Collapse
|
7
|
Fletcher JR, Crane DD, Wehrly TD, Martens CA, Bosio CM, Jones BD. The Ability to Acquire Iron Is Inversely Related to Virulence and the Protective Efficacy of Francisella tularensis Live Vaccine Strain. Front Microbiol 2018; 9:607. [PMID: 29670588 PMCID: PMC5893802 DOI: 10.3389/fmicb.2018.00607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/15/2018] [Indexed: 02/02/2023] Open
Abstract
Francisella tularensis is a highly infectious bacterial pathogen that causes the potentially fatal disease tularemia. The Live Vaccine Strain (LVS) of F. tularensis subsp. holarctica, while no longer licensed as a vaccine, is used as a model organism for identifying correlates of immunity and bacterial factors that mediate a productive immune response against F. tularensis. Recently, it was reported that two biovars of LVS differed in their virulence and vaccine efficacy. Genetic analysis showed that they differ in ferrous iron homeostasis; lower Fe2+ levels contributed to increased resistance to hydrogen peroxide in the vaccine efficacious LVS biovar. This also correlated with resistance to the bactericidal activity of interferon γ-stimulated murine bone marrow-derived macrophages. We have extended these findings further by showing that a mutant lacking bacterioferritin stimulates poor protection against Schu S4 challenge in a mouse model of tularemia. Together these results suggest that the efficacious biovar of LVS stimulates productive immunity by a mechanism that is dependent on its ability to limit the toxic effects of oxidative stress by maintaining optimally low levels of intracellular Fe2+.
Collapse
Affiliation(s)
- Joshua R. Fletcher
- Graduate Program in Genetics, University of Iowa, Iowa City, IA, United States
| | - Deborah D. Crane
- Immunity to Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Hamilton, MT, United States
| | - Tara D. Wehrly
- Immunity to Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Hamilton, MT, United States
| | - Craig A. Martens
- Genomics Core, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Hamilton, MT, United States
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Hamilton, MT, United States
| | - Bradley D. Jones
- Graduate Program in Genetics, University of Iowa, Iowa City, IA, United States
- Department of Microbiology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
8
|
Sampath V, McCaig WD, Thanassi DG. Amino acid deprivation and central carbon metabolism regulate the production of outer membrane vesicles and tubes by Francisella. Mol Microbiol 2018; 107:523-541. [PMID: 29240272 DOI: 10.1111/mmi.13897] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 12/23/2022]
Abstract
Francisella tularensis is a highly virulent Gram-negative bacterial pathogen that causes the zoonotic disease tularemia. F. novicida, a model tularemia strain, produces spherical outer membrane vesicles (OMV), as well as novel tubular vesicles and extensions of the cell surface. These OMV and tubes (OMV/T) are produced in a regulated manner and contain known virulence factors. Mechanisms by which bacterial vesicles are produced and regulated are not well understood. We performed a genetic screen in F. novicida to decipher the molecular basis for regulated OMV/T formation, and identified both hypo- and hyper-vesiculating mutants. Mutations in fumA and tktA, involved in central carbon metabolism, and in FTN_0908 and FTN_1037, of unknown function, resulted in severe defects in OMV/T production. Cysteine deprivation was identified as the signal that triggers OMV/T formation in F. novicida during growth in rich medium. We also found that fully virulent F. tularensis produces OMV/T in a similarly regulated manner. Further analysis revealed that OMV/T production is responsive to deprivation of essential amino acids in addition to cysteine, and that the hypo-vesiculating mutants are defective in responding to this signal. Thus, amino acid starvation, such as encountered by Francisella during host cell invasion, regulates the production of membrane-derived structures.
Collapse
Affiliation(s)
- Vinaya Sampath
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, NY 11794, USA
| | - William D McCaig
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, NY 11794, USA
| | - David G Thanassi
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
9
|
Post DMB, Slütter B, Schilling B, Chande AT, Rasmussen JA, Jones BD, D'Souza AK, Reinders LM, Harty JT, Gibson BW, Apicella MA. Characterization of Inner and Outer Membrane Proteins from Francisella tularensis Strains LVS and Schu S4 and Identification of Potential Subunit Vaccine Candidates. mBio 2017; 8:e01592-17. [PMID: 29018123 PMCID: PMC5635693 DOI: 10.1128/mbio.01592-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 01/02/2023] Open
Abstract
Francisella tularensis is the causative agent of tularemia and a potential bioterrorism agent. In the present study, we isolated, identified, and quantified the proteins present in the membranes of the virulent type A strain, Schu S4, and the attenuated type B strain, LVS (live vaccine strain). Spectral counting of mass spectrometric data showed enrichment for membrane proteins in both strains. Mice vaccinated with whole LVS membranes encapsulated in poly (lactic-co-glycolic acid) (PLGA) nanoparticles containing the adjuvant polyinosinic-polycytidylic acid [poly(I·C)] showed significant protection against a challenge with LVS compared to the results seen with naive mice or mice vaccinated with either membranes or poly(I·C) alone. The PLGA-encapsulated Schu S4 membranes with poly(I·C) alone did not significantly protect mice from a lethal intraperitoneal challenge with Schu S4; however, this vaccination strategy provided protection from LVS challenge. Mice that received the encapsulated Schu S4 membranes followed by a booster of LVS bacteria showed significant protection with respect to a lethal Schu S4 challenge compared to control mice. Western blot analyses of the sera from the Schu S4-vaccinated mice that received an LVS booster showed four immunoreactive bands. One of these bands from the corresponding one-dimensional (1D) SDS-PAGE experiment represented capsule. The remaining bands were excised, digested with trypsin, and analyzed using mass spectrometry. The most abundant proteins present in these immunoreactive samples were an outer membrane OmpA-like protein, FopA; the type IV pilus fiber building block protein; a hypothetical membrane protein; and lipoproteins LpnA and Lpp3. These proteins should serve as potential targets for future recombinant protein vaccination studies.IMPORTANCE The low infectious dose, the high potential mortality/morbidity rates, and the ability to be disseminated as an aerosol make Francisella tularensis a potential agent for bioterrorism. These characteristics led the Centers for Disease Control (CDC) to classify F. tularensis as a Tier 1 pathogen. Currently, there is no vaccine approved for general use in the United States.
Collapse
Affiliation(s)
| | - Bram Slütter
- Department of Microbiology, the University of Iowa, Iowa City, Iowa, USA
| | | | - Aroon T Chande
- Department of Microbiology, the University of Iowa, Iowa City, Iowa, USA
| | - Jed A Rasmussen
- Department of Microbiology, the University of Iowa, Iowa City, Iowa, USA
| | - Bradley D Jones
- Department of Microbiology, the University of Iowa, Iowa City, Iowa, USA
| | | | | | - John T Harty
- Department of Microbiology, the University of Iowa, Iowa City, Iowa, USA
| | - Bradford W Gibson
- Buck Institute for Research on Aging, Novato, California, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, USA
| | - Michael A Apicella
- Department of Microbiology, the University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
10
|
Holland KM, Rosa SJ, Kristjansdottir K, Wolfgeher D, Franz BJ, Zarrella TM, Kumar S, Sunagar R, Singh A, Bakshi CS, Namjoshi P, Barry EM, Sellati TJ, Kron SJ, Gosselin EJ, Reed DS, Hazlett KRO. Differential Growth of Francisella tularensis, Which Alters Expression of Virulence Factors, Dominant Antigens, and Surface-Carbohydrate Synthases, Governs the Apparent Virulence of Ft SchuS4 to Immunized Animals. Front Microbiol 2017; 8:1158. [PMID: 28690600 PMCID: PMC5479911 DOI: 10.3389/fmicb.2017.01158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 06/07/2017] [Indexed: 12/29/2022] Open
Abstract
The gram-negative bacterium Francisella tularensis (Ft) is both a potential biological weapon and a naturally occurring microbe that survives in arthropods, fresh water amoeba, and mammals with distinct phenotypes in various environments. Previously, we used a number of measurements to characterize Ft grown in Brain-Heart Infusion (BHI) broth as (1) more similar to infection-derived bacteria, and (2) slightly more virulent in naïve animals, compared to Ft grown in Mueller Hinton Broth (MHB). In these studies we observed that the free amino acids in MHB repress expression of select Ft virulence factors by an unknown mechanism. Here, we tested the hypotheses that Ft grown in BHI (BHI-Ft) accurately displays a full protein composition more similar to that reported for infection-derived Ft and that this similarity would make BHI-Ft more susceptible to pre-existing, vaccine-induced immunity than MHB-Ft. We performed comprehensive proteomic analysis of Ft grown in MHB, BHI, and BHI supplemented with casamino acids (BCA) and compared our findings to published “omics” data derived from Ft grown in vivo. Based on the abundance of ~1,000 proteins, the fingerprint of BHI-Ft is one of nutrient-deprived bacteria that—through induction of a stringent-starvation-like response—have induced the FevR regulon for expression of the bacterium's virulence factors, immuno-dominant antigens, and surface-carbohydrate synthases. To test the notion that increased abundance of dominant antigens expressed by BHI-Ft would render these bacteria more susceptible to pre-existing, vaccine-induced immunity, we employed a battery of LVS-vaccination and S4-challenge protocols using MHB- and BHI-grown Ft S4. Contrary to our hypothesis, these experiments reveal that LVS-immunization provides a barrier to infection that is significantly more effective against an MHB-S4 challenge than a BHI-S4 challenge. The differences in apparent virulence to immunized mice are profoundly greater than those observed with primary infection of naïve mice. Our findings suggest that tularemia vaccination studies should be critically evaluated in regard to the growth conditions of the challenge agent.
