1
|
Velappan N, Biryukov SS, Rill NO, Klimko CP, Rosario-Acevedo R, Shoe JL, Hunter M, Dankmeyer JL, Fetterer DP, Bedinger D, Phipps ME, Watt AJ, Abergel RJ, Dichosa A, Kozimor SA, Cote CK, Lillo AM. Characterization of two affinity matured Anti-Yersinia pestis F1 human antibodies with medical countermeasure potential. PLoS One 2024; 19:e0305034. [PMID: 38954719 PMCID: PMC11218954 DOI: 10.1371/journal.pone.0305034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 05/23/2024] [Indexed: 07/04/2024] Open
Abstract
Yersinia pestis, the causative agent of plague and a biological threat agent, presents an urgent need for novel medical countermeasures due to documented cases of naturally acquired antibiotic resistance and potential person-to-person spread during a pneumonic infection. Immunotherapy has been proposed as a way to circumvent current and future antibiotic resistance. Here, we describe the development and characterization of two affinity matured human antibodies (αF1Ig AM2 and αF1Ig AM8) that promote survival of mice after exposure to aerosolized Y. pestis. We share details of the error prone PCR and yeast display technology-based affinity maturation process that we used. The resultant matured antibodies have nanomolar affinity for Y. pestis F1 antigen, are produced in high yield, and are resilient to 37°C stress for up to 6 months. Importantly, in vitro assays using a murine macrophage cell line demonstrated that αF1Ig AM2 and αF1Ig AM8 are opsonic. Even more importantly, in vivo studies using pneumonic plague mouse models showed that 100% of the mice receiving 500 μg of IgGs αF1Ig AM2 and αF1Ig AM8 survived lethal challenge with aerosolized Y. pestis CO92. Combined, these results provide evidence of the quality and robustness of αF1Ig AM2 and αF1Ig AM8 and support their development as potential medical countermeasures against plague.
Collapse
Affiliation(s)
- Nileena Velappan
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Sergei S. Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Nathaniel O. Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Raysa Rosario-Acevedo
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Jennifer L. Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - David P. Fetterer
- Biostatisitics Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | | | - Mary E. Phipps
- Los Alamos National Laboratory, Center Alamos for Integrated Nanotechnologies, Los Alamos, NM, United States of America
| | - Austin J. Watt
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Rebecca J. Abergel
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States of America
- Department of Nuclear Engineering, University of California, Berkeley, CA, United States of America
| | - Armand Dichosa
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Stosh A. Kozimor
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Antonietta M. Lillo
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| |
Collapse
|
2
|
Davies ML, Biryukov SS, Rill NO, Klimko CP, Hunter M, Dankmeyer JL, Miller JA, Shoe JL, Mlynek KD, Talyansky Y, Toothman RG, Qiu J, Bozue JA, Cote CK. Sex differences in immune protection in mice conferred by heterologous vaccines for pneumonic plague. Front Immunol 2024; 15:1397579. [PMID: 38835755 PMCID: PMC11148226 DOI: 10.3389/fimmu.2024.1397579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/25/2024] [Indexed: 06/06/2024] Open
Abstract
Background Yersinia pestis is the etiological agent of plague, which can manifest as bubonic, septicemic, and/or pneumonic disease. Plague is a severe and rapidly progressing illness that can only be successfully treated with antibiotics initiated early after infection. There are no FDA-approved vaccines for plague, and some vaccine candidates may be less effective against pneumonic plague than bubonic plague. Y. pestis is not known to impact males and females differently in mechanisms of pathogenesis or severity of infection. However, one previous study reported sex-biased vaccine effectiveness after intranasal Y. pestis challenge. As part of developing a safe and effective vaccine, it is essential that potential sex differences are characterized. Methods In this study we evaluated novel vaccines in male and female BALB/c mice using a heterologous prime-boost approach and monitored survival, bacterial load in organs, and immunological correlates. Our vaccine strategy consisted of two subcutaneous immunizations, followed by challenge with aerosolized virulent nonencapsulated Y. pestis. Mice were immunized with a combination of live Y. pestis pgm- pPst-Δcaf1, live Y. pestis pgm- pPst-Δcaf1/ΔyopD, or recombinant F1-V (rF1-V) combined with adjuvants. Results The most effective vaccine regimen was initial priming with rF1-V, followed by boost with either of the live attenuated strains. However, this and other strategies were more protective in female mice. Males had higher bacterial burden and differing patterns of cytokine expression and serum antibody titers. Male mice did not demonstrate synergy between vaccination and antibiotic treatment as repeatedly observed in female mice. Conclusions This study provides new knowledge about heterologous vaccine strategies, sex differences in plague-vaccine efficacy, and the immunological factors that differ between male and female mice.
