1
|
Schott S, Scheuer R, Ermoli F, Glatter T, Evguenieva-Hackenberg E, Diepold A. A ParDE toxin-antitoxin system is responsible for the maintenance of the Yersinia virulence plasmid but not for type III secretion-associated growth inhibition. Front Cell Infect Microbiol 2023; 13:1166077. [PMID: 37228670 PMCID: PMC10203498 DOI: 10.3389/fcimb.2023.1166077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
Many Gram-negative pathogens utilize the type III secretion system (T3SS) to translocate virulence-promoting effector proteins into eukaryotic host cells. The activity of this system results in a severe reduction of bacterial growth and division, summarized as secretion-associated growth inhibition (SAGI). In Yersinia enterocolitica, the T3SS and related proteins are encoded on a virulence plasmid. We identified a ParDE-like toxin-antitoxin system on this virulence plasmid in genetic proximity to yopE, encoding a T3SS effector. Effectors are strongly upregulated upon activation of the T3SS, indicating a potential role of the ParDE system in the SAGI or maintenance of the virulence plasmid. Expression of the toxin ParE in trans resulted in reduced growth and elongated bacteria, highly reminiscent of the SAGI. Nevertheless, the activity of ParDE is not causal for the SAGI. T3SS activation did not influence ParDE activity; conversely, ParDE had no impact on T3SS assembly or activity itself. However, we found that ParDE ensures the presence of the T3SS across bacterial populations by reducing the loss of the virulence plasmid, especially under conditions relevant to infection. Despite this effect, a subset of bacteria lost the virulence plasmid and regained the ability to divide under secreting conditions, facilitating the possible emergence of T3SS-negative bacteria in late acute and persistent infections.
Collapse
Affiliation(s)
- Saskia Schott
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Robina Scheuer
- Department of Microbiology and Molecular Biology, Justus Liebig University Gießen, Gießen, Germany
| | - Francesca Ermoli
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Timo Glatter
- Core Facility for Mass spectrometry & Proteomics, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | | | - Andreas Diepold
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| |
Collapse
|
2
|
Biryukov SS, Wu H, Dankmeyer JL, Rill NO, Klimko CP, Egland KA, Shoe JL, Hunter M, Fetterer DP, Qiu J, Davies ML, Bausch CL, Sullivan EJ, Luke T, Cote CK. Polyclonal Antibodies Derived from Transchromosomic Bovines Vaccinated with the Recombinant F1-V Vaccine Increase Bacterial Opsonization In Vitro and Protect Mice from Pneumonic Plague. Antibodies (Basel) 2023; 12:antib12020033. [PMID: 37218899 DOI: 10.3390/antib12020033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Plague is an ancient disease that continues to be of concern to both the public health and biodefense research communities. Pneumonic plague is caused by hematogenous spread of Yersinia pestis bacteria from a ruptured bubo to the lungs or by directly inhaling aerosolized bacteria. The fatality rate associated with pneumonic plague is significant unless effective antibiotic therapy is initiated soon after an early and accurate diagnosis is made. As with all bacterial pathogens, drug resistance is a primary concern when developing strategies to combat these Yersinia pestis infections in the future. While there has been significant progress in vaccine development, no FDA-approved vaccine strategy exists; thus, other medical countermeasures are needed. Antibody treatment has been shown to be effective in animal models of plague. We produced fully human polyclonal antibodies in transchromosomic bovines vaccinated with the recombinant F1-V plague vaccine. The resulting human antibodies opsonized Y. pestis bacteria in the presence of RAW264.7 cells and afforded significant protection to BALB/c mice after exposure to aerosolized Y. pestis. These data demonstrate the utility of this technology to produce large quantities of non-immunogenic anti-plague human antibodies to prevent or possibly treat pneumonic plague in human.
Collapse
Affiliation(s)
- Sergei S Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Hua Wu
- SAB Biotherapeutics, 2100 E 54th St. N, Sioux Falls, SD 57104, USA
| | - Jennifer L Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Nathaniel O Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Kristi A Egland
- SAB Biotherapeutics, 2100 E 54th St. N, Sioux Falls, SD 57104, USA
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - David P Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Ju Qiu
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Michael L Davies
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | | | - Eddie J Sullivan
- SAB Biotherapeutics, 2100 E 54th St. N, Sioux Falls, SD 57104, USA
| | - Thomas Luke
- SAB Biotherapeutics, 2100 E 54th St. N, Sioux Falls, SD 57104, USA
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| |
Collapse
|
3
|
Fang X, Kang L, Qiu YF, Li ZS, Bai Y. Yersinia enterocolitica in Crohn’s disease. Front Cell Infect Microbiol 2023; 13:1129996. [PMID: 36968108 PMCID: PMC10031030 DOI: 10.3389/fcimb.2023.1129996] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Increasing attention is being paid to the unique roles gut microbes play in both physiological and pathological processes. Crohn’s disease (CD) is a chronic, relapsing, inflammatory disease of the gastrointestinal tract with unknown etiology. Currently, gastrointestinal infection has been proposed as one initiating factor of CD. Yersinia enterocolitica, a zoonotic pathogen that exists widely in nature, is one of the most common bacteria causing acute infectious gastroenteritis, which displays clinical manifestations similar to CD. However, the specific role of Y. enterocolitica in CD is controversial. In this Review, we discuss the current knowledge on how Y. enterocolitica and derived microbial compounds may link to the pathogenesis of CD. We highlight examples of Y. enterocolitica-targeted interventions in the diagnosis and treatment of CD, and provide perspectives for future basic and translational investigations on this topic.
Collapse
Affiliation(s)
| | | | | | | | - Yu Bai
- *Correspondence: Zhao-Shen Li, ; Yu Bai,
| |
Collapse
|
4
|
Yang R, Atkinson S, Chen Z, Cui Y, Du Z, Han Y, Sebbane F, Slavin P, Song Y, Yan Y, Wu Y, Xu L, Zhang C, Zhang Y, Hinnebusch BJ, Stenseth NC, Motin VL. Yersinia pestis and Plague: some knowns and unknowns. ZOONOSES (BURLINGTON, MASS.) 2023; 3:5. [PMID: 37602146 PMCID: PMC10438918 DOI: 10.15212/zoonoses-2022-0040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Since its first identification in 1894 during the third pandemic in Hong Kong, there has been significant progress of understanding the lifestyle of Yersinia pestis, the pathogen that is responsible for plague. Although we now have some understanding of the pathogen's physiology, genetics, genomics, evolution, gene regulation, pathogenesis and immunity, there are many unknown aspects of the pathogen and its disease development. Here, we focus on some of the knowns and unknowns relating to Y. pestis and plague. We notably focus on some key Y. pestis physiological and virulence traits that are important for its mammal-flea-mammal life cycle but also its emergence from the enteropathogen Yersinia pseudotuberculosis. Some aspects of the genetic diversity of Y. pestis, the distribution and ecology of plague as well as the medical countermeasures to protect our population are also provided. Lastly, we present some biosafety and biosecurity information related to Y. pestis and plague.
Collapse
Affiliation(s)
- Ruifu Yang
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Steve Atkinson
- School of Life Sciences, Centre for Biomolecular Science, University of Nottingham, Nottingham, United Kingdom
| | - Ziqi Chen
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Yujun Cui
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Zongmin Du
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yanping Han
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Florent Sebbane
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Philip Slavin
- Division of History and Politics, University of Stirling, Stirling FK9 4LJ, UK
| | - Yajun Song
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yanfeng Yan
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yarong Wu
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Lei Xu
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Chutian Zhang
- College of Natural Resources and Environment, Northwest A&F University, Yangling 712100, China
| | - Yun Zhang
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - B. Joseph Hinnebusch
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, USA
| | - Nils Chr. Stenseth
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, N-0316 Oslo, Norway
| | - Vladimir L. Motin
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
5
|
Goldberg JB, Crisan CV, Luu JM. Pseudomonas aeruginosa Antivirulence Strategies: Targeting the Type III Secretion System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:257-280. [PMID: 36258075 DOI: 10.1007/978-3-031-08491-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The Pseudomonas aeruginosa type III secretion system (T3SS) is a complex molecular machine that delivers toxic proteins from the bacterial cytoplasm directly into host cells. This apparatus spans the inner and outer membrane and employs a needle-like structure that penetrates through the eucaryotic cell membrane into the host cell cytosol. The expression of the P. aeruginosa T3SS is highly regulated by environmental signals including low calcium and host cell contact. P. aeruginosa strains with mutations in T3SS genes are less pathogenic, suggesting that the T3SS is a virulence mechanism. Given that P. aeruginosa is naturally antibiotic resistant and multidrug resistant isolates are rapidly emerging, new antibiotics to target P. aeruginosa are needed. Furthermore, even if new antibiotics were to be developed, the timeline between when an antibiotic is released and resistance development is relatively short. Therefore, the concept of targeting virulence factors has garnered attention. So-called "antivirulence" approaches do not kill the microbe but instead focus on rendering it harmless and therefore unable to cause damage. Since these therapies target a particular system or pathway, the normal microbiome is unlikely to be affected and there is less concern about the spread to other microbes. Finally, and most importantly, since any antivirulence drug does not kill the microbe, there should be less selective pressure to develop resistance to these inhibitors. The P. aeruginosa T3SS has been well studied due to its importance for pathogenesis in numerous human and animal infections. Thus, many P. aeruginosa T3SS inhibitors have been described as potential antivirulence therapeutics, some of which have progressed to clinical trials.