Collapse
Affiliation(s)
- Kristen M Holland
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Sarah J Rosa
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | | | - Donald Wolfgeher
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicago, IL, United States
| | - Brian J Franz
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Tiffany M Zarrella
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Raju Sunagar
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Anju Singh
- Trudeau InstituteSaranac Lake, NY, United States
| | - Chandra S Bakshi
- Department of Microbiology and Immunology, New York Medical CollegeValhalla, NY, United States
| | - Prachi Namjoshi
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Eileen M Barry
- School of Medicine, University of MarylandBaltimore, MD, United States
| | | | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicago, IL, United States
| | - Edmund J Gosselin
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Douglas S Reed
- Center for Vaccine Research, University of PittsburghPittsburgh, PA, United States
| | - Karsten R O Hazlett
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| |
Collapse
|
11
|
Petersson E, Athlin S. Cat-bite-induced Francisella tularensis infection with a false-positive serological reaction for Bartonella quintana. JMM Case Rep 2017; 4:e005071. [PMID: 28348802 PMCID: PMC5361632 DOI: 10.1099/jmmcr.0.005071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/04/2016] [Indexed: 11/28/2022] Open
Abstract
Introduction. Tularaemia is caused by infection with Francisella tularensistransmitted via direct contact with an infected hare carcass or indirectly through the bites of vectors, but may be cat-bite-associated as well. Medical history and reliable diagnostic analysis are important in order to differentiate it from other cat-associated infections, e.g. Bartonella spp. Casepresentation. A healthy 56-year-old man was examined because of a cat-bite-associated ulceroglandular wound on his right thumb. Nineteen days after the cat bite occurred, a serology test was positive for anti-Bartonella quintana, but negative for anti-F. tularensis. Since Bartonella infections are rare in Sweden, another serology test was analysed 2 weeks later with a positive result for anti-F. tularensis. The patient was treated with doxycycline for 14 days and recovered. The patient was re-sampled after 18 months to obtain a convalescent sample. The acute and the convalescent samples were both analysed at a reference centre, with negative results for anti-Bartonella spp. this time. Conclusion. This case is enlightening about the importance of extending the medical history and re-sampling the patient for antibody detection when the clinical suspicion of cat-bite-associated tularaemia is high. The false-positive result for anti-B. quintana antibodies may have been due to technical issues with the assay, cross-reactivity or both.
Collapse
Affiliation(s)
| | - Simon Athlin
- Department of Infectious Diseases, Faculty of Medicine and Health, Örebro University , Örebro SE 701 82 , Sweden
| |
Collapse
|
12
|
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
|
13
|
Chandler JC, Sutherland MD, Harton MR, Molins CR, Anderson RV, Heaslip DG, Bosio CM, Belisle JT. Francisella tularensis LVS surface and membrane proteins as targets of effective post-exposure immunization for tularemia. J Proteome Res 2014; 14:664-75. [PMID: 25494920 PMCID: PMC4324441 DOI: 10.1021/pr500628k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Francisella tularensis causes disease (tularemia)
in a large number of mammals, including man. We previously demonstrated
enhanced efficacy of conventional antibiotic therapy for tularemia
by postexposure passive transfer of immune sera developed against
a F. tularensis LVS membrane protein fraction (MPF).
However, the protein composition of this immunogenic fraction was
not defined. Proteomic approaches were applied to define the protein
composition and identify the immunogens of MPF. MPF consisted of at
least 299 proteins and 2-D Western blot analyses using sera from MPF-immunized
and F. tularensis LVS-vaccinated mice coupled to
liquid chromatography–tandem mass spectrometry identified 24
immunoreactive protein spots containing 45 proteins. A reverse vaccinology
approach that applied labeling of F. tularensis LVS
surface proteins and bioinformatics was used to reduce the complexity
of potential target immunogens. Bioinformatics analyses of the immunoreactive
proteins reduced the number of immunogen targets to 32. Direct surface
labeling of F. tularensis LVS resulted in the identification
of 31 surface proteins. However, only 13 of these were reactive with
MPF and/or F. tularensis LVS immune sera. Collectively,
this use of orthogonal proteomic approaches reduced the complexity
of potential immunogens in MPF by 96% and allowed for prioritization
of target immunogens for antibody-based immunotherapies against tularemia.
Collapse
Affiliation(s)
- Jeffrey C Chandler
- Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Department of Microbiology, Immunology, and Pathology, Colorado State University , Campus Delivery 0922, Fort Collins 80523, Colorado, United States
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM, Casadevall A, Flynn J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin Immunol 2014; 26:588-600. [PMID: 25458990 PMCID: PMC4314354 DOI: 10.1016/j.smim.2014.10.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 10/09/2014] [Accepted: 10/10/2014] [Indexed: 12/24/2022]
Abstract
Mycobacterium tuberculosis remains a major public health burden. It is generally thought that while B cell- and antibody-mediated immunity plays an important role in host defense against extracellular pathogens, the primary control of intracellular microbes derives from cellular immune mechanisms. Studies on the immune regulatory mechanisms during infection with M. tuberculosis, a facultative intracellular organism, has established the importance of cell-mediated immunity in host defense during tuberculous infection. Emerging evidence suggest a role for B cell and humoral immunity in the control of intracellular pathogens, including obligatory species, through interactions with the cell-mediated immune compartment. Recent studies have shown that B cells and antibodies can significantly impact on the development of immune responses to the tubercle bacillus. In this review, we present experimental evidence supporting the notion that the importance of humoral and cellular immunity in host defense may not be entirely determined by the niche of the pathogen. A comprehensive approach that examines both humoral and cellular immunity could lead to better understanding of the immune response to M. tuberculosis.
Collapse
Affiliation(s)
- John Chan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Simren Mehta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sushma Bharrhan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yong Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jacqueline M Achkar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Arturo Casadevall
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - JoAnne Flynn
- Departments of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
15
|
Plzakova L, Kubelkova K, Krocova Z, Zarybnicka L, Sinkorova Z, Macela A. B cell subsets are activated and produce cytokines during early phases of Francisella tularensis LVS infection. Microb Pathog 2014; 75:49-58. [PMID: 25200734 DOI: 10.1016/j.micpath.2014.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 08/25/2014] [Accepted: 08/27/2014] [Indexed: 11/29/2022]
Abstract
Francisella tularensis, a facultative intracellular Gram-negative bacterium, causes the illness tularemia. The infection of mice with live vaccine strain is considered to be a model of human tularemia. F. tularensis infects predominantly such phagocytic cells as macrophages or neutrophils, but it also infects non-phagocytic hepatocytes, epithelial cells, and murine and human B cell lines. Based on work with the murine tularemia model, we report here that F. tularensis LVS infects peritoneal CD19(+) cells - exclusively B-1a cells - early after intraperitoneal infection in vivo. The peritoneal and consequently spleen CD19(+) cells are activated by the F. tularensis LVS infection to express the activation markers from MHC class II, CD25, CD54, CD69, and the co-stimulatory molecules CD80 and CD86. As early as 12 h post-infection, the peritoneal CD19(+) cells produce IFN-γ, IL-1β, IL-4, IL-6, IL-12, IL-17, IL-23, and TNF-α. The spleen CD19(+) cells respond to infection with some delay. Moreover, the F. tularensis infected A20 B cell line activates CD3(+) spleen cells isolated from naïve mice. Thus, the data presented here suggest that B cells have all the attributes to actively participate in the induction and regulation of the adaptive immune response during early stages of F. tularensis infection.
Collapse
Affiliation(s)
- Lenka Plzakova
- Institute of Molecular Pathology, Faculty of Military Health Sciences (FMHS), University of Defense (UO), Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Klara Kubelkova
- Centre of Advanced Studies, FMHS, UO, Hradec Kralove, Czech Republic
| | - Zuzana Krocova
- Institute of Molecular Pathology, Faculty of Military Health Sciences (FMHS), University of Defense (UO), Trebesska 1575, 500 01 Hradec Kralove, Czech Republic.