Collapse
Affiliation(s)
- Michael L Davies
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Sergei S Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Nathaniel O Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jennifer L Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jeremy A Miller
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Kevin D Mlynek
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Yuli Talyansky
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Ronald G Toothman
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Ju Qiu
- Regulated Research Administration: Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Joel A Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| |
Collapse
|
3
|
Guo X, Xin Y, Tong Z, Cao S, Zhang Y, Wu G, Chen H, Wang T, Song Y, Zhang Q, Yang R, Du Z. A novel sORF gene mutant strain of Yersinia pestis vaccine EV76 offers enhanced safety and improved protection against plague. PLoS Pathog 2024; 20:e1012129. [PMID: 38547321 PMCID: PMC11020802 DOI: 10.1371/journal.ppat.1012129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/16/2024] [Accepted: 03/15/2024] [Indexed: 04/18/2024] Open
Abstract
We recently identified two virulence-associated small open reading frames (sORF) of Yersinia pestis, named yp1 and yp2, and null mutants of each individual genes were highly attenuated in virulence. Plague vaccine strain EV76 is known for strong reactogenicity, making it not suitable for use in humans. To improve the immune safety of EV76, three mutant strains of EV76, Δyp1, Δyp2, and Δyp1&yp2 were constructed and their virulence attenuation, immunogenicity, and protective efficacy in mice were evaluated. All mutant strains were attenuated by the subcutaneous (s.c.) route and exhibited more rapid clearance in tissues than the parental strain EV76. Under iron overload conditions, only the mice infected with EV76Δyp1 survived, accompanied by less draining lymph nodes damage than those infected by EV76. Analysis of cytokines secreted by splenocytes of immunized mice found that EV76Δyp2 induced higher secretion of multiple cytokines including TNF-α, IL-2, and IL-12p70 than EV76. On day 42, EV76Δyp2 or EV76Δyp1&yp2 immunized mice exhibited similar protective efficacy as EV76 when exposed to Y. pestis 201, both via s.c. or intranasal (i.n.) routes of administration. Moreover, when exposed to 200-400 LD50 Y. pestis strain 201Δcaf1 (non-encapsulated Y. pestis), EV76Δyp2 or EV76Δyp1&yp2 are able to afford about 50% protection to i.n. challenges, significantly better than the protection afforded by EV76. On 120 day, mice immunized with EV76Δyp2 or EV76Δyp1&yp2 cleared the i.n. challenge of Y. pestis 201-lux as quickly as those immunized with EV76, demonstrating 90-100% protection. Our results demonstrated that deletion of the yp2 gene is an effective strategy to attenuate virulence of Y. pestis EV76 while improving immunogenicity. Furthermore, EV76Δyp2 is a promising candidate for conferring protection against the pneumonic and bubonic forms of plague.