Collapse
Affiliation(s)
- Joanna B Goldberg
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airway Disease Research, Emory University School of Medicine, Atlanta, GA, USA.
| | - Cristian V Crisan
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airway Disease Research, Emory University School of Medicine, Atlanta, GA, USA
| | - Justin M Luu
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airway Disease Research, Emory University School of Medicine, Atlanta, GA, USA
- Microbiology and Molecular Genetics Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| |
Collapse
|
6
|
Combinatorial Viral Vector-Based and Live Attenuated Vaccines without an Adjuvant to Generate Broader Immune Responses to Effectively Combat Pneumonic Plague. mBio 2021; 12:e0322321. [PMID: 34872353 PMCID: PMC8649767 DOI: 10.1128/mbio.03223-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mice immunized with a combination of an adenovirus vector (Ad5-YFV) and live-attenuated (LMA)-based vaccines were evaluated for protective efficacy against pneumonic plague. While the Ad5-YFV vaccine harbors a fusion cassette of three genes encoding YscF, F1, and LcrV, LMA represents a mutant of parental Yersinia pestis CO92 deleted for genes encoding Lpp, MsbB, and Ail. Ad5-YFV and LMA were either administered simultaneously (1-dose regimen) or 21 days apart in various orders and route of administration combinations (2-dose regimen). The 2-dose regimen induced robust immune responses to provide full protection to animals against parental CO92 and its isogenic F1 deletion mutant (CAF−) challenges during both short- and long-term studies. Mice intranasally (i.n.) immunized with Ad5-YFV first followed by LMA (i.n. or intramuscularly [i.m.]) had higher T- and B-cell proliferative responses and LcrV antibody titers than those in mice vaccinated with LMA (i.n. or i.m.) first ahead of Ad5-YFV (i.n.) during the long-term study. Specifically, the needle- and adjuvant-free vaccine combination (i.n.) is ideal for use in plague regions of endemicity. Conversely, with a 1-dose regimen, mice vaccinated with Ad5-YFV i.n. and LMA by the i.m. route provided complete protection to animals against CO92 and its CAF− mutant challenges and elicited Th1/Th2, as well as Th17 responses, making it suitable for emergency vaccination during a plague outbreak or bioterrorist attack. This is a first study in which a viral vector-based and live-attenuated vaccines were effectively used in combination, representing adjuvant- and/or needle-free immunization, with each vaccine triggering a distinct cellular immune response.
Collapse
|
7
|
Rosario-Acevedo R, Biryukov SS, Bozue JA, Cote CK. Plague Prevention and Therapy: Perspectives on Current and Future Strategies. Biomedicines 2021; 9:1421. [PMID: 34680537 PMCID: PMC8533540 DOI: 10.3390/biomedicines9101421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 01/14/2023] Open
Abstract
Plague, caused by the bacterial pathogen Yersinia pestis, is a vector-borne disease that has caused millions of human deaths over several centuries. Presently, human plague infections continue throughout the world. Transmission from one host to another relies mainly on infected flea bites, which can cause enlarged lymph nodes called buboes, followed by septicemic dissemination of the pathogen. Additionally, droplet inhalation after close contact with infected mammals can result in primary pneumonic plague. Here, we review research advances in the areas of vaccines and therapeutics for plague in context of Y. pestis virulence factors and disease pathogenesis. Plague continues to be both a public health threat and a biodefense concern and we highlight research that is important for infection mitigation and disease treatment.
Collapse
Affiliation(s)
| | | | | | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD 21702, USA; (R.R.-A.); (S.S.B.); (J.A.B.)
| |
Collapse
|
8
|
Swietnicki W. Secretory System Components as Potential Prophylactic Targets for Bacterial Pathogens. Biomolecules 2021; 11:892. [PMID: 34203937 PMCID: PMC8232601 DOI: 10.3390/biom11060892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/18/2023] Open
Abstract
Bacterial secretory systems are essential for virulence in human pathogens. The systems have become a target of alternative antibacterial strategies based on small molecules and antibodies. Strategies to use components of the systems to design prophylactics have been less publicized despite vaccines being the preferred solution to dealing with bacterial infections. In the current review, strategies to design vaccines against selected pathogens are presented and connected to the biology of the system. The examples are given for Y. pestis, S. enterica, B. anthracis, S. flexneri, and other human pathogens, and discussed in terms of effectiveness and long-term protection.
Collapse
Affiliation(s)
- Wieslaw Swietnicki
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. R. Weigla 12, 53-114 Wroclaw, Poland
| |
Collapse
|
9
|
Sebbane F, Lemaître N. Antibiotic Therapy of Plague: A Review. Biomolecules 2021; 11:724. [PMID: 34065940 PMCID: PMC8151713 DOI: 10.3390/biom11050724] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
Plague-a deadly disease caused by the bacterium Yersinia pestis-is still an international public health concern. There are three main clinical forms: bubonic plague, septicemic plague, and pulmonary plague. In all three forms, the symptoms appear suddenly and progress very rapidly. Early antibiotic therapy is essential for countering the disease. Several classes of antibiotics (e.g., tetracyclines, fluoroquinolones, aminoglycosides, sulfonamides, chloramphenicol, rifamycin, and β-lactams) are active in vitro against the majority of Y. pestis strains and have demonstrated efficacy in various animal models. However, some discrepancies have been reported. Hence, health authorities have approved and recommended several drugs for prophylactic or curative use. Only monotherapy is currently recommended; combination therapy has not shown any benefits in preclinical studies or case reports. Concerns about the emergence of multidrug-resistant strains of Y. pestis have led to the development of new classes of antibiotics and other therapeutics (e.g., LpxC inhibitors, cationic peptides, antivirulence drugs, predatory bacteria, phages, immunotherapy, host-directed therapy, and nutritional immunity). It is difficult to know which of the currently available treatments or therapeutics in development will be most effective for a given form of plague. This is due to the lack of standardization in preclinical studies, conflicting data from case reports, and the small number of clinical trials performed to date.
Collapse
Affiliation(s)
- Florent Sebbane
- Univ. Lille, Inserm, CNRS, Institut Pasteur Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Nadine Lemaître
- Univ. Lille, Inserm, CNRS, Institut Pasteur Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, F-59000 Lille, France
- Laboratoire de Bactériologie-Hygiène, Centre Hospitalier Universitaire Amiens Picardie, UR 4294, Agents Infectieux, Résistance et Chimiothérapie (AGIR), Université de Picardie Jules Verne, F-80000 Amiens, France
| |
Collapse
|
10
|
Cote CK, Biryukov SS, Klimko CP, Shoe JL, Hunter M, Rosario-Acevedo R, Fetterer DP, Moody KL, Meyer JR, Rill NO, Dankmeyer JL, Worsham PL, Bozue JA, Welkos SL. Protection Elicited by Attenuated Live Yersinia pestis Vaccine Strains against Lethal Infection with Virulent Y. pestis. Vaccines (Basel) 2021; 9:vaccines9020161. [PMID: 33669472 PMCID: PMC7920443 DOI: 10.3390/vaccines9020161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/22/2022] Open
Abstract
The etiologic agent of plague, Yersinia pestis, is a globally distributed pathogen which poses both a natural and adversarial threat. Due largely to the rapid course and high mortality of pneumonic plague, vaccines are greatly needed. Two-component protein vaccines have been unreliable and potentially vulnerable to vaccine resistance. We evaluated the safety and efficacy of eight live Y. pestis strains derived from virulent strains CO92 or KIM6+ and mutated in one or more virulence-associated gene(s) or cured of plasmid pPst. Stringent, single-dose vaccination allowed down-selection of the two safest and most protective vaccine candidates, CO92 mutants pgm- pPst- and ΔyscN. Both completely protected BALB/c mice against subcutaneous and aerosol challenge with Y. pestis. Strain CD-1 outbred mice were more resistant to bubonic (but not pneumonic) plague than BALB/c mice, but the vaccines elicited partial protection of CD-1 mice against aerosol challenge, while providing full protection against subcutaneous challenge. A ΔyscN mutant of the nonencapsulated C12 strain was expected to display antigens previously concealed by the capsule. C12 ΔyscN elicited negligible titers to F1 but comparable antibody levels to whole killed bacteria, as did CO92 ΔyscN. Although one dose of C12 ΔyscN was not protective, vaccination with two doses of either CO92 ΔyscN, or a combination of the ΔyscN mutants of C12 and CO92, protected optimally against lethal bubonic or pneumonic plague. Protection against encapsulated Y. pestis required inclusion of F1 in the vaccine and was associated with high anti-F1 titers.
Collapse
|
11
|
Antibody Opsonization Enhances Early Interactions between Yersinia pestis and Neutrophils in the Skin and Draining Lymph Node in a Mouse Model of Bubonic Plague. Infect Immun 2020; 89:IAI.00061-20. [PMID: 33077628 DOI: 10.1128/iai.00061-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 10/14/2020] [Indexed: 11/20/2022] Open
Abstract
Bubonic plague results when Yersinia pestis is deposited in the skin via the bite of an infected flea. Bacteria then traffic to the draining lymph node (dLN) where they replicate to large numbers. Without treatment, this infection can result in highly fatal septicemia. Several plague vaccine candidates are currently at various stages of development, but no licensed vaccine is available in the United States. Though polyclonal and monoclonal antibodies (Ab) can provide complete protection against bubonic plague in animal models, the mechanisms responsible for this antibody-mediated immunity (AMI) to Y. pestis remain poorly understood. Here, we examine the effects of Ab opsonization on Y. pestis interactions with phagocytes in vitro and in vivo Opsonization of Y. pestis with polyclonal antiserum modestly increased phagocytosis/killing by an oxidative burst of murine neutrophils in vitro Intravital microscopy (IVM) showed increased association of Ab-opsonized Y. pestis with neutrophils in the dermis in a mouse model of bubonic plague. IVM of popliteal LNs after intradermal (i.d.) injection of bacteria in the footpad revealed increased Y. pestis-neutrophil interactions and increased neutrophil crawling and extravasation in response to Ab-opsonized bacteria. Thus, despite only having a modest effect in in vitro assays, opsonizing Ab had a dramatic effect in vivo on Y. pestis-neutrophil interactions in the dermis and dLN very early after infection. These data shed new light on the importance of neutrophils in AMI to Y. pestis and may provide a new correlate of protection for evaluation of plague vaccine candidates.