| | - Lenka Zarybnicka
- Department of Radiobiology, FMHS, UO, Hradec Kralove, Czech Republic
| | - Zuzana Sinkorova
- Department of Radiobiology, FMHS, UO, Hradec Kralove, Czech Republic
| | - Ales Macela
- Institute of Molecular Pathology, Faculty of Military Health Sciences (FMHS), University of Defense (UO), Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| |
Collapse
|
16
|
Dulay SB, Gransee R, Julich S, Tomaso H, O׳ Sullivan CK. Automated microfluidically controlled electrochemical biosensor for the rapid and highly sensitive detection of Francisella tularensis. Biosens Bioelectron 2014; 59:342-9. [DOI: 10.1016/j.bios.2014.03.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/08/2014] [Accepted: 03/11/2014] [Indexed: 11/29/2022]
|
17
|
Lu Z, Rynkiewicz MJ, Madico G, Li S, Yang CY, Perkins HM, Sompuram SR, Kodela V, Liu T, Morris T, Wang D, Roche MI, Seaton BA, Sharon J. B-cell epitopes in GroEL of Francisella tularensis. PLoS One 2014; 9:e99847. [PMID: 24968190 PMCID: PMC4072690 DOI: 10.1371/journal.pone.0099847] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/16/2014] [Indexed: 01/01/2023] Open
Abstract
The chaperonin protein GroEL, also known as heat shock protein 60 (Hsp60), is a prominent antigen in the human and mouse antibody response to the facultative intracellular bacterium Francisella tularensis (Ft), the causative agent of tularemia. In addition to its presumed cytoplasmic location, FtGroEL has been reported to be a potential component of the bacterial surface and to be released from the bacteria. In the current study, 13 IgG2a and one IgG3 mouse monoclonal antibodies (mAbs) specific for FtGroEL were classified into eleven unique groups based on shared VH-VL germline genes, and seven crossblocking profiles revealing at least three non-overlapping epitope areas in competition ELISA. In a mouse model of respiratory tularemia with the highly pathogenic Ft type A strain SchuS4, the Ab64 and N200 IgG2a mAbs, which block each other’s binding to and are sensitive to the same two point mutations in FtGroEL, reduced bacterial burden indicating that they target protective GroEL B-cell epitopes. The Ab64 and N200 epitopes, as well as those of three other mAbs with different crossblocking profiles, Ab53, N3, and N30, were mapped by hydrogen/deuterium exchange–mass spectrometry (DXMS) and visualized on a homology model of FtGroEL. This model was further supported by its experimentally-validated computational docking to the X-ray crystal structures of Ab64 and Ab53 Fabs. The structural analysis and DXMS profiles of the Ab64 and N200 mAbs suggest that their protective effects may be due to induction or stabilization of a conformational change in FtGroEL.
Collapse
Affiliation(s)
- Zhaohua Lu
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Michael J. Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Guillermo Madico
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sheng Li
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Chiou-Ying Yang
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Hillary M. Perkins
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Seshi R. Sompuram
- Medical Discovery Partners, LLC, Boston, Massachusetts, United States of America
| | - Vani Kodela
- Medical Discovery Partners, LLC, Boston, Massachusetts, United States of America
| | - Tong Liu
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Timothy Morris
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Daphne Wang
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, United States of America
| | - Marly I. Roche
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Barbara A. Seaton
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jacqueline Sharon
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
18
|
Sharon J, Rynkiewicz MJ, Lu Z, Yang CY. Discovery of protective B-cell epitopes for development of antimicrobial vaccines and antibody therapeutics. Immunology 2014; 142:1-23. [PMID: 24219801 PMCID: PMC3992043 DOI: 10.1111/imm.12213] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 11/05/2013] [Accepted: 11/08/2013] [Indexed: 01/07/2023] Open
Abstract
Protective antibodies play an essential role in immunity to infection by neutralizing microbes or their toxins and recruiting microbicidal effector functions. Identification of the protective B-cell epitopes, those parts of microbial antigens that contact the variable regions of the protective antibodies, can lead to development of antibody therapeutics, guide vaccine design, enable assessment of protective antibody responses in infected or vaccinated individuals, and uncover or localize pathogenic microbial functions that could be targeted by novel antimicrobials. Monoclonal antibodies are required to link in vivo or in vitro protective effects to specific epitopes and may be obtained from experimental animals or from humans, and their binding can be localized to specific regions of antigens by immunochemical assays. The epitopes are then identified with mapping methods such as X-ray crystallography of antigen-antibody complexes, antibody inhibition of hydrogen-deuterium exchange in the antigen, antibody-induced alteration of the nuclear magnetic resonance spectrum of the antigen, and experimentally validated computational docking of antigen-antibody complexes. The diversity in shape, size and structure of protective B-cell epitopes, and the increasing importance of protective B-cell epitope discovery to development of vaccines and antibody therapeutics are illustrated through examples from different microbe categories, with emphasis on epitopes targeted by broadly neutralizing antibodies to pathogens of high antigenic variation. Examples include the V-shaped Ab52 glycan epitope in the O-antigen of Francisella tularensis, the concave CR6261 peptidic epitope in the haemagglutinin stem of influenza virus H1N1, and the convex/concave PG16 glycopeptidic epitope in the gp120 V1/V2 loop of HIV type 1.
Collapse
MESH Headings
- Animals
- Antibodies, Bacterial/immunology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antigen-Antibody Reactions
- Antigens, Bacterial/chemistry
- Antigens, Bacterial/immunology
- Antigens, Viral/chemistry
- Antigens, Viral/immunology
- Bacterial Vaccines/immunology
- Bacterial Vaccines/therapeutic use
- Epitope Mapping
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/immunology
- Humans
- Models, Molecular
- Protein Conformation
- Viral Vaccines/immunology
- Viral Vaccines/therapeutic use
Collapse
Affiliation(s)
- Jacqueline Sharon
- Department of Pathology and Laboratory Medicine, Boston University School of MedicineBoston, MA, USA
| | - Michael J Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of MedicineBoston, MA, USA
| | - Zhaohua Lu
- Department of Pathology and Laboratory Medicine, Boston University School of MedicineBoston, MA, USA
| | - Chiou-Ying Yang
- Department of Pathology and Laboratory Medicine, Boston University School of MedicineBoston, MA, USA
| |
Collapse
|
19
|
Boisset S, Caspar Y, Sutera V, Maurin M. New therapeutic approaches for treatment of tularaemia: a review. Front Cell Infect Microbiol 2014; 4:40. [PMID: 24734221 PMCID: PMC3975101 DOI: 10.3389/fcimb.2014.00040] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 03/13/2014] [Indexed: 12/23/2022] Open
Abstract
Antibiotic treatment of tularaemia is based on a few drugs, including the fluoroquinolones (e.g., ciprofloxacin), the tetracyclines (e.g., doxycycline), and the aminoglycosides (streptomycin and gentamicin). Because no effective and safe vaccine is currently available, tularaemia prophylaxis following proven exposure to F. tularensis also relies on administration of antibiotics. A number of reasons make it necessary to search for new therapeutic alternatives: the potential toxicity of first-line drugs, especially in children and pregnant women; a high rate of treatment relapses and failures, especially for severe and/or suppurated forms of the disease; and the possible use of antibiotic-resistant strains in the context of a biological threat. This review presents novel therapeutic approaches that have been explored in recent years to improve tularaemia patients' management and prognosis. These new strategies have been evaluated in vitro, in axenic media and cell culture systems and/or in animal models. First, the activities of newly available antibiotic compounds were evaluated against F. tularensis, including tigecycline (a glycylcycline), ketolides (telithromycin and cethromycin), and fluoroquinolones (moxifloxacin, gatifloxacin, trovafloxacin and grepafloxacin). The liposome delivery of some antibiotics was evaluated. The effect of antimicrobial peptides against F. tularensis was also considered. Other drugs were evaluated for their ability to suppress the intracellular multiplication of F. tularensis. The effects of the modulation of the innate immune response (especially via TLR receptors) on the course of F. tularensis infection was characterized. Another approach was the administration of specific antibodies to induce passive resistance to F. tularensis infection. All of these studies highlight the need to develop new therapeutic strategies to improve the management of patients with tularaemia. Many possibilities exist, some unexplored. Moreover, it is likely that new therapeutic alternatives that are effective against this intracellular pathogen could be, at least partially, extrapolated to other human pathogens.
Collapse
Affiliation(s)
- Sandrine Boisset
- Laboratoire de Bactériologie, Institut de Biologie et de Pathologie, CHU de Grenoble Grenoble, France ; Université Joseph Fourier-Grenoble 1 Grenoble, France ; Laboratoire Adaptation et Pathogénie des Micro-Organismes, CNRS/UJF, UMR 5163 Grenoble, France
| | - Yvan Caspar
- Laboratoire de Bactériologie, Institut de Biologie et de Pathologie, CHU de Grenoble Grenoble, France ; Université Joseph Fourier-Grenoble 1 Grenoble, France ; Laboratoire Adaptation et Pathogénie des Micro-Organismes, CNRS/UJF, UMR 5163 Grenoble, France
| | - Vivien Sutera
- Laboratoire de Bactériologie, Institut de Biologie et de Pathologie, CHU de Grenoble Grenoble, France ; Université Joseph Fourier-Grenoble 1 Grenoble, France ; Laboratoire Adaptation et Pathogénie des Micro-Organismes, CNRS/UJF, UMR 5163 Grenoble, France
| | - Max Maurin
- Laboratoire de Bactériologie, Institut de Biologie et de Pathologie, CHU de Grenoble Grenoble, France ; Université Joseph Fourier-Grenoble 1 Grenoble, France ; Laboratoire Adaptation et Pathogénie des Micro-Organismes, CNRS/UJF, UMR 5163 Grenoble, France
| |
Collapse
|
20
|
Ma Z, Banik S, Rane H, Mora VT, Rabadi SM, Doyle CR, Thanassi DG, Bakshi CS, Malik M. EmrA1 membrane fusion protein of Francisella tularensis LVS is required for resistance to oxidative stress, intramacrophage survival and virulence in mice. Mol Microbiol 2014; 91:976-95. [PMID: 24397487 DOI: 10.1111/mmi.12509] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2014] [Indexed: 01/11/2023]
Abstract
Francisella tularensis is a category A biodefence agent that causes a fatal human disease known as tularaemia. The pathogenicity of F. tularensis depends on its ability to persist inside host immune cells primarily by resisting an attack from host-generated reactive oxygen and nitrogen species (ROS/RNS). Based on the ability of F. tularensis to resist high ROS/RNS levels, we have hypothesized that additional unknown factors act in conjunction with known antioxidant defences to render ROS resistance. By screening a transposon insertion library of F. tularensis LVS in the presence of hydrogen peroxide, we have identified an oxidant-sensitive mutant in putative EmrA1 (FTL_0687) secretion protein. The results demonstrate that the emrA1 mutant is highly sensitive to oxidants and several antimicrobial agents, and exhibits diminished intramacrophage growth that can be restored to wild-type F. tularensis LVS levels by either transcomplementation, inhibition of ROS generation or infection in NADPH oxidase deficient (gp91Phox(-/-)) macrophages. The emrA1 mutant is attenuated for virulence, which is restored by infection in gp91Phox(-/-) mice. Further, EmrA1 contributes to oxidative stress resistance by affecting secretion of Francisella antioxidant enzymes SodB and KatG. This study exposes unique links between transporter activity and the antioxidant defence mechanisms of F. tularensis.