Collapse
Affiliation(s)
- Xiao Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Youquan Xin
- Key Laboratory for Plague Prevention and Control of Qinghai Province, Qinghai Institute for Endemic Disease Prevention and Control, Xining, China
| | - Zehui Tong
- School of Basic Medical Sciences, Anhui Medical University Hefei, China
| | - Shiyang Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuan Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Gengshan Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Hongyan Chen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Tong Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yajun Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Qingwen Zhang
- Key Laboratory for Plague Prevention and Control of Qinghai Province, Qinghai Institute for Endemic Disease Prevention and Control, Xining, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zongmin Du
- School of Basic Medical Sciences, Anhui Medical University Hefei, China
- Key Laboratory for Plague Prevention and Control of Qinghai Province, Qinghai Institute for Endemic Disease Prevention and Control, Xining, China
| |
Collapse
|
4
|
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
|
5
|
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: 2.7] [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
|
6
|
Ii AN, Lin SC, Lepene B, Zhou W, Kehn-Hall K, van Hoek ML. Use of magnetic nanotrap particles in capturing Yersinia pestis virulence factors, nucleic acids and bacteria. J Nanobiotechnology 2021; 19:186. [PMID: 34154629 PMCID: PMC8215484 DOI: 10.1186/s12951-021-00859-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022] Open
Abstract
Background Many pathogens, including Yersinia pestis, cannot be consistently and reliably cultured from blood. New approaches are needed to facilitate the detection of proteins, nucleic acid and microorganisms in whole blood samples to improve downstream assay performance. Detection of biomarkers in whole blood is difficult due to the presence of host proteins that obscure standard detection mechanisms. Nanotrap® particles are micron-sized hydrogel structures containing a dye molecule as the affinity bait and used to detect host biomarkers, viral nucleic acids and proteins as well as some bacterial markers. Nanotraps have been shown to bind and enrich a wide variety of biomarkers and viruses in clinically relevant matrices such as urine and plasma. Our objective was to characterize the binding ability of Nanotrap particle type CN3080 to Y. pestis bacteria, bacterial proteins and nucleic acids from whole human blood in order to potentially improve detection and diagnosis. Results CN3080 Nanotraps bind tightly to Yersinia bacteria, even after washing, and we were able to visualize the co-localized Nanotraps and bacteria by electron microscopy. These magnetic hydrogel Nanotraps were able to bind Yersinia DNA, supporting the utility of Nanotraps for enhancing nucleic acid-based detection methods. Nanotraps were capable of increasing Y. pestis nucleic acid yield by fourfold from whole human blood compared to standard nucleic acid extraction. Interestingly, we found CN3080 Nanotraps to have a high affinity for multiple components of the Yersinia type III secretion system (T3SS), including chaperone proteins, Yop effector proteins and virulence factor protein LcrV (V). Using Nanotraps as a rapid upstream sample-prep tool, we were able to detect LcrV in human blood by western blotting with minimal blood interference in contrast to direct western blotting of blood samples in which LcrV was obscured. We were able to computationally model the interaction of LcrV with the CN3080 Nanotrap dye and found that it had a low delta-G, suggesting high affinity. Importantly, Nanotraps were also able to enhance detection of secreted Yersinia proteins by mass spectrometry. Conclusion Upstream use of magnetic CN3080 Nanotrap particles may improve the downstream workflow though binding and enrichment of biomarkers and speed of processing. Utilization of Nanotrap particles can improve detection of Yersinia pestis proteins and nucleic acid from whole human blood and contribute to downstream assays and diagnostics including molecular methods such as sequencing and PCR and protein-based methods. Graphic Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00859-8.
Collapse
Affiliation(s)
- Alexandra N Ii
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Shih-Chao Lin
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA.,College of Life Sciences, National Taiwan Ocean University, 2 Pei-Ning Rd, Keelung, 202301, Taiwan
| | - Benjamin Lepene
- Ceres Nanosciences, 9460 Innovation Drive, Manassas, VA, 20110, USA
| | - Weidong Zhou
- Center for Applied Proteomics and Personalized Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Kylene Kehn-Hall
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA.,Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24060, USA
| | - Monique L van Hoek
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA.