Collapse
|
12
|
Hotinger JA, May AE. Antibodies Inhibiting the Type III Secretion System of Gram-Negative Pathogenic Bacteria. Antibodies (Basel) 2020; 9:antib9030035. [PMID: 32726928 PMCID: PMC7551047 DOI: 10.3390/antib9030035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Pathogenic bacteria are a global health threat, with over 2 million infections caused by Gram-negative bacteria every year in the United States. This problem is exacerbated by the increase in resistance to common antibiotics that are routinely used to treat these infections, creating an urgent need for innovative ways to treat and prevent virulence caused by these pathogens. Many Gram-negative pathogenic bacteria use a type III secretion system (T3SS) to inject toxins and other effector proteins directly into host cells. The T3SS has become a popular anti-virulence target because it is required for pathogenesis and knockouts have attenuated virulence. It is also not required for survival, which should result in less selective pressure for resistance formation against T3SS inhibitors. In this review, we will highlight selected examples of direct antibody immunizations and the use of antibodies in immunotherapy treatments that target the bacterial T3SS. These examples include antibodies targeting the T3SS of Pseudomonas aeruginosa, Yersinia pestis, Escherichia coli, Salmonella enterica, Shigella spp., and Chlamydia trachomatis.
Collapse
|
13
|
Kinoshita M, Shimizu M, Akiyama K, Kato H, Moriyama K, Sawa T. Epidemiological survey of serum titers from adults against various Gram-negative bacterial V-antigens. PLoS One 2020; 15:e0220924. [PMID: 32155175 PMCID: PMC7064248 DOI: 10.1371/journal.pone.0220924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/24/2020] [Indexed: 11/18/2022] Open
Abstract
The V-antigen, a virulence-associated protein, was first identified in Yersinia pestis more than half a century ago. Since then, other V-antigen homologs and orthologs have been discovered and are now considered as critical molecules for the toxic effects mediated by the type III secretion system during infections caused by various pathogenic Gram-negative bacteria. After purifying recombinant V-antigen proteins, including PcrV from Pseudomonas aeruginosa, LcrV from Yersinia, LssV from Photorhabdus luminescens, AcrV from Aeromonas salmonicida, and VcrV from Vibrio parahaemolyticus, we developed an enzyme-linked immunoabsorbent assay to measure titers against each V-antigen in sera collected from 186 adult volunteers. Different titer-specific correlation levels were determined for the five V-antigens. The anti-LcrV and anti-AcrV titers shared the highest correlation with each other with a correlation coefficient of 0.84. The next highest correlation coefficient was between anti-AcrV and anti-VcrV titers at 0.79, while the lowest correlation was found between anti-LcrV and anti-VcrV titers, which were still higher than 0.7. Sera from mice immunized with one of the five recombinant V-antigens displayed cross-antigenicity with some of the other four V-antigens, supporting the results from the human sera. Thus, the serum anti-V-antigen titer measurement system may be used for epidemiological investigations of various pathogenic Gram-negative bacteria.
Collapse
Affiliation(s)
- Mao Kinoshita
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaru Shimizu
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Akiyama
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideya Kato
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kiyoshi Moriyama
- Department of Anesthesiology, School of Medicine, Kyorin University, Tokyo, Japan
| | - Teiji Sawa
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- * E-mail:
| |
Collapse
|
14
|
Wei T, Gong J, Qu G, Wang M, Xu H. Interactions between Yersinia pestis V-antigen (LcrV) and human Toll-like receptor 2 (TLR2) in a modelled protein complex and potential mechanistic insights. BMC Immunol 2019; 20:48. [PMID: 31842739 PMCID: PMC6916237 DOI: 10.1186/s12865-019-0329-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022] Open
Abstract
Background Yersinia pestis, the etiological pathogen of plague, is capable of repressing the immune response of white blood cells to evade phagocytosis. The V-antigen (LcrV) was found to be involved in this process by binding to human Toll-like Receptor 2 (TLR2). The detailed mechanism behind this LcrV and TLR2 mediated immune response repression, however, is yet to be fully elucidated due to the lack of structural information. Results In this work, with protein structure modelling, we were able to construct a structure model of the heterotetramer of Y. pestis LcrV and human TLR2. Molecular dynamics simulation suggests the stability of this structure in aquatic environment. The LcrV model has a dumbbell-like structure with two globule domains (G1 at N-terminus and G2 away from membrane) connected with a coiled-coil linker (CCL) domain. The two horseshoe-shape TLR2 subunits form a V-shape structure, are not in direct contact with each other, and are held together by the LcrV homodimer. In this structure model, both the G1 and CCL domains are involved in the formation of LcrV homodimer, while all three domains are involved in LcrV-TLR2 binding. A mechanistic model was proposed based on this heterotetrameric structure model: The LcrV homodimer separates the TLR2 subunits to inhibit the dimerization of TLR2 and subsequent signal transfer for immune response; while LcrV could also inhibit the formation of heterodimers of TLR2 with other TLRs, and leads to immune response repression. Conclusions A heterotetrameric structure of Y. pestis LcrV and human TLR2 was modelled in this work. Analysis of this modelled structure showed its stability in aquatic environments and the role of LcrV domains and residues in protein-protein interaction. A mechanistic model for the role of LcrV in Y. pestis pathogenesis is raised based on this heterotetrameric structure model. This work provides a hypothesis of LcrV function, with which further experimental validation may elucidate the role of LcrV in human immune response repression.
Collapse
Affiliation(s)
- Tiandi Wei
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China.,Taishan College, Shandong University, Qingdao, China
| | - Jing Gong
- School of Life Sciences, Shandong University, Qingdao, China
| | - Guojing Qu
- Taishan College, Shandong University, Qingdao, China
| | - Mingyu Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China. .,Taishan College, Shandong University, Qingdao, China.
| | - Hai Xu
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| |
Collapse
|
15
|
Sawa T, Kinoshita M, Inoue K, Ohara J, Moriyama K. Immunoglobulin for Treating Bacterial Infections: One More Mechanism of Action. Antibodies (Basel) 2019; 8:antib8040052. [PMID: 31684203 PMCID: PMC6963986 DOI: 10.3390/antib8040052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 02/08/2023] Open
Abstract
The mechanisms underlying the effects of immunoglobulins on bacterial infections are thought to involve bacterial cell lysis via complement activation, phagocytosis via bacterial opsonization, toxin neutralization, and antibody-dependent cell-mediated cytotoxicity. Nevertheless, recent advances in the study of the pathogenicity of Gram-negative bacteria have raised the possibility of an association between immunoglobulin and bacterial toxin secretion. Over time, new toxin secretion systems like the type III secretion system have been discovered in many pathogenic Gram-negative bacteria. With this system, the bacterial toxins are directly injected into the cytoplasm of the target cell through a special secretory apparatus without any exposure to the extracellular environment, and therefore with no opportunity for antibodies to neutralize the toxin. However, antibodies against the V-antigen, which is located on the needle-shaped tip of the bacterial secretion apparatus, can inhibit toxin translocation, thus raising the hope that the toxin may be susceptible to antibody targeting. Because multi-drug resistant bacteria are now prevalent, inhibiting this secretion mechanism is an attractive alternative or adjunctive therapy against lethal bacterial infections. Thus, it is not unreasonable to define the blocking effect of anti-V-antigen antibodies as the fifth mechanism for immunoglobulin action against bacterial infections.
Collapse
Affiliation(s)
- Teiji Sawa
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Mao Kinoshita
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Keita Inoue
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Junya Ohara
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Kiyoshi Moriyama
- Department of Anesthesiology, Kyorin University School of Medicine, Tokyo 181-8611, Japan.
| |
Collapse
|
16
|
Kopylov PK, Svetoch TE, Ivanov SA, Kombarova TI, Perovskaya ON, Titareva GM, Anisimov AP. Characteristics of the Chromatographic Cleaning and Protectiveness of the LcrV Isoform of Yersinia pestis. APPL BIOCHEM MICRO+ 2019. [DOI: 10.1134/s0003683819040082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Daniel C, Dewitte A, Poiret S, Marceau M, Simonet M, Marceau L, Descombes G, Boutillier D, Bennaceur N, Bontemps-Gallo S, Lemaître N, Sebbane F. Polymorphism in the Yersinia LcrV Antigen Enables Immune Escape From the Protection Conferred by an LcrV-Secreting Lactococcus Lactis in a Pseudotuberculosis Mouse Model. Front Immunol 2019; 10:1830. [PMID: 31428104 PMCID: PMC6688116 DOI: 10.3389/fimmu.2019.01830] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/19/2019] [Indexed: 11/13/2022] Open
Abstract
Yersinioses caused by Yersinia pestis, Yersinia pseudotuberculosis, and Yersinia enterocolitica are significant concerns in human and veterinary health. The link between virulence and the potent LcrV antigen has prompted the latter's selection as a major component of anti-Yersinia vaccines. Here, we report that (i) the group of Yersinia species encompassing Y. pestis and Y. pseudotuberculosis produces at least five different clades of LcrV and (ii) vaccination of mice with an LcrV-secreting Lactococcus lactis only protected against Yersinia strains producing the same LcrV clade as that of used for vaccination. By vaccinating with engineered LcrVs and challenging mice with strains producing either type of LcrV or a LcrV mutated for regions of interest, we highlight key polymorphic residues responsible for the absence of cross-protection. Our results show that an anti-LcrV-based vaccine should contain multiple LcrV clades if protection against the widest possible array of Yersinia strains is sought.