Collapse
Affiliation(s)
- Zhuo Ma
- Department of Basic and Social Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lu Z, Rynkiewicz MJ, Yang CY, Madico G, Perkins HM, Wang Q, Costello CE, Zaia J, Seaton BA, Sharon J. The binding sites of monoclonal antibodies to the non-reducing end of Francisella tularensis O-antigen accommodate mainly the terminal saccharide. Immunology 2013; 140:374-89. [PMID: 23844703 DOI: 10.1111/imm.12150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/19/2013] [Accepted: 07/05/2013] [Indexed: 11/27/2022] Open
Abstract
We have previously described two types of protective B-cell epitopes in the O-antigen (OAg) of the Gram-negative bacterium Francisella tularensis: repeating internal epitopes targeted by the vast majority of anti-OAg monoclonal antibodies (mAbs), and a non-overlapping epitope at the non-reducing end targeted by the previously unique IgG2a mAb FB11. We have now generated and characterized three mAbs specific for the non-reducing end of F. tularensis OAg, partially encoded by the same variable region germline genes, indicating that they target the same epitope. Like FB11, the new mAbs, Ab63 (IgG3), N213 (IgG3) and N62 (IgG2b), had higher antigen-binding bivalent avidity than internally binding anti-OAg mAbs, and an oligosaccharide containing a single OAg repeat was sufficient for optimal inhibition of their antigen-binding. The X-ray crystal structure of N62 Fab showed that the antigen-binding site is lined mainly by aromatic amino acids that form a small cavity, which can accommodate no more than one and a third sugar residues, indicating that N62 binds mainly to the terminal Qui4NFm residue at the nonreducing end of OAg. In efficacy studies with mice infected intranasally with the highly virulent F. tularensis strain SchuS4, N62, N213 and Ab63 prolonged survival and reduced blood bacterial burden. These results yield insights into how antibodies to non-reducing ends of microbial polysaccharides can contribute to immune protection despite the smaller size of their target epitopes compared with antibodies to internal polysaccharide regions.
Collapse
Affiliation(s)
- Zhaohua Lu
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Francisella tularensis is a gram-negative bacterium that causes the zoonotic disease tularemia. Francisella is highly infectious via the respiratory route (~10 CFUs) and pulmonary infections due to type A strains of F. tularensis are highly lethal in untreated patients (>30%). In addition, no vaccines are licensed to prevent tularemia in humans. Due to the high infectivity and mortality of pulmonary tularemia, F. tularensis has been weaponized, including via the introduction of antibiotic resistance, by several countries. Because of the lack of efficacious vaccines, and concerns about F. tularensis acquiring resistance to antibiotics via natural or illicit means, augmentation of host immunity, and humoral immunotherapy have been investigated as countermeasures against tularemia. This manuscript will review advances made and challenges in the field of immunotherapy against tularemia.
Collapse
Affiliation(s)
- Jerod A Skyberg
- Department of Veterinary Pathobiology and Laboratory for Infectious Disease Research; University of Missouri; Columbia, MO USA
| |
Collapse
|
23
|
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: 2.1] [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
|
24
|
Crane DD, Griffin AJ, Wehrly TD, Bosio CM. B1a cells enhance susceptibility to infection with virulent Francisella tularensis via modulation of NK/NKT cell responses. THE JOURNAL OF IMMUNOLOGY 2013; 190:2756-66. [PMID: 23378429 DOI: 10.4049/jimmunol.1202697] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B1a cells are an important source of natural Abs, Abs directed against T-independent Ags, and are a primary source of IL-10. Bruton's tyrosine kinase (btk) is a cytoplasmic kinase that is essential for mediating signals from the BCR and is critical for development of B1a cells. Consequentially, animals lacking btk have few B1a cells, minimal Ab responses, and can preferentially generate Th1-type immune responses following infection. B1a cells have been shown to aid in protection against infection with attenuated Francisella tularensis, but their role in infection mediated by fully virulent F. tularensis is not known. Therefore, we used mice with defective btk (CBA/CaHN-Btk(XID)/J [XID mice]) to determine the contribution of B1a cells in defense against the virulent F. tularensis ssp. tularensis strain SchuS4. Surprisingly, XID mice displayed increased resistance to pulmonary infection with F. tularensis. Specifically, XID mice had enhanced clearance of bacteria from the lung and spleen and significantly greater survival of infection compared with wild-type controls. We revealed that resistance to infection in XID mice was associated with decreased numbers of IL-10-producing B1a cells and concomitant increased numbers of IL-12-producing macrophages and IFN-γ-producing NK/NKT cells. Adoptive transfer of wild-type B1a cells into XID mice reversed the control of bacterial replication. Similarly, depletion of NK/NKT cells also increased bacterial burdens in XID mice. Together, our data suggest B cell-NK/NKT cell cross-talk is a critical pivot controlling survival of infection with virulent F. tularensis.
Collapse
Affiliation(s)
- Deborah D Crane
- Immunity to Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Hamilton, MT 59840, USA
| | | | | | | |
Collapse
|
25
|
Production of outer membrane vesicles and outer membrane tubes by Francisella novicida. J Bacteriol 2012; 195:1120-32. [PMID: 23264574 DOI: 10.1128/jb.02007-12] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella spp. are highly infectious and virulent bacteria that cause the zoonotic disease tularemia. Knowledge is lacking for the virulence factors expressed by Francisella and how these factors are secreted and delivered to host cells. Gram-negative bacteria constitutively release outer membrane vesicles (OMV), which may function in the delivery of virulence factors to host cells. We identified growth conditions under which Francisella novicida produces abundant OMV. Purification of the vesicles revealed the presence of tube-shaped vesicles in addition to typical spherical OMV, and examination of whole bacteria revealed the presence of tubes extending out from the bacterial surface. Recently, both prokaryotic and eukaryotic cells have been shown to produce membrane-enclosed projections, termed nanotubes, which appear to function in cell-cell communication and the exchange of molecules. In contrast to these previously characterized structures, the F. novicida tubes are produced in liquid as well as on solid medium and are derived from the OM rather than the cytoplasmic membrane. The production of the OMV and tubes (OMV/T) by F. novicida was coordinately regulated and responsive to both growth medium and growth phase. Proteomic analysis of purified OMV/T identified known Francisella virulence factors among the constituent proteins, suggesting roles for the vesicles in pathogenesis. In support of this, production of OM tubes by F. novicida was stimulated during infection of macrophages and addition of purified OMV/T to macrophages elicited increased release of proinflammatory cytokines. Finally, vaccination with purified OMV/T protected mice from subsequent challenge with highly lethal doses of F. novicida.
Collapse
|
26
|
Kuznetsova EM, Volokh OA, Shepelev IA, Nikiforov AK. Components of the Francisella tularensis protective antigen complex. MOLECULAR GENETICS MICROBIOLOGY AND VIROLOGY 2012. [DOI: 10.3103/s0891416812030044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Peng Y, Zhang Y, Mitchell WJ, Zhang G. Development of a lipopolysaccharide-targeted peptide mimic vaccine against Q fever. THE JOURNAL OF IMMUNOLOGY 2012; 189:4909-20. [PMID: 23053512 DOI: 10.4049/jimmunol.1201622] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Coxiella burnetii is a Gram-negative bacterium that causes acute and chronic Q fever in humans. Creation of a safe and effective new generation vaccine to prevent Q fever remains an important public health goal. Previous studies suggested that Ab-mediated immunity to C. burnetii phase I LPS (PI-LPS) is protective. To identify the potential peptides that can mimic the protective epitopes on PI-LPS, a PI-LPS-specific mAb 1E4 was generated, characterized, and used to screen a phage display library. Interestingly, our results indicate that 1E4 was able to inhibit C. burnetii infection in vivo, suggesting that 1E4 is a protective mAb. After three rounds of biopanning by 1E4 from the phage display library, a mimetic peptide, m1E41920, was identified, chemically synthesized, and conjugated to keyhole limpet hemocyanin (KLH) for examining its immunogenicity. The results indicate that the synthetic peptide m1E41920 was able to inhibit the binding of 1E4 to PI Ag, suggesting m1E41920 shares the same binding site of 1E4 with the epitopes of PI Ag. In addition, m1E41920-KLH elicited a specific IgG response to PI Ag, and immune sera from m1E41920-KLH-immunized mice was able to inhibit C. burnetii infection in vivo, suggesting that m1E41920 may specifically mimic the protective epitope of PI-LPS. Furthermore, m1E41920-KLH was able to confer significant protection against C. burnetii challenge. Thus, m1E41920-KLH is a protective Ag and may be useful for developing a safe and effective vaccine against Q fever. This study demonstrates the feasibility of developing a peptide mimic vaccine against Q fever.