| |
Collapse
|
7
|
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: 7] [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
|
8
|
Trevino SR, Dankmeyer JL, Fetterer DP, Klimko CP, Raymond JLW, Moreau AM, Soffler C, Waag DM, Worsham PL, Amemiya K, Ruiz SI, Cote CK, Krakauer T. Comparative virulence of three different strains of Burkholderia pseudomallei in an aerosol non-human primate model. PLoS Negl Trop Dis 2021; 15:e0009125. [PMID: 33571211 PMCID: PMC7904162 DOI: 10.1371/journal.pntd.0009125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 02/24/2021] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is a major cause of sepsis and mortality in endemic regions of Southeast Asia and Northern Australia. B. pseudomallei is a potential bioterrorism agent due to its high infectivity, especially via inhalation, and its inherent resistance to antimicrobials. There is currently no vaccine for melioidosis and antibiotic treatment can fail due to innate drug resistance, delayed diagnosis and treatment, or insufficient duration of treatment. A well-characterized animal model that mimics human melioidosis is needed for the development of new medical countermeasures. This study first characterized the disease progression of melioidosis in the African green monkey (AGM) and rhesus macaque (RM) for non-human primate model down-selection. All AGMs developed acute lethal disease similar to that described in human acute infection following exposure to aerosolized B. pseudomallei strain HBPUB10134a. Only 20% of RMs succumbed to acute disease. Disease progression, immune response and pathology of two other strains of B. pseudomallei, K96243 and MSHR5855, were also compared using AGMs. These three B. pseudomallei strains represent a highly virulent strain from Thailand (HBPUB101034a), a highly virulent strains from Australia (MSHR5855), and a commonly used laboratory strains originating from Thailand (K96243). Animals were observed for clinical signs of infection and blood samples were analyzed for cytokine responses, blood chemistry and leukocyte changes in order to characterize bacterial infection. AGMs experienced fever after exposure to aerosolized B. pseudomallei at the onset of acute disease. Inflammation, abscesses and/or pyogranulomas were observed in lung with all three strains of B. pseudomallei. Inflammation, abscesses and/or pyogranulomas were observed in lymph nodes, spleen, liver and/or kidney with B. pseudomallei, HBPUB10134a and K96243. Additionally, the Australian strain MSHR5855 induced brain lesions in one AGM similar to clinical cases of melioidosis seen in Australia. Elevated serum levels of IL-1β, IL-1 receptor antagonist, IL-6, MCP-1, G-CSF, HGF, IFNγ, MIG, I-TAC, and MIP-1β at terminal end points can be significantly correlated with non-survivors with B. pseudomallei infection in AGM. The AGM model represents an acute model of B. pseudomallei infection for all three strains from two geographical locations and will be useful for efficacy testing of vaccines and therapeutics against melioidosis. In summary, a dysregulated immune response leading to excessive persistent inflammation and inflammatory cell death is the key driver of acute melioidosis. Early intervention in these pathways will be necessary to counter B. pseudomallei and mitigate the pathological consequences of melioidosis. Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is endemic in tropical regions globally and is an emerging threat in non-tropical areas worldwide. Its mortality rate is high in endemic areas due to its high infectivity, antimicrobial resistance, lack of available vaccines and limited treatment options. Animal model development and pathogenicity studies of various isolates are critical for the development of countermeasures against this pathogen. In this study, we compared the virulence of three different isolates of B. pseudomallei from two geographical locations in an aerosol non-human primate model. We found that early elevations of both pro-inflammatory and anti-inflammatory mediators, as well as the persistence of these mediators in the terminal phase of bacterial infection correlate with mortality. Histopathological analysis showed that the severity of lesions in various organs also correlates with the virulence of the B. pseudomallei strains, HBPUB10134a, MSHR5855 and K96243. Thus, a dysregulated immune response leading to excessive IL-1β and IL-6 at terminal end points and necrosis are key drivers of acute melioidosis. Development of drugs targeting these host response processes will be necessary to counter B. pseudomallei and mitigate the pathological consequences of melioidosis. This non-human primate model will facilitate the screening of vaccines and novel therapeutics.