Collapse
Affiliation(s)
- Catherine Daniel
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Amélie Dewitte
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Sabine Poiret
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Michaël Marceau
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Michel Simonet
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Laure Marceau
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Guillaume Descombes
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Denise Boutillier
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Nadia Bennaceur
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Sébastien Bontemps-Gallo
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Nadine Lemaître
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| | - Florent Sebbane
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
18
|
Picking WD, Barta ML. The Tip Complex: From Host Cell Sensing to Translocon Formation. Curr Top Microbiol Immunol 2019; 427:173-199. [PMID: 31218507 DOI: 10.1007/82_2019_171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Type III secretion systems are used by some Gram-negative bacteria to inject effector proteins into targeted eukaryotic cells for the benefit of the bacterium. The type III secretion injectisome is a complex nanomachine comprised of four main substructures including a cytoplasmic sorting platform, an envelope-spanning basal body, an extracellular needle and an exposed needle tip complex. Upon contact with a host cell, secretion is induced, resulting in the formation of a translocon pore in the host membrane. Translocon formation completes the conduit needed for effector secretion into the host cell. Control of type III secretion occurs in response to environmental signals, with the final signal being host cell contact. Secretion control occurs primarily at two sites-the cytoplasmic sorting platform, which determines secretion hierarchy, and the needle tip complex, which is critical for sensing and responding to environmental signals. The best-characterized injectisomes are those from Yersinia, Shigella and Salmonella species where there is a wealth of information on the tip complex and the two translocator proteins. Of these systems, the best characterized from a secretion regulation standpoint is Shigella. In the Shigella system, the tip complex and the first secreted translocon both contribute to secretion control and, thus, both are considered components of the tip complex. In this review, all three of these type III secretion systems are described with discussion focused on the structure and formation of the injectisome tip complex and what is known of the transition from nascent tip complex to assembled translocon pore.
Collapse
Affiliation(s)
- William D Picking
- Department of Pharmaceutical Chemistry, University of Kansas, 2030 Becker Drive, Lawrence, 66047, KS, USA.
| | - Michael L Barta
- Higuchi Biosciences, 2099 Constant Ave., Lawrence, 66047, KS, USA.,Catalent Pharma Solutions, 10245 Hickman Mills Drive, Kansas City, 64137, MO, USA
| |
Collapse
|
19
|
Abramov VM, Kosarev IV, Motin VL, Khlebnikov VS, Vasilenko RN, Sakulin VK, Machulin AV, Uversky VN, Karlyshev AV. Binding of LcrV protein from Yersinia pestis to human T-cells induces apoptosis, which is completely blocked by specific antibodies. Int J Biol Macromol 2018; 122:1062-1070. [PMID: 30218736 DOI: 10.1016/j.ijbiomac.2018.09.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 11/29/2022]
Abstract
The V antigen (LcrV) of the plague bacterium Yersinia pestis is a potent protective protein that is considered as a vaccine component for humans. LcrV mediates the delivery of Yop toxins into host cells and upregulates TLR2-dependent IL-10 production. Although LcrV can interact with the receptor-bound human interferon-γ (hIFN-γ), the significance of these interactions in plague pathogenesis is not known. In this study, we determined the parameters of specific interactions of LcrV and LcrV68-326 with primary human thymocytes and Jurkat T-leukemia cells in the presence of receptor-bound hIFN-γ. Although the C-terminal region of hIFN-γ contains a GRRA138-141 site needed for high-affinity binding of LcrV and LcrV68-326, in the hIFN-γ homodimer, these GRRA138-141 target sites becomes accessible for targeting by LcrV or LcrV68-326 only after immobilization of the hIFN-γ homodimer on the hIFN-γ receptors of thymocytes or Jurkat T-cells. The interaction of LcrV or LcrV68-326 with receptor-bound hIFN-γ on the thymocytes or Jurkat T-cells caused apoptosis of both cell types, which can be completely blocked by the addition of monoclonal antibodies specific to the LEEL32-35 and DEEI203-206 sites of LcrV. The ability of LcrV to utilize hIFN-γ is insidious and may account in part for the severe symptoms of plague in humans.
Collapse
Affiliation(s)
- Vyacheslav M Abramov
- Department of Immunology and Biodefence, Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Igor V Kosarev
- Department of Immunology and Biodefence, Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Vladimir L Motin
- Department of Pathology/Microbiology & Immunology, University of Texas Medical Branch, Galverston, TX 77555, USA
| | - Valentin S Khlebnikov
- Department of Immunology and Biodefence, Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Raisa N Vasilenko
- Department of Immunology and Biodefence, Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Vadim K Sakulin
- Department of Immunology and Biodefence, Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Andrey V Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino 142290, Russia.
| | - Andrey V Karlyshev
- Department of Science, Engineering and Computing, Kingston University, Kingston, UK
| |
Collapse
|
20
|
|
21
|
A phase I safety and immunogenicity dose escalation trial of plague vaccine, Flagellin/F1/V, in healthy adult volunteers (DMID 08-0066). Vaccine 2017; 35:6759-6765. [PMID: 29037578 DOI: 10.1016/j.vaccine.2017.09.070] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Intentional aerosolization of Yersinia pestis may result in pneumonic plague which is highly fatal if not treated early. METHODS We conducted a phase 1 randomized, double blind (within each group), placebo controlled, dose escalation trial to evaluate a plague vaccine, Flagellin/F1/V, in healthy adults aged 8 through 45years. Vaccine was administered intramuscularly on Days 0 and 28 at a dose of 1, 3, 6 or 10mcg. Subjects were observed for 4h after vaccination for cytokine release syndrome. Reactogenicity and adverse events (AE) were collected for 14 and 28days, respectively, after each vaccination. Serious AE were collected for the entire study. ELISA antibody and cytokines were measured at multiple time points. Subject's participation lasted 13months. RESULTS Sixty healthy subjects were enrolled; 52% males, 100% non-Hispanic, 91.7% white and mean age 30.8years. No severe reactogenicity events occurred; most AE were mild. No serious AE related to vaccine occurred. A dose response effect was observed to F1, V and flagellin. The peak ELISA IgG antibody titers (95% CI) after two 10mcg doses of vaccine were 260.0 (102.6-659.0) and 983.6 (317.3-3048.8), respectively, against F1 and V antigens. The 6mcg dose group provided similar titers. Titers were low for the placebo, 1mcg and 3mcg recipients. A positive antibody dose response was observed to F1, V and flagellin. Vaccine antigen specific serum IgE was not detected. There were no significant rises in serum or cellular cytokine responses and no significant IgG increase to flagellin after the second dose. CONCLUSION The Flagellin/F1/V vaccine exhibited a dose dependent increase in immunogenicity and was well tolerated at all doses. Antibody specific responses to F1, V and flagellin increased as dose increased. Given the results from this trial, testing higher doses of the vaccine may be merited.
Collapse
|
22
|
Grabowski B, Schmidt MA, Rüter C. Immunomodulatory Yersinia outer proteins (Yops)-useful tools for bacteria and humans alike. Virulence 2017; 8:1124-1147. [PMID: 28296562 DOI: 10.1080/21505594.2017.1303588] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human-pathogenic Yersinia produce plasmid-encoded Yersinia outer proteins (Yops), which are necessary to down-regulate anti-bacterial responses that constrict bacterial survival in the host. These Yops are effectively translocated directly from the bacterial into the target cell cytosol by the type III secretion system (T3SS). Cell-penetrating peptides (CPPs) in contrast are characterized by their ability to autonomously cross cell membranes and to transport cargo - independent of additional translocation systems. The recent discovery of bacterial cell-penetrating effector proteins (CPEs) - with the prototype being the T3SS effector protein YopM - established a new class of autonomously translocating immunomodulatory proteins. CPEs represent a vast source of potential self-delivering, anti-inflammatory therapeutics. In this review, we give an update on the characteristic features of the plasmid-encoded Yops and, based on recent findings, propose the further development of these proteins for potential therapeutic applications as natural or artificial cell-penetrating forms of Yops might be of value as bacteria-derived biologics.
Collapse
Affiliation(s)
- Benjamin Grabowski
- a Institute of Infectiology - Centre for Molecular Biology of Inflammation (ZMBE), University of Münster , Münster , Germany
| | - M Alexander Schmidt
- a Institute of Infectiology - Centre for Molecular Biology of Inflammation (ZMBE), University of Münster , Münster , Germany
| | - Christian Rüter
- a Institute of Infectiology - Centre for Molecular Biology of Inflammation (ZMBE), University of Münster , Münster , Germany
| |
Collapse
|
23
|
Verma SK, Tuteja U. Plague Vaccine Development: Current Research and Future Trends. Front Immunol 2016; 7:602. [PMID: 28018363 PMCID: PMC5155008 DOI: 10.3389/fimmu.2016.00602] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/30/2016] [Indexed: 02/05/2023] Open
Abstract
Plague is one of the world’s most lethal human diseases caused by Yersinia pestis, a Gram-negative bacterium. Despite overwhelming studies for many years worldwide, there is no safe and effective vaccine against this fatal disease. Inhalation of Y. pestis bacilli causes pneumonic plague, a fast growing and deadly dangerous disease. F1/LcrV-based vaccines failed to provide adequate protection in African green monkey model in spite of providing protection in mice and cynomolgus macaques. There is still no explanation for this inconsistent efficacy, and scientists leg behind to search reliable correlate assays for immune protection. These paucities are the main barriers to improve the effectiveness of plague vaccine. In the present scenario, one has to pay special attention to elicit strong cellular immune response in developing a next-generation vaccine against plague. Here, we review the scientific contributions and existing progress in developing subunit vaccines, the role of molecular adjuvants; DNA vaccines; live delivery platforms; and attenuated vaccines developed to counteract virulent strains of Y. pestis.