Collapse
Affiliation(s)
- Ying Peng
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri-Columbia, Columbia, MO 65211, USA
| | | | | | | |
Collapse
|
28
|
Kuriakose JA, Zhang X, Luo T, McBride JW. Molecular basis of antibody mediated immunity against Ehrlichia chaffeensis involves species-specific linear epitopes in tandem repeat proteins. Microbes Infect 2012; 14:1054-63. [PMID: 22658957 PMCID: PMC3445803 DOI: 10.1016/j.micinf.2012.05.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 05/10/2012] [Accepted: 05/18/2012] [Indexed: 01/06/2023]
Abstract
Humoral immune mechanisms are an important component of protective immunity to Ehrlichia species. However, the molecular basis of antibody mediated immunity is not completely defined, and the role of most molecularly characterized major immunoreactive proteins is unknown. In previous studies, we mapped major species-specific continuous epitopes in three surface exposed and secreted tandem repeat proteins (TRP32, TRP47 and TRP120). In this study, we report that protection is provided by antibodies against these molecularly defined TRP epitopes using in vitro and in vivo models. Protection was demonstrated in vitro after prophylactic and therapeutic administration of epitope-specific anti-TRP antibodies, suggesting that the protective mechanisms involve extracellular and intracellular antibody-mediated effects. In vivo passive transfer of individual epitope-specific TRP sera significantly reduced the ehrlichial load and splenomegaly, and protected mice against lethal infection. Moreover, the combination of antibodies to all three TRPs provided enhanced reduction in ehrlichial load similar to that of Ehrlichia chaffeensis immune sera. IgG1 was the predominant antibody isotype in the epitope-specific TRP mouse sera. These results demonstrate that antibodies against linear epitopes in TRP32, TRP47 and TRP120 are protective during E. chaffeensis infection and involves extracellular and intracellular antibody-mediated mechanisms.
Collapse
Affiliation(s)
- Jeeba A. Kuriakose
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Xiaofeng Zhang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Tian Luo
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
29
|
Carrion J, Scisci E, Miles B, Sabino GJ, Zeituni AE, Gu Y, Bear A, Genco CA, Brown DL, Cutler CW. Microbial carriage state of peripheral blood dendritic cells (DCs) in chronic periodontitis influences DC differentiation, atherogenic potential. THE JOURNAL OF IMMUNOLOGY 2012; 189:3178-87. [PMID: 22891282 DOI: 10.4049/jimmunol.1201053] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The low-grade oral infection chronic periodontitis (CP) has been implicated in coronary artery disease risk, but the mechanisms are unclear. In this study, a pathophysiological role for blood dendritic cells (DCs) in systemic dissemination of oral mucosal pathogens to atherosclerotic plaques was investigated in humans. The frequency and microbiome of CD19(-)BDCA-1(+)DC-SIGN(+) blood myeloid DCs (mDCs) were analyzed in CP subjects with or without existing acute coronary syndrome and in healthy controls. FACS analysis revealed a significant increase in blood mDCs in the following order: healthy controls < CP < acute coronary syndrome/CP. Analysis of the blood mDC microbiome by 16S rDNA sequencing showed Porphyromonas gingivalis and other species, including (cultivable) Burkholderia cepacia. The mDC carriage rate with P. gingivalis correlated with oral carriage rate and with serologic exposure to P. gingivalis in CP subjects. Intervention (local debridement) to elicit a bacteremia increased the mDC carriage rate and frequency in vivo. In vitro studies established that P. gingivalis enhanced by 28% the differentiation of monocytes into immature mDCs; moreover, mDCs secreted high levels of matrix metalloproteinase-9 and upregulated C1q, heat shock protein 60, heat shock protein 70, CCR2, and CXCL16 transcripts in response to P. gingivalis in a fimbriae-dependent manner. Moreover, the survival of the anaerobe P. gingivalis under aerobic conditions was enhanced when within mDCs. Immunofluorescence analysis of oral mucosa and atherosclerotic plaques demonstrate infiltration with mDCs, colocalized with P. gingivalis. Our results suggest a role for blood mDCs in harboring and disseminating pathogens from oral mucosa to atherosclerosis plaques, which may provide key signals for mDC differentiation and atherogenic conversion.
Collapse
Affiliation(s)
- Julio Carrion
- School of Dental Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lu Z, Madico G, Roche MI, Wang Q, Hui JH, Perkins HM, Zaia J, Costello CE, Sharon J. Protective B-cell epitopes of Francisella tularensis O-polysaccharide in a mouse model of respiratory tularaemia. Immunology 2012; 136:352-60. [PMID: 22486311 DOI: 10.1111/j.1365-2567.2012.03589.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Antibodies to the lipopolysaccharide (LPS) of Francisella tularensis have been shown to be protective against respiratory tularaemia in mouse models, and we have previously described mouse monoclonal antibodies (mAbs) to non-overlapping terminal and internal epitopes of the F. tularensis LPS O-polysaccharide (OAg). In the current study, we used F. tularensis LPS oligosaccharides of defined OAg repeat length as molecular rulers in competition ELISA to demonstrate that the epitope targeted by the terminal OAg-binding mAb FB11 is contained within one tetrasaccharide repeat whereas the epitope targeted by the internal OAg-binding mAb Ab52 spans two tetrasaccharide repeats. Both mAbs conferred survival to BALB/c mice infected intranasally with the F. tularensis type B live vaccine strain and prolonged survival of BALB/c mice infected intranasally with the highly virulent F. tularensis type A strain SchuS4. The protective effects correlated with reduced bacterial burden in mAb-treated infected mice. These results indicate that an oligosaccharide with two OAg tetrasaccharide repeats covers both terminal and internal protective OAg epitopes, which may inform the design of vaccines for tularaemia. Furthermore, the FB11 and Ab52 mAbs could serve as reporters to monitor the response of vaccine recipients to protective B-cell epitopes of F. tularensis OAg.
Collapse
Affiliation(s)
- Zhaohua Lu
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kubelkova K, Krocova Z, Balonova L, Pejchal J, Stulik J, Macela A. Specific antibodies protect gamma-irradiated mice against Francisella tularensis infection. Microb Pathog 2012; 53:259-68. [PMID: 22841607 DOI: 10.1016/j.micpath.2012.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 01/27/2023]
Abstract
The role of antibodies in the course of Francisella tularensis (F. tularensis) infection is still a subject of debate. The understanding of the poorly described role of humoral immunity is more than important for the effort to develop effective prophylactic procedure against the infection with Francisella virulent strains. We utilized the model of gamma-irradiated mice for the studies of the protective role of anti-F. tularensis antibodies in order to partially eliminate cellular responses. The model of gamma-irradiated mice can also demonstrate the responses of immunocompromised host to intracellular bacterial infection. The gamma-irradiation by doses greater than 3 Gy completely impairs the resistance to infection and causes a disbalance of cytokine production in mice. In this study, we demonstrate that passive transfer of immune sera protected irradiated mice against subsequent infection with strains of F. tularensis subsp. holarctica. Naïve mice of BALB/c or C3H/CBi strains were subjected to passive transfer of sera obtained from immunized mice with live vaccine strain (LVS) F. tularensis LVS, F. tularensis subsp. holarctica strain 15, heat-killed F. tularensis LVS, or heat-killed strain 15 two hours before infection with lethal doses of LVS or strain 15. The passive transfer of sera obtained from immunized mice conferred full protection of naïve unirradiated as well as sublethally irradiated mice against low lethal doses of infection with F. tularensis LVS or strain 15, in all variants of the experiments. In addition, the passively protected mice that survived the primary infection with F. tularensis LVS were protected also against further secondary challenge with a highly virulent strain of F. tularensis subsp. tularensis SchuS4. Moreover, the first evidence of combination of successful passive transfer of immunity by specific antisera and subsequent active immunization of immunocompromised animals is demonstrated. In summary, we demonstrate that B cell-mediated effector responses together with the induction of T cell-mediated immunity both play an important role in naïve and also in immunocompromised mice and this fact it would be appropriate to take into the account in the design of new vaccines.
Collapse
Affiliation(s)
- Klara Kubelkova
- University of Defence, Faculty of Military Health Sciences, 1575 Trebesska, 500 01 Hradec Kralove, Czech Republic.
| | | | | | | | | | | |
Collapse
|
32
|
Rynkiewicz MJ, Lu Z, Hui JH, Sharon J, Seaton BA. Structural analysis of a protective epitope of the Francisella tularensis O-polysaccharide. Biochemistry 2012; 51:5684-94. [PMID: 22747335 DOI: 10.1021/bi201711m] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Francisella tularensis (Ft), the Gram-negative facultative intracellular bacterium that causes tularemia, is considered a biothreat because of its high infectivity and the high mortality rate of respiratory disease. The Ft lipopolysaccharide (Ft LPS) is thought to be a main protective antigen in mice and humans, and we have previously demonstrated the protective effect of the Ft LPS-specific monoclonal antibody Ab52 in a mouse model of respiratory tularemia. Immunochemical characterization has shown that the epitope recognized by Ab52 is contained within two internal repeat units of the O-polysaccharide [O-antigen (OAg)] of Ft LPS. To further localize the Ab52 epitope and understand the molecular interactions between the antibody and the saccharide, we determined the X-ray crystal structure of the Fab fragment of Ab52 and derived an antibody-antigen complex using molecular docking. The docked complex, refined through energy minimization, reveals an antigen binding site in the shape of a large canyon with a central pocket that accommodates a V-shaped epitope consisting of six sugar residues, α-D-GalpNAcAN(1→4)-α-D-GalpNAcAN(1→3)-β-D-QuipNAc(1→2)-β-D-Quip4NFm(1→4)-α-D-GalpNAcAN(1→4)-α-D-GalpNAcAN. These results inform the development of vaccines and immunotherapeutic/immunoprophylactic antibodies against Ft by suggesting a desired topology for binding of the antibody to internal epitopes of Ft LPS. This is the first report of an X-ray crystal structure of a monoclonal antibody that targets a protective Ft B cell epitope.
Collapse
Affiliation(s)
- Michael J Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
33
|
Crane DD, Scott DP, Bosio CM. Generation of a convalescent model of virulent Francisella tularensis infection for assessment of host requirements for survival of tularemia. PLoS One 2012; 7:e33349. [PMID: 22428026 PMCID: PMC3299770 DOI: 10.1371/journal.pone.0033349] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/12/2012] [Indexed: 01/04/2023] Open
Abstract
Francisella tularensis is a facultative intracellular bacterium and the causative agent of tularemia. Development of novel vaccines and therapeutics for tularemia has been hampered by the lack of understanding of which immune components are required to survive infection. Defining these requirements for protection against virulent F. tularensis, such as strain SchuS4, has been difficult since experimentally infected animals typically die within 5 days after exposure to as few as 10 bacteria. Such a short mean time to death typically precludes development, and therefore assessment, of immune responses directed against virulent F. tularensis. To enable identification of the components of the immune system that are required for survival of virulent F. tularensis, we developed a convalescent model of tularemia in C57Bl/6 mice using low dose antibiotic therapy in which the host immune response is ultimately responsible for clearance of the bacterium. Using this model we demonstrate αβTCR+ cells, γδTCR+ cells, and B cells are necessary to survive primary SchuS4 infection. Analysis of mice deficient in specific soluble mediators shows that IL-12p40 and IL-12p35 are essential for survival of SchuS4 infection. We also show that IFN-γ is required for survival of SchuS4 infection since mice lacking IFN-γR succumb to disease during the course of antibiotic therapy. Finally, we found that both CD4+ and CD8+ cells are the primary producers of IFN-γand that γδTCR+ cells and NK cells make a minimal contribution toward production of this cytokine throughout infection. Together these data provide a novel model that identifies key cells and cytokines required for survival or exacerbation of infection with virulent F. tularensis and provides evidence that this model will be a useful tool for better understanding the dynamics of tularemia infection.
Collapse
Affiliation(s)
- Deborah D. Crane
- Immunity to Pulmonary Pathogens, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID, National Institutes of Health, Hamilton, Montana, United States of America
| | - Dana P. Scott
- Veterinary Pathology Section, Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID, National Institutes of Health, Hamilton, Montana, United States of America
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| |
Collapse
|
34
|
Evaluation of the immune response induced by DNA vaccines expressing MIF and MCD-1 genes ofTrichinella spiralisin BALB/c mice. J Helminthol 2011; 86:430-9. [DOI: 10.1017/s0022149x11000654] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AbstractPlasmids expressing macrophage migration inhibitory factor (MIF) ofTrichinella spiralis(TsMIF), multi-cystatin-like domain protein (MCD-1) ofT. spiralis(TsMCD-1), or co-expressingTsMIF andTsMCD-1 were constructed with a pVAX1 vector. Their ability to generate a protective immune response againstT. spiralisinfection was evaluated in BALB/c mice. Groups of mice were immunized twice at 2-week intervals with 100 μg of recombinant plasmids pVAX1-Tsmif, pVAX1-Tsmcd-1or pVAX1-Tsmif-Tsmcd-1. Control animals were immunized with phosphate-buffered saline (PBS) or blank vector plasmid. Specific antibody levels (IgG, IgG1, IgG2a, IgG2b, IgM, IgA, IgE) against the recombinant proteinTsMIF-TsMCD-1, serum cytokines (interferon (IFN)-γ, interleukin (IL)-4, IL-5, transforming growth factor (TGF)-β1 and IL-17) and CD4+/CD8+T cells were monitored. Challenge infection was performed 2 weeks following the second immunization and worm burden was assayed at 35 days post-challenge. Vaccination with pVAX1-Tsmifinduced moderate serum IFN-γ and increases of CD4+and CD8+T cells, but no specific immunoglobulin antibody response. Vaccination with pVAX1-Tsmcd-1induced a predominant Th1 antibody (IgG2a and IgG2b) response and strong levels of serum IFN-γ, and increases of CD4+T cells. Importantly, co-expression ofTsMIF andTsMCD-1 in DNA immunization produced more serum IFN-γ and markedly enhanced CD4+and CD8+T cells than the single DNA vaccine of the two genes. Challenge infection demonstrated that immunization with pVAX1-Tsmif-Tsmcd-1reduced worm burdens (by 23.17%;P < 0.05).
Collapse
|
35
|
Froude JW, Stiles B, Pelat T, Thullier P. Antibodies for biodefense. MAbs 2011; 3:517-27. [PMID: 22123065 PMCID: PMC3242838 DOI: 10.4161/mabs.3.6.17621] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/03/2011] [Indexed: 12/11/2022] Open
Abstract
Potential bioweapons are biological agents (bacteria, viruses, and toxins) at risk of intentional dissemination. Biodefense, defined as development of therapeutics and vaccines against these agents, has seen an increase, particularly in the US following the 2001 anthrax attack. This review focuses on recombinant antibodies and polyclonal antibodies for biodefense that have been accepted for clinical use. These antibodies aim to protect against primary potential bioweapons, or category A agents as defined by the Centers for Disease Control and Prevention (Bacillus anthracis, Yersinia pestis, Francisella tularensis, botulinum neurotoxins, smallpox virus, and certain others causing viral hemorrhagic fevers) and certain category B agents. Potential for prophylactic use is presented, as well as frequent use of oligoclonal antibodies or synergistic effect with other molecules. Capacities and limitations of antibodies for use in biodefense are discussed, and are generally applicable to the field of infectious diseases.
Collapse
Affiliation(s)
- Jeffrey W Froude
- US Army Medical Research and Material Command; Fort Detrick, MD USA
- Unité de biotechnologie des anticorps et des toxines; Département de Microbiologie; Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); La Tronche Cedex, France
| | - Bradley Stiles
- US Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD USA
| | - Thibaut Pelat
- Unité de biotechnologie des anticorps et des toxines; Département de Microbiologie; Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); La Tronche Cedex, France
| | - Philippe Thullier
- Unité de biotechnologie des anticorps et des toxines; Département de Microbiologie; Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); La Tronche Cedex, France
| |
Collapse
|
36
|
Cyge B, Fischer V, Takemaru KI, Li FQ. Generation and characterization of monoclonal antibodies against human Chibby protein. Hybridoma (Larchmt) 2011; 30:163-8. [PMID: 21529289 DOI: 10.1089/hyb.2010.0098] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chibby (Cby) was originally identified as an antagonist of the Wnt/β-catenin signaling pathway. It physically interacts with the key co-activator β-catenin and inhibits β-catenin-mediated transcriptional activation. More recently, we demonstrated that Cby protein localizes to the base of motile cilia and is required for ciliogenesis in the respiratory epithelium of mice. To gain further insight into the physiological function of Cby, we developed mouse monoclonal antibodies (MAbs) against human Cby protein and characterized two Cby MAbs, designated 8-2 and 27-11, in depth. Western blot analysis revealed that 8-2 reacts with both human and mouse Cby proteins, whereas 27-11 is specific to human Cby. The epitopes of 8-2 and 27-11 were narrowed down to the middle portion (aa 49-63) and N-terminal region (aa 1-31) of the protein, respectively. We also determined their isotypes and found that 8-2 and 27-11 belong to IgG2a and IgG1 with κ light chains, respectively. Both MAbs can be employed for immunoprecipitation assays. Moreover, 8-2 detects endogenous Cby protein on Western blots, and marks the ciliary base of motile cilia in the murine lung and trachea as shown by immunofluorescence staining. These Cby MAbs therefore hold promise as useful tools for the investigation of Wnt signaling and ciliogenesis.