Collapse
Affiliation(s)
| | - Urmil Tuteja
- Microbiology Division, Defence Research & Development Establishment , Gwalior , India
| |
Collapse
|
24
|
Daniel C, Titecat M, Poiret S, Cayet D, Boutillier D, Simonet M, Sirard JC, Lemaître N, Sebbane F. Characterization of the protective immune response to Yersinia pseudotuberculosis infection in mice vaccinated with an LcrV-secreting strain of Lactococcus lactis. Vaccine 2016; 34:5762-5767. [PMID: 27742220 DOI: 10.1016/j.vaccine.2016.09.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Pseudotuberculosis is an infection caused by the bacterial enteropathogen Yersinia pseudotuberculosis and is considered to be a significant problem in veterinary medicine. We previously found that intranasal administration of a recombinant Lactococcus lactis strain that secretes the low-calcium response V (LcrV) antigen from Y. pseudotuberculosis (Ll-LcrV) confers protection against a lethal Y. pseudotuberculosis infection. Here, we aimed at characterizing the immunological basis of this LcrV-elicited protective response and at determining the duration of vaccine-induced immunity. METHODS Splenocytes from BALB/c mice intranasally immunized with Ll-LcrV or Ll as control were immunostained then analyzed by flow cytometry. Protection against a lethal intravenous injection of Y. pseudotuberculosis was also determined (i) in immunized BALB/c mice depleted or not of CD4+, CD8+ or CD25+ cells and (ii) in naïve BALB/c mice receiving serum from immunized mice by counting the number of bacteria in liver and spleen. Lastly, survival rate of immunized BALB/c mice following a lethal intravenous injection of Y. pseudotuberculosis was followed up to 9-months. RESULTS We found that T and B lymphocytes but not non-conventional lymphoid cells were affected by Ll-LcrV immunization. We also observed that depletion of CD4+ and CD25+ but not CD8+ cells in immunized mice eradicated protection against a lethal systemic Y. pseudotuberculosis infection, suggesting that activated CD4+ T lymphocytes are required for vaccine-induced protection. Adoptive transfer of LcrV-specific antibodies from Ll-LcrV-immunized animals significantly reduced the bacterial counts in the liver compared to non-vaccinated mice. Lastly, the protective immunity conferred by Ll-LcrV decreased slightly over time; nevertheless almost 60% of the mice survived a lethal bacterial challenge at 9months post-vaccination. CONCLUSION Mucosal vaccination of mice with Ll-LcrV induced cell- and antibody-mediated protective immunity against Y. pseudotuberculosis infection in the mouse and the protection is long-lasting.
Collapse
Affiliation(s)
- Catherine Daniel
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Marie Titecat
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Sabine Poiret
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Delphine Cayet
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Denise Boutillier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Michel Simonet
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Jean-Claude Sirard
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Nadine Lemaître
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Florent Sebbane
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France.
| |
Collapse
|
25
|
Sanapala S, Rahav H, Patel H, Sun W, Curtiss R. Multiple antigens of Yersinia pestis delivered by live recombinant attenuated Salmonella vaccine strains elicit protective immunity against plague. Vaccine 2016; 34:2410-2416. [PMID: 27060051 DOI: 10.1016/j.vaccine.2016.03.094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/12/2016] [Accepted: 03/29/2016] [Indexed: 01/14/2023]
Abstract
Based on our improved novel Salmonella vaccine delivery platform, we optimized the recombinant attenuated Salmonella typhimurium vaccine (RASV) χ12094 to deliver multiple Yersinia pestis antigens. These included LcrV196 (amino acids, 131-326), Psn encoded on pYA5383 and F1 encoded in the chromosome, their synthesis did not cause adverse effects on bacterial growth. Oral immunization with χ12094(pYA5383) simultaneously stimulated high antibody titers to LcrV, Psn and F1 in mice and presented complete protection against both subcutaneous (s.c.) and intranasal (i.n.) challenges with high lethal doses of Y. pestis CO92. Moreover, no deaths or other disease symptoms were observed in SCID mice orally immunized with χ12094(pYA5383) over a 60-day period. Therefore, the trivalent S. typhimurium-based live vaccine shows promise for a next-generation plague vaccine.
Collapse
Affiliation(s)
- Shilpa Sanapala
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Hannah Rahav
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Hetal Patel
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Wei Sun
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Roy Curtiss
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
26
|
Physiology of Yersinia pestis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 918:79-99. [DOI: 10.1007/978-94-024-0890-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Abstract
Three major plague pandemics caused by the gram-negative bacterium Yersinia pestis have killed nearly 200 million people in human history. Due to its extreme virulence and the ease of its transmission, Y. pestis has been used purposefully for biowarfare in the past. Currently, plague epidemics are still breaking out sporadically in most of parts of the world, including the United States. Approximately 2000 cases of plague are reported each year to the World Health Organization. However, the potential use of the bacteria in modern times as an agent of bioterrorism and the emergence of a Y. pestis strain resistant to eight antibiotics bring out severe public health concerns. Therefore, prophylactic vaccination against this disease holds the brightest prospect for its long-term prevention. Here, we summarize the progress of the current vaccine development for counteracting plague.
Collapse
Affiliation(s)
- Wei Sun
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, 110880, Gainesville, FL, 32611-0880, USA.
| |
Collapse
|
28
|
Sun W, Sanapala S, Rahav H, Curtiss R. Oral administration of a recombinant attenuated Yersinia pseudotuberculosis strain elicits protective immunity against plague. Vaccine 2015; 33:6727-35. [PMID: 26514425 DOI: 10.1016/j.vaccine.2015.10.074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/16/2015] [Accepted: 10/14/2015] [Indexed: 01/14/2023]
Abstract
A Yersinia pseudotuberculosis PB1+ (Yptb PB1+) mutant strain combined with chromosome insertion of the caf1R-caf1A-caf1M-caf1 operon and deletions of yopJ and yopK, χ10068 [pYV-ω2 (ΔyopJ315 ΔyopK108) ΔlacZ044::caf1R-caf1M-caf1A-caf1] was constructed. Results indicated that gene insertion and deletion did not affect the growth rate of χ10068 compared to wild-type Yptb cultured at 26 °C. In addition, the F1 antigen in χ10068 was synthesized and secreted on the surface of bacteria at 37 °C (mammalian body temperature), not at ambient culture temperature (26 °C). Immunization with χ10068 primed antibody responses and specific T-cell responses to F1 and YpL (Y. pestis whole cell lysate). Oral immunization with a single dose of χ10068 provided 70% protection against a subcutaneous (s.c.) challenge with ∼ 2.6 × 10(5) LD50 of Y. pestis KIM6+ (pCD1Ap) (KIM6+Ap) and 90% protection against an intranasal (i.n.) challenge with ∼ 500 LD50 of KIM6+Ap in mice. Our results suggest that χ10068 can be used as an effective precursor to make a safe vaccine to prevent plague in humans and to eliminate plague circulation among humans and animals.
Collapse
Affiliation(s)
- Wei Sun
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Shilpa Sanapala
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Hannah Rahav
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Roy Curtiss
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
29
|
Efficacy of primate humoral passive transfer in a murine model of pneumonic plague is mouse strain-dependent. J Immunol Res 2014; 2014:807564. [PMID: 25097863 PMCID: PMC4109106 DOI: 10.1155/2014/807564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/21/2014] [Accepted: 06/18/2014] [Indexed: 11/23/2022] Open
Abstract
New vaccines against biodefense-related and emerging pathogens are being prepared for licensure using the US Federal Drug Administration's “Animal Rule.” This allows licensure of drugs and vaccines using protection data generated in animal models. A new acellular plague vaccine composed of two separate recombinant proteins (rF1 and rV) has been developed and assessed for immunogenicity in humans. Using serum obtained from human volunteers immunised with various doses of this vaccine and from immunised cynomolgus macaques, we assessed the pharmacokinetic properties of human and cynomolgus macaque IgG in BALB/c and the NIH Swiss derived Hsd:NIHS mice, respectively. Using human and cynomolgus macaque serum with known ELISA antibody titres against both vaccine components, we have shown that passive immunisation of human and nonhuman primate serum provides a reproducible delay in median time to death in mice exposed to a lethal aerosol of plague. In addition, we have shown that Hsd:NIHS mice are a better model for humoral passive transfer studies than BALB/c mice.
Collapse
|
30
|
Sawa T, Katoh H, Yasumoto H. V-antigen homologs in pathogenic gram-negative bacteria. Microbiol Immunol 2014; 58:267-85. [DOI: 10.1111/1348-0421.12147] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/16/2014] [Accepted: 03/17/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Teiji Sawa
- Department of Anesthesiology; Kyoto Prefectural University of Medicine; Kajii-cho 465 Kamigyo Kyoto 602-8566 Japan
| | - Hideya Katoh
- Department of Anesthesiology; Kyoto Prefectural University of Medicine; Kajii-cho 465 Kamigyo Kyoto 602-8566 Japan
| | - Hiroaki Yasumoto
- Department of Anesthesiology; Kyoto Prefectural University of Medicine; Kajii-cho 465 Kamigyo Kyoto 602-8566 Japan
| |
Collapse
|
31
|
Phanse Y, Carrillo-Conde BR, Ramer-Tait AE, Broderick S, Kong CS, Rajan K, Flick R, Mandell RB, Narasimhan B, Wannemuehler MJ. A systems approach to designing next generation vaccines: combining α-galactose modified antigens with nanoparticle platforms. Sci Rep 2014; 4:3775. [PMID: 24441019 PMCID: PMC3895907 DOI: 10.1038/srep03775] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/20/2013] [Indexed: 12/21/2022] Open
Abstract
Innovative vaccine platforms are needed to develop effective countermeasures against emerging and re-emerging diseases. These platforms should direct antigen internalization by antigen presenting cells and promote immunogenic responses. This work describes an innovative systems approach combining two novel platforms, αGalactose (αGal)-modification of antigens and amphiphilic polyanhydride nanoparticles as vaccine delivery vehicles, to rationally design vaccine formulations. Regimens comprising soluble αGal-modified antigen and nanoparticle-encapsulated unmodified antigen induced a high titer, high avidity antibody response with broader epitope recognition of antigenic peptides than other regimen. Proliferation of antigen-specific CD4+ T cells was also enhanced compared to a traditional adjuvant. Combining the technology platforms and augmenting immune response studies with peptide arrays and informatics analysis provides a new paradigm for rational, systems-based design of next generation vaccine platforms against emerging and re-emerging pathogens.