Collapse
Affiliation(s)
- Benjamin Cyge
- Graduate Program in Molecular and Cellular Pharmacology, Department of Pharmacological Sciences, SUNY at Stony Brook, Stony Brook, New York, USA
| | | | | | | |
Collapse
|
37
|
Hickey AJ, Hazlett KRO, Kirimanjeswara GS, Metzger DW. Identification of Francisella tularensis outer membrane protein A (FopA) as a protective antigen for tularemia. Vaccine 2011; 29:6941-7. [PMID: 21803089 DOI: 10.1016/j.vaccine.2011.07.075] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/08/2011] [Accepted: 07/11/2011] [Indexed: 12/31/2022]
Abstract
Francisella tularensis is a highly pathogenic gram negative bacterium that infects multiple sites in a host, including the skin and the respiratory tract, which can lead to the onset of a deadly disease with a 50% mortality rate. The live vaccine strain (LVS) of F. tularensis, while attenuated in humans but still virulent in mice, is not an option for vaccine use in the United States due to safety concerns, and currently no FDA approved vaccine exists. The purpose of the present work was to assess the ability of recombinant Francisella outer membrane protein A (FopA) to induce a protective response in mice. The gene encoding FopA from F. tularensis LVS was cloned and expressed in Escherichia coli. The resulting recombinant protein was affinity-purified from the E. coli outer membrane, incorporated into liposomes and administered to mice via multiple routes. FopA-immunized mice produced FopA-specific antibodies and were protected against both lethal intradermal and intranasal challenges with F. tularensis LVS. The vaccinated mice had reduced bacterial numbers in their lungs, livers and spleens during infection, and complete bacterial clearance was observed by day 28 post infection. Passive transfer of FopA-immune serum protected naïve mice against lethal F. tularensis LVS challenge, showing that humoral immunity played an important role in vaccine efficacy. FopA-immunization was unable to protect against challenge with the fully virulent SchuS4 strain of F. tularensis; however, the findings demonstrate proof of principle that an immune response generated against a component of a subunit vaccine is protective against lethal respiratory and intradermal tularemia.
Collapse
Affiliation(s)
- Anthony J Hickey
- Albany Medical College, Center for Immunology and Microbial Disease, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
38
|
Zarrella TM, Singh A, Bitsaktsis C, Rahman T, Sahay B, Feustel PJ, Gosselin EJ, Sellati TJ, Hazlett KRO. Host-adaptation of Francisella tularensis alters the bacterium's surface-carbohydrates to hinder effectors of innate and adaptive immunity. PLoS One 2011; 6:e22335. [PMID: 21799828 PMCID: PMC3142145 DOI: 10.1371/journal.pone.0022335] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 06/27/2011] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The gram-negative bacterium Francisella tularensis survives in arthropods, fresh water amoeba, and mammals with both intracellular and extracellular phases and could reasonably be expected to express distinct phenotypes in these environments. The presence of a capsule on this bacterium has been controversial with some groups finding such a structure while other groups report that no capsule could be identified. Previously we reported in vitro culture conditions for this bacterium which, in contrast to typical methods, yielded a bacterial phenotype that mimics that of the bacterium's mammalian, extracellular phase. METHODS/FINDINGS SDS-PAGE and carbohydrate analysis of differentially-cultivated F. tularensis LVS revealed that bacteria displaying the host-adapted phenotype produce both longer polymers of LPS O-antigen (OAg) and additional HMW carbohydrates/glycoproteins that are reduced/absent in non-host-adapted bacteria. Analysis of wildtype and OAg-mutant bacteria indicated that the induced changes in surface carbohydrates involved both OAg and non-OAg species. To assess the impact of these HMW carbohydrates on the access of outer membrane constituents to antibody we used differentially-cultivated bacteria in vitro to immunoprecipitate antibodies directed against outer membrane moieties. We observed that the surface-carbohydrates induced during host-adaptation shield many outer membrane antigens from binding by antibody. Similar assays with normal mouse serum indicate that the induced HMW carbohydrates also impede complement deposition. Using an in vitro macrophage infection assay, we find that the bacterial HMW carbohydrate impedes TLR2-dependent, pro-inflammatory cytokine production by macrophages. Lastly we show that upon host-adaptation, the human-virulent strain, F. tularensis SchuS4 also induces capsule production with the effect of reducing macrophage-activation and accelerating tularemia pathogenesis in mice. CONCLUSION F. tularensis undergoes host-adaptation which includes production of multiple capsular materials. These capsules impede recognition of bacterial outer membrane constituents by antibody, complement, and Toll-Like Receptor 2. These changes in the host-pathogen interface have profound implications for pathogenesis and vaccine development.
Collapse
Affiliation(s)
- Tiffany M. Zarrella
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Anju Singh
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Constantine Bitsaktsis
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Tabassum Rahman
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Bikash Sahay
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Paul J. Feustel
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York, United States of America
| | - Edmund J. Gosselin
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Timothy J. Sellati
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Karsten R. O. Hazlett
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| |
Collapse
|
39
|
Cole LE, Mann BJ, Shirey KA, Richard K, Yang Y, Gearhart PJ, Chesko KL, Viscardi RM, Vogel SN. Role of TLR signaling in Francisella tularensis-LPS-induced, antibody-mediated protection against Francisella tularensis challenge. J Leukoc Biol 2011; 90:787-97. [PMID: 21750122 DOI: 10.1189/jlb.0111014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Immunization with Ft-LPS provokes an antigen-specific, B-1a cell-derived antibody response that protects WT mice against an otherwise lethal challenge with Ft LVS. However, this same regimen offers limited protection to TLR2(-/-) mice, despite production of WT levels of anti-Ft-LPS antibodies. As Ft-LPS exhibits no TLR2 agonist activity, and macrophage-induced cytokine production in response to Ft LVS is overwhelmingly TLR2-dependent, we hypothesized that treatment of TLR2(-/-) mice with an alternative, MyD88-dependent TLR agonist would compensate for reduced recognition of Ft LVS in TLR2(-/-) mice and thereby, restore Ft-LPS-mediated protection. Administration of the nontoxic TLR4 agonist, synthetic Escherichia coli MPL, at the time of Ft-LPS immunization or Ft LVS challenge, fully protected TLR2(-/-) mice, whereas treatment of WT or TLR2(-/-) mice with MPL alone conferred partial protection. The TLR5 agonist, flagellin, also synergized with Ft-LPS to protect TLR2(-/-) mice from lethal Ft LVS challenge. In contrast to Ft LVS, Ft-LPS pretreatment failed to protect mice against i.n. challenge with Ft Schu S4, whereas MPL, administered in the absence or presence of Ft-LPS, conferred significant, albeit partial, protection. MPL treatment of macrophages increased the uptake of Ft LVS and decreased intracellular bacterial survival while shifting the macrophage-differentiation phenotype from "alternatively activated" to "classically activated". Collectively, our data suggest that optimal, Ft-LPS-mediated protection against Ft LVS infection requires two discrete events, i.e., production of Ft-LPS-specific antibody, as well as TLR-mediated macrophage activation, to fully control Francisella infection.
Collapse
Affiliation(s)
- Leah E Cole
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pierson T, Matrakas D, Taylor YU, Manyam G, Morozov VN, Zhou W, van Hoek ML. Proteomic Characterization and Functional Analysis of Outer Membrane Vesicles of Francisella novicida Suggests Possible Role in Virulence and Use as a Vaccine. J Proteome Res 2011; 10:954-67. [DOI: 10.1021/pr1009756] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Tony Pierson
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
| | - Demetrios Matrakas
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
| | - Yuka U. Taylor
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
| | - Ganiraju Manyam
- Department of Bioinformatics & Computational Biology, The UT MD Anderson Cancer Center, Houston, Texas, United States
| | - Victor N. Morozov
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia 20110, United States
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia 20110, United States
| | - Monique L. van Hoek
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia 20110, United States
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia 20110, United States
| |
Collapse
|
41
|
Bosio CM. The subversion of the immune system by francisella tularensis. Front Microbiol 2011; 2:9. [PMID: 21687406 PMCID: PMC3109352 DOI: 10.3389/fmicb.2011.00009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 01/17/2011] [Indexed: 12/11/2022] Open
Abstract
Francisella tularensis is a highly virulent bacterial pathogen and the causative agent of tularemia. Perhaps the most impressive feature of this bacterium is its ability to cause lethal disease following inoculation of as few as 15 organisms. This remarkable virulence is, in part, attributed to the ability of this microorganism to evade, disrupt, and modulate host immune responses. The objective of this review is to discuss the mechanisms utilized by F. tularensis to evade and inhibit innate and adaptive immune responses. The capability of F. tularensis to interfere with developing immunity in the host was appreciated decades ago. Early studies in humans were the first to demonstrate the ability of F. tularensis to suppress innate immunity. This work noted that humans suffering from tularemia failed to respond to a secondary challenge of endotoxin isolated from unrelated bacteria. Further, anecdotal observations of individuals becoming repeatedly infected with virulent strains of F. tularensis suggests that this bacterium also interferes with the generation of adequate adaptive immunity. Recent advances utilizing the mouse model for in vivo studies and human cells for in vitro work have identified specific bacterial and host compounds that play a role in mediating ubiquitous suppression of the host immune response. Compilation of this work will undoubtedly aid in enhancing our understanding of the myriad of mechanisms utilized by virulent F. tularensis for successful infection, colonization, and pathogenesis in the mammalian host.