Collapse
Affiliation(s)
- Yashdeep Phanse
- 1] Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011 [2]
| | - Brenda R Carrillo-Conde
- 1] Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011 [2]
| | - Amanda E Ramer-Tait
- 1] Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011 [2] Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Scott Broderick
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Chang Sun Kong
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Krishna Rajan
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Ramon Flick
- BioProtection Systems Corporation, a subsidiary of NewLink Genetics Corporation, Ames, IA 50010
| | - Robert B Mandell
- 1] BioProtection Systems Corporation, a subsidiary of NewLink Genetics Corporation, Ames, IA 50010 [2]
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011
| |
Collapse
|
32
|
|
33
|
Hickey AJ, Lin JS, Kummer LW, Szaba FM, Duso DK, Tighe M, Parent MA, Smiley ST. Intranasal prophylaxis with CpG oligodeoxynucleotide can protect against Yersinia pestis infection. Infect Immun 2013; 81:2123-32. [PMID: 23545300 PMCID: PMC3676034 DOI: 10.1128/iai.00316-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 03/18/2013] [Indexed: 01/03/2023] Open
Abstract
Immunomodulatory agents potentially represent a new class of broad-spectrum antimicrobials. Here, we demonstrate that prophylaxis with immunomodulatory cytosine-phosphate-guanidine (CpG) oligodeoxynucleotide (ODN), a toll-like receptor 9 (TLR9) agonist, confers protection against Yersinia pestis, the etiologic agent of plague. The data establish that intranasal administration of CpG ODN 1 day prior to lethal pulmonary exposure to Y. pestis strain KIM D27 significantly improves survival of C57BL/6 mice and reduces bacterial growth in hepatic tissue, despite paradoxically increasing bacterial growth in the lung. All of these CpG ODN-mediated impacts, including the increased pulmonary burden, are TLR9 dependent, as they are not observed in TLR9-deficient mice. The capacity of prophylactic intranasal CpG ODN to enhance survival does not require adaptive immunity, as it is evident in mice lacking B and/or T cells; however, the presence of T cells improves long-term survival. The prophylactic regimen also improves survival and reduces hepatic bacterial burden in mice challenged intraperitoneally with KIM D27, indicating that intranasal delivery of CpG ODN has systemic impacts. Indeed, intranasal prophylaxis with CpG ODN provides significant protection against subcutaneous challenge with Y. pestis strain CO92 even though it fails to protect mice from intranasal challenge with that fully virulent strain.
Collapse
|
34
|
|
35
|
Plague vaccines: current developments and future perspectives. Emerg Microbes Infect 2012; 1:e36. [PMID: 26038406 PMCID: PMC3630923 DOI: 10.1038/emi.2012.34] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 09/12/2012] [Accepted: 09/13/2012] [Indexed: 12/21/2022]
Abstract
Despite many decades of intensive studies of Yersinia pestis, the causative agent of plague, there is no safe and efficient vaccine against this devastating disease. A recently developed F1/V subunit vaccine candidate, which relies mainly on humoral immunity, showed promising results in animal studies; however, its efficacy in humans still has to be carefully evaluated. In addition, those developing next-generation plague vaccines need to pay particular attention to the importance of eliciting cell-mediated immunity. In this review, we analyzed the current progress in developing subunit, DNA and live carrier platforms of delivery by bacterial and viral vectors, as well as approaches for controlled attenuation of virulent strains of Y. pestis.
Collapse
|
36
|
Sun W, Curtiss R. Amino acid substitutions in LcrV at putative sites of interaction with Toll-like receptor 2 do not affect the virulence of Yersinia pestis. Microb Pathog 2012; 53:198-206. [PMID: 22841961 DOI: 10.1016/j.micpath.2012.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 06/28/2012] [Accepted: 07/03/2012] [Indexed: 11/28/2022]
Abstract
LcrV, a component of the type III secretion system (T3SS) translocon in Yersinia pestis, has been concerned in suppressing inflammation through Toll-like receptor 2 (TLR2) by inducing expression of the anti-inflammatory cytokine interleukin-10 (IL-10). Previous studies have reported that LcrV aa E33, E34, K42 and/or E204 and E205 were important for interactions with TLR2 in vitro. While, recently there have been conflicting reports doubting this interaction and its importance in vivo. To further investigate the role of these residues, we replaced the wild-type lcrV gene on the pCD1Ap virulence plasmid of Y. pestis with lcrV2345 gene, which encodes a mutant protein by substituting all five of the amino acid residues with glutamine. The characteristics of the wild-type LcrV and mutant LcrV2345 were evaluated in tissue culture and mice. When purified protein was incubated with HEK293 cells synthesizing human TLR2 with or without CD14, LcrV2345 induced higher levels of IL-8 than wild-type LcrV, indicating that the LcrV2345 was not impaired in its ability to interact with TLR2. LcrV2345 stimulated higher levels of tumor necrosis factor-alpha (TNF-α) production than LcrV in J774A.1 cells, while neither protein elicited significant levels of IL-10. We also found there was no statistically significant difference in virulence between strains with wild-type LcrV and with mutated LcrV2345 administered by either subcutaneous or intranasal route in mice. Additionally, there were no discernible differences in survival kinetics. Serum levels of cytokines, such as IL-10 and TNF-α, bacterial burden, and the extent of organ inflammation were also indistinguishable in both strains. Our data confirm that immunomodulation mediated by LcrV/TLR2 interactions does not play a significant role in the pathogenicity of Y. pestis.
Collapse
Affiliation(s)
- Wei Sun
- Center for Infectious Disease and Vaccinology, The Biodesign Institute, Arizona State University, P.O. Box 875401, 1001 S. McAllister Avenue, Tempe, AZ 85287-5401, USA.
| | | |
Collapse
|
37
|
Bozue J, Cote CK, Webster W, Bassett A, Tobery S, Little S, Swietnicki W. A Yersinia pestis YscN ATPase mutant functions as a live attenuated vaccine against bubonic plague in mice. FEMS Microbiol Lett 2012; 332:113-21. [PMID: 22537022 DOI: 10.1111/j.1574-6968.2012.02583.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 04/19/2012] [Accepted: 04/22/2012] [Indexed: 11/30/2022] Open
Abstract
Yersinia pestis is the causative agent responsible for bubonic and pneumonic plague. The bacterium uses the pLcr plasmid-encoded type III secretion system to deliver virulence factors into host cells. Delivery requires ATP hydrolysis by the YscN ATPase encoded by the yscN gene also on pLcr. A yscN mutant was constructed in the fully virulent CO92 strain containing a nonpolar, in-frame internal deletion within the gene. We demonstrate that CO92 with a yscN mutation was not able to secrete the LcrV protein (V-Antigen) and attenuated in a subcutaneous model of plague demonstrating that the YscN ATPase was essential for virulence. However, if the yscN mutant was complemented with a functional yscN gene in trans, virulence was restored. To evaluate the mutant as a live vaccine, Swiss-Webster mice were vaccinated twice with the ΔyscN mutant at varying doses and were protected against bubonic plague in a dose-dependent manner. Antibodies to F1 capsule but not to LcrV were detected in sera from the vaccinated mice. These preliminary results suggest a proof-of-concept for an attenuated, genetically engineered, live vaccine effective against bubonic plague.
Collapse
Affiliation(s)
- Joel Bozue
- Division of Bacteriology, US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Polymorphisms in the lcrV gene of Yersinia enterocolitica and their effect on plague protective immunity. Infect Immun 2012; 80:1572-82. [PMID: 22252870 DOI: 10.1128/iai.05637-11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current efforts to develop plague vaccines focus on LcrV, a polypeptide that resides at the tip of type III secretion needles. LcrV-specific antibodies block Yersinia pestis type III injection of Yop effectors into host immune cells, thereby enabling phagocytes to kill the invading pathogen. Earlier work reported that antibodies against Y. pestis LcrV cannot block type III injection by Yersinia enterocolitica strains and suggested that lcrV polymorphisms may provide for escape from LcrV-mediated plague immunity. We show here that polyclonal or monoclonal antibodies raised against Y. pestis KIM D27 LcrV (LcrV(D27)) bind LcrV from Y. enterocolitica O:9 strain W22703 (LcrV(W22703)) or O:8 strain WA-314 (LcrV(WA-314)) but are otherwise unable to block type III injection by Y. enterocolitica strains. Replacing the lcrV gene on the pCD1 virulence plasmid of Y. pestis KIM D27 with either lcrV(W22703) or lcrV(WA-314) does not affect the ability of plague bacteria to secrete proteins via the type III pathway, to inject Yops into macrophages, or to cause lethal plague infections in mice. LcrV(D27)-specific antibodies blocked type III injection by Y. pestis expressing lcrV(W22703) or lcrV(WA-314) and protected mice against intravenous lethal plague challenge with these strains. Thus, although antibodies raised against LcrV(D27) are unable to block the type III injection of Y. enterocolitica strains, expression of lcrV(W22703) or lcrV(WA-314) in Y. pestis did not allow these strains to escape LcrV-mediated plague protective immunity in the intravenous challenge model.
Collapse
|
39
|
Byvalov AA, Ovodov IS. [Immunobiological properties of Yersinia pestis antigens]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2011; 37:452-63. [PMID: 22096987 DOI: 10.1134/s1068162011040042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The present review contains information concerning immunobiological properties of plague microbe antigens. All of the identified antigens are evaluated in relation to pathogenicity of Yersinia pestis namely a resistance to phagocytosis, toxicity, adhesiveness etc. as well as persistence ability and adaptation to variable environment. In addition, the role of antigens in immunogenicity of living plague microbe for experimental animals is considered. The data concerning mechanisms of antigenic contribution to the development of adaptive immunity are presented.
Collapse
|
40
|
Advanced Development of the rF1V and rBV A/B Vaccines: Progress and Challenges. Adv Prev Med 2011; 2012:731604. [PMID: 22028978 PMCID: PMC3199075 DOI: 10.1155/2012/731604] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 07/20/2011] [Accepted: 07/21/2011] [Indexed: 01/25/2023] Open
Abstract
The development of vaccines for microorganisms and bacterial toxins with the potential to be used as biowarfare and bioterrorism agents is an important component of the US biodefense program. DVC is developing two vaccines, one against inhalational exposure to botulinum neurotoxins A1 and B1 and a second for Yersinia pestis, with the ultimate goal of licensure by the FDA under the Animal Rule. Progress has been made in all technical areas, including manufacturing, nonclinical, and clinical development and testing of the vaccines, and in assay development. The current status of development of these vaccines, and remaining challenges are described in this chapter.
Collapse
|
41
|
Embry A, Meng X, Cantwell A, Dube PH, Xiang Y. Enhancement of immune response to an antigen delivered by vaccinia virus by displaying the antigen on the surface of intracellular mature virion. Vaccine 2011; 29:5331-9. [PMID: 21664218 PMCID: PMC3139018 DOI: 10.1016/j.vaccine.2011.05.088] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 05/23/2011] [Accepted: 05/25/2011] [Indexed: 12/01/2022]
Abstract
Vaccinia virus (VACV) is the vaccine for smallpox and a widely used vaccine vector for infectious diseases and cancers. The majority of the antibodies elicited by live VACV vaccination respond to virion structural proteins, including many integral membrane proteins on the intracellular mature virion (MV). Here, we showed that antibody response to an exogenous antigen delivered by VACV was greatly enhanced by incorporating the antigen as an integral membrane protein of MV. We constructed recombinant VACV expressing a Yersinia pestis protective antigen, LcrV, unmodified or fused with either a signal peptide or with the transmembrane domain of VACV D8 protein (LcrV-TM). Electron microscopy showed that LcrV-TM was displayed on the surface of MV. Importantly, VACV expressing LcrV-TM elicited a significantly higher titer of anti-LcrV antibody in mice than viruses expressing other forms of LcrV. Only mice immunized with LcrV-TM-expressing VACV were protected from lethal Y. pestis and VACV WR challenges. Antigen engineering through fusion with D8 transmembrane domain may be broadly applicable for enhancing the immune response to antigens delivered by a VACV vector. The recombinant virus described here could also serve as the basis for developing a vaccine against both smallpox and plague.
Collapse
Affiliation(s)
- Addie Embry
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xiangzhi Meng
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Angelene Cantwell
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Peter H. Dube
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Yan Xiang
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| |
Collapse
|
42
|
Quenee LE, Ciletti NA, Elli D, Hermanas TM, Schneewind O. Prevention of pneumonic plague in mice, rats, guinea pigs and non-human primates with clinical grade rV10, rV10-2 or F1-V vaccines. Vaccine 2011; 29:6572-83. [PMID: 21763383 DOI: 10.1016/j.vaccine.2011.06.119] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 06/30/2011] [Accepted: 06/30/2011] [Indexed: 10/18/2022]
Abstract
Yersinia pestis causes plague, a disease with high mortality in humans that can be transmitted by fleabite or aerosol. A US Food and Drug Administration (FDA)-licensed plague vaccine is currently not available. Vaccine developers have focused on two subunits of Y. pestis: LcrV, a protein at the tip of type III secretion needles, and F1, the fraction 1 pilus antigen. F1-V, a hybrid generated via translational fusion of both antigens, is being developed for licensure as a plague vaccine. The rV10 vaccine is a non-toxigenic variant of LcrV lacking residues 271-300. Here we developed Current Good Manufacturing Practice (cGMP) protocols for rV10. Comparison of clinical grade rV10 with F1-V did not reveal significant differences in plague protection in mice, guinea pigs or cynomolgus macaques. We also developed cGMP protocols for rV10-2, a variant of rV10 with an altered affinity tag. Immunization with rV10-2 adsorbed to aluminum hydroxide elicited antibodies against LcrV and conferred pneumonic plague protection in mice, rats, guinea pigs, cynomolgus macaques and African Green monkeys. The data support further development of rV10-2 for FDA Investigational New Drug (IND) authorization review and clinical testing.
Collapse
Affiliation(s)
- Lauriane E Quenee
- Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
43
|
Sato H, Frank DW. Multi-Functional Characteristics of the Pseudomonas aeruginosa Type III Needle-Tip Protein, PcrV; Comparison to Orthologs in other Gram-negative Bacteria. Front Microbiol 2011; 2:142. [PMID: 21772833 PMCID: PMC3131520 DOI: 10.3389/fmicb.2011.00142] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Accepted: 06/15/2011] [Indexed: 01/02/2023] Open
Abstract
Pseudomonas aeruginosa possesses a type III secretion system (T3SS) to intoxicate host cells and evade innate immunity. This virulence-related machinery consists of a molecular syringe and needle assembled on the bacterial surface, which allows delivery of T3 effector proteins into infected cells. To accomplish a one-step effector translocation, a tip protein is required at the top end of the T3 needle structure. Strains lacking expression of the functional tip protein fail to intoxicate host cells. P. aeruginosa encodes a T3S that is highly homologous to the proteins encoded by Yersinia spp. The needle-tip proteins of Yersinia, LcrV, and P. aeruginosa, PcrV, share 37% identity and 65% similarity. Other known tip proteins are AcrV (Aeromonas), IpaD (Shigella), SipD (Salmonella), BipD (Burkholderia), EspA (EPEC, EHEC), Bsp22 (Bordetella), with additional proteins identified from various Gram-negative species, such as Vibrio and Bordetella. The tip proteins can serve as a protective antigen or may be critical for sensing host cells and evading innate immune responses. Recognition of the host microenvironment transcriptionally activates synthesis of T3SS components. The machinery appears to be mechanically controlled by the assemblage of specific junctions within the apparatus. These junctions include the tip and base of the T3 apparatus, the needle proteins and components within the bacterial cytoplasm. The tip proteins likely have chaperone functions for translocon proteins, allowing the proper assembly of translocation channels in the host membrane and completing vectorial delivery of effector proteins into the host cytoplasm. Multi-functional features of the needle-tip proteins appear to be intricately controlled. In this review, we highlight the functional aspects and complex controls of T3 needle-tip proteins with particular emphasis on PcrV and LcrV.
Collapse
Affiliation(s)
- Hiromi Sato
- Center for Infectious Disease Research, Medical College of Wisconsin Milwaukee, WI, USA
| | | |
Collapse
|
44
|
Abstract
Bioterrorism is the deliberate release of biological toxins, pathogenic viruses, bacteria, parasites, or other infectious agents into the public sphere with the objective of causing panic, illness, and/or death on a local, regional, or possibly national scale. The list of potential biological agents compiled by the Centers for Disease Control and Prevention is long and diverse. However, a trait common to virtually all the potential bioterrorism agents is the fact that they are likely to be disseminated by either aerosol or in food/water supplies with the intention of targeting the mucosal surfaces of the respiratory or gastrointestinal tracts, respectively. In some instances, inhalation or ingestion would mimic the natural route by which humans are exposed to these agents. In other instances, (e.g., the inhalation of a toxin is normally associated with food borne illness), it would represent an unnatural route of exposure. For most potential bioterrorism agents, the respiratory or gastrointestinal mucosa may simply serve as a route of entry by which they gain access to the systemic compartment where intoxication/replication occurs. For others, however, the respiratory or gastrointestinal mucosa is the primary tissue associated with pathogenesis, and therefore, the tissue for which countermeasures must be developed.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- , Louisiana Gene Therapy Research Consorti, LSU Health Sciences Center, Poydras Street 1615, New Orleans, 70112 Louisiana USA
| | | | | |
Collapse
|
45
|
Modified needle-tip PcrV proteins reveal distinct phenotypes relevant to the control of type III secretion and intoxication by Pseudomonas aeruginosa. PLoS One 2011; 6:e18356. [PMID: 21479247 PMCID: PMC3066235 DOI: 10.1371/journal.pone.0018356] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 02/27/2011] [Indexed: 12/14/2022] Open
Abstract
The type III secretion system (T3SS) is employed to deliver effector proteins to the cytosol of eukaryotic hosts by multiple species of Gram-negative bacteria, including Pseudomonas aeruginosa. Translocation of effectors is dependent on the proteins encoded by the pcrGVHpopBD operon. These proteins form a T3S translocator complex, composed of a needle-tip complex (PcrV), translocons (PopB and PopD), and chaperones (PcrG and PcrH). PcrV mediates the folding and insertion of PopB/PopD in host plasmic membranes, where assembled translocons form a translocation channel. Assembly of this complex and delivery of effectors through this machinery is tightly controlled by PcrV, yet the multifunctional aspects of this molecule have not been defined. In addition, PcrV is a protective antigen for P. aeruginosa infection as is the ortholog, LcrV, for Yersinia. We constructed PcrV derivatives containing in-frame linker insertions and site-specific mutations. The expression of these derivatives was regulated by a T3S-specific promoter in a pcrV-null mutant of PA103. Nine derivatives disrupted the regulation of effector secretion and constitutively released an effector protein into growth medium. Three of these regulatory mutants, in which the linker was inserted in the N-terminal globular domain, were competent for the translocation of a cytotoxin, ExoU, into eukaryotic host cells. We also isolated strains expressing a delayed-toxicity phenotype, which secrete translocators slowly despite the normal level of effector secretion. Most of the cytotoxic translocation-competent strains retained the protective epitope of PcrV derivatives, and Mab166 was able to protect erythrocytes during infection with these strains. The use of defined PcrV derivatives possessing distinct phenotypes may lead to a better understanding of the functional aspects of T3 needle-tip proteins and the development of therapeutic agents or vaccines targeting T3SS-mediated intoxication.