Collapse
Affiliation(s)
- Catharine M Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton, MT, USA
| |
Collapse
|
42
|
Antibodies contribute to effective vaccination against respiratory infection by type A Francisella tularensis strains. Infect Immun 2011; 79:1770-8. [PMID: 21282410 DOI: 10.1128/iai.00605-10] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pneumonic tularemia is a life-threatening disease caused by inhalation of the highly infectious intracellular bacterium Francisella tularensis. The most serious form of the disease associated with the type A strains can be prevented in experimental animals through vaccination with the attenuated live vaccine strain (LVS). The protection is largely cell mediated, but the contribution of antibodies remains controversial. We addressed this issue in a series of passive immunization studies in Fischer 344 (F344) rats. Subcutaneous LVS vaccination induced a robust serum antibody response dominated by IgM, IgG2a, and IgG2b antibodies. Prophylactic administration of LVS immune serum or purified immune IgG reduced the severity and duration of disease in naïve rats challenged intratracheally with a lethal dose of the virulent type A strain SCHU S4. The level of resistance increased with the volume of immune serum given, but the maximum survivable SCHU S4 challenge dose was at least 100-fold lower than that shown for LVS-vaccinated rats. Protection correlated with reduced systemic bacterial growth, less severe histopathology in the liver and spleen during the early phase of infection, and bacterial clearance by a T cell-dependent mechanism. Our results suggest that treatment with immune serum limited the sequelae associated with infection, thereby enabling a sterilizing T cell response to develop and resolve the infection. Thus, antibodies induced by LVS vaccination may contribute to the defense of F344 rats against respiratory infection by type A strains of F. tularensis.
Collapse
|
43
|
Stundick MV, Metz M, Sampath A, Larsen JC. State-of-the-art therapeutic medical countermeasures for bacterial threat agents. Drug Dev Res 2011. [DOI: 10.1002/ddr.20462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
44
|
Nieves W, Heang J, Asakrah S, Höner zu Bentrup K, Roy CJ, Morici LA. Immunospecific responses to bacterial elongation factor Tu during Burkholderia infection and immunization. PLoS One 2010; 5:e14361. [PMID: 21179405 PMCID: PMC3003680 DOI: 10.1371/journal.pone.0014361] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 11/15/2010] [Indexed: 01/18/2023] Open
Abstract
Burkholderia pseudomallei is the etiological agent of melioidosis, a disease endemic in parts of Southeast Asia and Northern Australia. Currently there is no licensed vaccine against infection with this biological threat agent. In this study, we employed an immunoproteomic approach and identified bacterial Elongation factor-Tu (EF-Tu) as a potential vaccine antigen. EF-Tu is membrane-associated, secreted in outer membrane vesicles (OMVs), and immunogenic during Burkholderia infection in the murine model of melioidosis. Active immunization with EF-Tu induced antigen-specific antibody and cell-mediated immune responses in mice. Mucosal immunization with EF-Tu also reduced lung bacterial loads in mice challenged with aerosolized B. thailandensis. Our data support the utility of EF-Tu as a novel vaccine immunogen against bacterial infection.
Collapse
Affiliation(s)
- Wildaliz Nieves
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Julie Heang
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Saja Asakrah
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Kerstin Höner zu Bentrup
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Chad J. Roy
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Lisa A. Morici
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
45
|
Correlates of protection induced by vaccination. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1055-65. [PMID: 20463105 DOI: 10.1128/cvi.00131-10] [Citation(s) in RCA: 1221] [Impact Index Per Article: 87.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This paper attempts to summarize current knowledge about immune responses to vaccines that correlate with protection. Although the immune system is redundant, almost all current vaccines work through antibodies in serum or on mucosa that block infection or bacteremia/viremia and thus provide a correlate of protection. The functional characteristics of antibodies, as well as quantity, are important. Antibody may be highly correlated with protection or synergistic with other functions. Immune memory is a critical correlate: effector memory for short-incubation diseases and central memory for long-incubation diseases. Cellular immunity acts to kill or suppress intracellular pathogens and may also synergize with antibody. For some vaccines, we have no true correlates, but only useful surrogates, for an unknown protective response.
Collapse
|
46
|
GroEL and lipopolysaccharide from Francisella tularensis live vaccine strain synergistically activate human macrophages. Infect Immun 2010; 78:1797-806. [PMID: 20123721 DOI: 10.1128/iai.01135-09] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis, the causative agent of tularemia, interacts with host cells of innate immunity in an atypical manner. For most Gram-negative bacteria, the release of lipopolysaccharide (LPS) from their outer membranes stimulates an inflammatory response. When LPS from the attenuated live vaccine strain (LVS) or the highly virulent Schu S4 strain of F. tularensis was incubated with human umbilical vein endothelial cells, neither species of LPS induced expression of the adhesion molecule E-selectin or secretion of the chemokine CCL2. Moreover, a high concentration (10 microg/ml) of LVS or Schu S4 LPS was required to stimulate production of CCL2 by human monocyte-derived macrophages (huMDM). A screen for alternative proinflammatory factors of F. tularensis LVS identified the heat shock protein GroEL as a potential candidate. Recombinant LVS GroEL at a concentration of 10 microg/ml elicited secretion of CXCL8 and CCL2 by huMDM through a TLR4-dependent mechanism. When 1 microg of LVS GroEL/ml was added to an equivalent amount of LVS LPS, the two components synergistically activated the huMDM to produce CXCL8. Schu S4 GroEL was less stimulatory than LVS GroEL and showed a lesser degree of synergy when combined with Schu S4 LPS. These findings suggest that the intrinsically low proinflammatory activity of F. tularensis LPS may be increased in the infected human host through interactions with other components of the bacterium.
Collapse
|
47
|
Crane DD, Warner SL, Bosio CM. A novel role for plasmin-mediated degradation of opsonizing antibody in the evasion of host immunity by virulent, but not attenuated, Francisella tularensis. THE JOURNAL OF IMMUNOLOGY 2009; 183:4593-600. [PMID: 19752236 DOI: 10.4049/jimmunol.0901655] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Opsonization by Abs represents a critical component of the host immune response against many pathogens. The mechanisms by which virulent microbes evade this protective response are not completely understood. In disease mediated by Francisella tularensis, Ab can effectively protect against infections with attenuated strains, for example, LVS, but not virulent strains such as SchuS4. Thus, it is likely that SchuS4 has mechanisms, which are not present in LVS, that allow evasion of opsonization by Ab, dampening the protective effects of these host molecules. Here we demonstrate that evasion of Ab-mediated opsonization and phagocytosis by the highly virulent SchuS4 is associated with its ability to bind the host serine protease plasmin. SchuS4, but not the closely related LVS, bound active plasmin. Plasmin bound SchuS4 degraded exogenous and opsonizing Abs, whereas LVS failed to do so. Furthermore, plasmin-mediated inhibition of Ab opsonization by SchuS4 also inhibited Ab-mediated uptake of this bacterium by macrophages. Ab-mediated uptake of uncoated and opsonized SchuS4 elicited a strong proinflammatory response in infected macrophages. However, plasmin-coated, opsonized SchuS4 poorly elicited production of these protective proinflammatory cytokines. This unique host-pathogen interplay is a novel immune evasion strategy utilized by virulent F. tularensis, and it provides one explanation for the ability of Ab to protect against attenuated, but not virulent, strains of F. tularensis. This mechanism may also represent a more common hereto unrecognized strategy by which virulent bacteria evade detection and clearance by Ig.
Collapse
Affiliation(s)
- Deborah D Crane
- Immunity to Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | | | | |
Collapse
|
48
|
Racine R, Winslow GM. IgM in microbial infections: taken for granted? Immunol Lett 2009; 125:79-85. [PMID: 19539648 DOI: 10.1016/j.imlet.2009.06.003] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 06/08/2009] [Accepted: 06/08/2009] [Indexed: 12/20/2022]
Abstract
Much has been learned about the structure, function, and production of IgM, since the antibody's initial characterization. It is widely accepted that IgM provides a first line of defense during microbial infections, prior to the generation of adaptive, high-affinity IgG responses that are important for long-lived immunity and immunological memory. Although IgM responses are commonly used as a measure of exposure to infectious diseases, it is perhaps surprising that the role of and requirement for IgM in many microbial infections has not been well explored in vivo. This is in part due to the lack of capabilities, until relatively recently, to evaluate the requirement for IgM in the absence of coincident IgG responses. Such evaluations are now possible, using gene-targeted mouse strains that produce only IgM, or isotype-switched IgG. A number of studies have revealed that IgM, produced either innately, or in response to antigen challenge, plays an important and perhaps under appreciated role in many microbial infections. Moreover, the characterization of the roles of various B cell subsets, in the production of IgM, and in host defense, has revealed important and divergent roles for B-1a and B-1b cells. This review will highlight studies in which IgM, in its own right, has been found to play an important role, not only in early immunity, but also in long-term protection, against a variety of microbial pathogens. Observations that long-lived IgM responses can be generated in vivo suggest that it may be feasible to target IgM production as part of vaccination strategies.
Collapse
Affiliation(s)
- Rachael Racine
- The Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY 12201-0509, United States
| | | |
Collapse
|