Collapse
|
46
|
Gendrin C, Sarrazin S, Bonnaffé D, Jault JM, Lortat-Jacob H, Dessen A. Hijacking of the pleiotropic cytokine interferon-γ by the type III secretion system of Yersinia pestis. PLoS One 2010; 5:e15242. [PMID: 21179438 PMCID: PMC3001473 DOI: 10.1371/journal.pone.0015242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 11/02/2010] [Indexed: 11/19/2022] Open
Abstract
Yersinia pestis, the causative agent of bubonic plague, employs its type III secretion system to inject toxins into target cells, a crucial step in infection establishment. LcrV is an essential component of the T3SS of Yersinia spp, and is able to associate at the tip of the secretion needle and take part in the translocation of anti-host effector proteins into the eukaryotic cell cytoplasm. Upon cell contact, LcrV is also released into the surrounding medium where it has been shown to block the normal inflammatory response, although details of this mechanism have remained elusive. In this work, we reveal a key aspect of the immunomodulatory function of LcrV by showing that it interacts directly and with nanomolar affinity with the inflammatory cytokine IFNγ. In addition, we generate specific IFNγ mutants that show decreased interaction capabilities towards LcrV, enabling us to map the interaction region to two basic C-terminal clusters of IFNγ. Lastly, we show that the LcrV-IFNγ interaction can be disrupted by a number of inhibitors, some of which display nanomolar affinity. This study thus not only identifies novel potential inhibitors that could be developed for the control of Yersinia-induced infection, but also highlights the diversity of the strategies used by Y. pestis to evade the immune system, with the hijacking of pleiotropic cytokines being a long-range mechanism that potentially plays a key role in the severity of plague.
Collapse
Affiliation(s)
- Claire Gendrin
- Institut de Biologie Structurale, UMR 5075 (Comissariat à l'Enérgie Atomique/Centre National de la Recherche Scientifique/Université Grenoble I), Grenoble, France
| | - Stéphane Sarrazin
- Institut de Biologie Structurale, UMR 5075 (Comissariat à l'Enérgie Atomique/Centre National de la Recherche Scientifique/Université Grenoble I), Grenoble, France
| | - David Bonnaffé
- Laboratoire de Chimie Organique Multifonctionnelle, Institut de Chimie Moléculaire et des Matériaux d'Orsay, UMR 8182, Université Paris-Sud 11, Orsay, France
| | - Jean-Michel Jault
- Institut de Biologie Structurale, UMR 5075 (Comissariat à l'Enérgie Atomique/Centre National de la Recherche Scientifique/Université Grenoble I), Grenoble, France
| | - Hugues Lortat-Jacob
- Institut de Biologie Structurale, UMR 5075 (Comissariat à l'Enérgie Atomique/Centre National de la Recherche Scientifique/Université Grenoble I), Grenoble, France
| | - Andréa Dessen
- Institut de Biologie Structurale, UMR 5075 (Comissariat à l'Enérgie Atomique/Centre National de la Recherche Scientifique/Université Grenoble I), Grenoble, France
- * E-mail:
| |
Collapse
|
47
|
Fellows P, Adamovicz J, Hartings J, Sherwood R, Mega W, Brasel T, Barr E, Holland L, Lin W, Rom A, Blackwelder W, Price J, Morris S, Snow D, Hart MK. Protection in mice passively immunized with serum from cynomolgus macaques and humans vaccinated with recombinant plague vaccine (rF1V). Vaccine 2010; 28:7748-56. [DOI: 10.1016/j.vaccine.2010.09.062] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 09/16/2010] [Accepted: 09/19/2010] [Indexed: 01/14/2023]
|
48
|
Evaluation of Psn, HmuR and a modified LcrV protein delivered to mice by live attenuated Salmonella as a vaccine against bubonic and pneumonic Yersinia pestis challenge. Vaccine 2010; 29:274-82. [PMID: 20979987 DOI: 10.1016/j.vaccine.2010.10.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 10/13/2010] [Indexed: 01/09/2023]
Abstract
We evaluated the ability of Yersinia pestis antigens HmuR, Psn and modified forms of LcrV delivered by live attenuated Salmonella strains to stimulate a protective immune response against subcutaneous or intranasal challenge with Y. pestis CO92. LcrV196 is a previously described truncated protein that includes aa 131-326 of LcrV and LcrV5214 has been modified to replace five key amino acids required for interaction with the TLR2 receptor. Psn is the outer membrane receptor for the siderophore, yersiniabactin, and the bacteriocin, pesticin. Mice immunized with Salmonella synthesizing Psn, LcrV196 or LcrV5214 developed serum IgG responses to the respective Yersinia antigen and were protected against pneumonic challenge with Y. pestis. Immunization with Salmonella synthesizing Psn or LcrV196 was sufficient to afford nearly full protection against bubonic challenge, while immunization with the strain synthesizing LcrV5214 was not protective. Immunization with Salmonella synthesizing HmuR, an outer membrane protein involved in heme acquisition in Y. pestis, was poorly immunogenic and did not elicit a protective response against either challenge route. These findings indicate that both Psn and LcrV196 delivered by Salmonella provide protection against both bubonic and pneumonic plague.
Collapse
|
49
|
Bhattacharya D, Mecsas J, Hu LT. Development of a vaccinia virus based reservoir-targeted vaccine against Yersinia pestis. Vaccine 2010; 28:7683-9. [PMID: 20875494 DOI: 10.1016/j.vaccine.2010.09.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 09/08/2010] [Accepted: 09/10/2010] [Indexed: 01/14/2023]
Abstract
Yersinia pestis, the causative organism of plague, is a zoonotic organism with a worldwide distribution. Although the last plague epidemic occurred in early 1900s, human cases continue to occur due to contact with infected wild animals. In this study, we have developed a reservoir-targeted vaccine against Y. pestis, to interrupt transmission of disease in wild animals as a potential strategy for decreasing human disease. A vaccinia virus delivery system was used to express the F1 capsular protein and the LcrV type III secretion component of Y. pestis as a fusion protein. Here we show that a single dose of this vaccine administered orally, generates a dose-dependent antibody response in mice. Antibody titers peak by 3 weeks after administration and remain elevated for a minimum of 45 weeks. Vaccination provided up to 100% protection against challenge with Y. pestis administered by intranasal challenge at 10 times the lethal dose with protection lasting a minimum of 45 weeks. An orally available, vaccinia virus expressed vaccine against Y. pestis may be a suitable vaccine for a reservoir targeted strategy for the prevention of enzootic plague.
Collapse
Affiliation(s)
- Debaditya Bhattacharya
- Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | | | | |
Collapse
|
50
|
Fine-tuning synthesis of Yersinia pestis LcrV from runaway-like replication balanced-lethal plasmid in a Salmonella enterica serovar typhimurium vaccine induces protection against a lethal Y. pestis challenge in mice. Infect Immun 2010; 78:2529-43. [PMID: 20308296 DOI: 10.1128/iai.00005-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
A balanced-lethal plasmid expression system that switches from low-copy-number to runaway-like high-copy-number replication (pYA4534) was constructed for the regulated delayed in vivo synthesis of heterologous antigens by vaccine strains. This is an antibiotic resistance-free maintenance system containing the asdA gene (essential for peptidoglycan synthesis) as a selectable marker to complement the lethal chromosomal DeltaasdA allele in live recombinant attenuated Salmonella vaccines (RASVs) such as Salmonella enterica serovar Typhimurium strain chi9447. pYA4534 harbors two origins of replication, pSC101 and pUC (low and high copy numbers, respectively). The pUC replication origin is controlled by a genetic switch formed by the operator/promoter of the P22 cro gene (O/P(cro)) (P(R)), which is negatively regulated by an arabinose-inducible P22 c2 gene located on both the plasmid and the chromosome (araC P(BAD) c2). The absence of arabinose, which is unavailable in vivo, triggers replication to a high-copy-number plasmid state. To validate these vector attributes, the Yersinia pestis virulence antigen LcrV was used to develop a vaccine against plague. An lcrV sequence encoding amino acids 131 to 326 (LcrV196) was optimized for expression in Salmonella, flanked with nucleotide sequences encoding the signal peptide (SS) and the carboxy-terminal domain (CT) of beta-lactamase, and cloned into pYA4534 under the control of the P(trc) promoter to generate plasmid pYA4535. Our results indicate that the live Salmonella vaccine strain chi9447 harboring pYA4535 efficiently stimulated a mixed Th1/Th2 immune response that protected mice against lethal challenge with Y. pestis strain CO92 introduced through either the intranasal or subcutaneous route.
Collapse
|