Collapse
Affiliation(s)
- Sylvia R. Trevino
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - David P. Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Jo Lynne W. Raymond
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Alicia M. Moreau
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Carl Soffler
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - David M. Waag
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Patricia L. Worsham
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Kei Amemiya
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Sara I. Ruiz
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
- * E-mail: (CKC); (TK)
| | - Teresa Krakauer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
- * E-mail: (CKC); (TK)
| |
Collapse
|
9
|
Feng J, Deng Y, Fu M, Hu X, Luo W, Lu Z, Dai L, Yang H, Zhao X, Du Z, Wen B, Jiang L, Zhou D, Jiao J, Xiong X. Construction of a Live-Attenuated Vaccine Strain of Yersinia pestis EV76-B-SHUΔ pla and Evaluation of Its Protection Efficacy in a Mouse Model by Aerosolized Intratracheal Inoculation. Front Cell Infect Microbiol 2020; 10:473. [PMID: 33014895 PMCID: PMC7509399 DOI: 10.3389/fcimb.2020.00473] [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: 02/24/2020] [Accepted: 07/31/2020] [Indexed: 12/15/2022] Open
Abstract
Plague, which is caused by Yersinia pestis, is one of the most dangerous infectious diseases. No FDA-approved vaccine against plague is available for human use at present. To improve the immune safety of Y. pestis EV76 based live attenuated vaccine and to explore the feasibility of aerosolized intratracheal inoculation (i.t.) route for vaccine delivery, a plasminogen activator protease (pla) gene deletion mutant of the attenuated Y. pestis strain EV76-B-SHU was constructed, and its residual virulence and protective efficacy were evaluated in a mouse model via aerosolized intratracheal inoculation (i.t.) or via subcutaneous injection (s.c.). The residual virulence of EV76-B-SHUΔpla was significantly reduced compared to that of the parental strain EV76-B-SHU following i.t. and s.c. infection. The EV76-B-SHUΔpla induced higher levels of mucosal antibody sIgA in the bronchoalveolar lavage fluid of mice immunized by i.t. but not by s.c.. Moreover, after lethal challenge with Y. pestis biovar Microtus strain 201 (avirulent in humans), the protective efficacy and bacterial clearance ability of the EV76-B-SHUΔpla-i.t. group were comparable to those of the EV76-B-SHUΔpla-s.c. and EV76-B-SHU immunized groups. Thus, the EV76-B-SHUΔpla represents an excellent live-attenuated vaccine candidate against pneumonic plague and aerosolized i.t. represents a promising immunization route in mouse model.
Collapse
Affiliation(s)
- Junxia Feng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China.,Capital Institute of Pediatrics, Beijing, China
| | - Yingying Deng
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mengjiao Fu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xueyuan Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Wenbo Luo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zhiyu Lu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | | | - Huiying Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaodong Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zongmin Du
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Bohai Wen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lingxiao Jiang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jun Jiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaolu Xiong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
10
|
Cote CK, Blanco II, Hunter M, Shoe JL, Klimko CP, Panchal RG, Welkos SL. Combinations of early generation antibiotics and antimicrobial peptides are effective against a broad spectrum of bacterial biothreat agents. Microb Pathog 2020; 142:104050. [PMID: 32050093 DOI: 10.1016/j.micpath.2020.104050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 10/25/2022]
Abstract
The misuse of infectious disease pathogens as agents of deliberate attack on civilians and military personnel is a serious national security concern, which is exacerbated by the emergence of natural or genetically engineered multidrug resistant strains. In this study, the therapeutic potential of combinations of an antibiotic and a broad-spectrum antimicrobial peptide (AMP) was evaluated against five bacterial biothreats, the etiologic agents of glanders (Burkholderia mallei), melioidosis (Burkholderia pseudomallei), plague (Yersinia pestis), tularemia (Francisella tularensis), and anthrax (Bacillus anthracis). The therapeutics included licensed early generation antibiotics which are now rarely used. Three antibiotics and one 24- amino acid AMP were selected based on MIC assay data. Combinations of the AMP and tigecycline, minocycline, or novobiocin were screened for synergistic activity by checkerboard MIC assay. The combinations each enhanced the susceptibility of several strains. The tetracycline-peptide combinations increased the sensitivities of Y. pestis, F. tularensis, B. anthracis and B. pseudomallei, and the novobiocin-AMP combination augmented the sensitivity of all five. In time-kill assays, down-selected combinations of the peptide and minocycline or tigecycline enhanced killing of B. anthracis, Y. pestis, F. tularensis, and Burkholderia mallei but not B. pseudomallei. The novobiocin-AMP pair significantly reduced viability of all strains except B. mallei, which was very sensitive to the antibiotic alone. The results suggested that antibiotic-AMP combinations are useful tools for combating diverse pathogens. Future studies employing cell culture and animal models will utilize virulent strains of the agents to investigate the in vivo availability, host cytotoxicity, and protective efficacy of these therapeutics.
Collapse
Affiliation(s)
- Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702-5011, USA.
| | - Irma I Blanco
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702-5011, USA
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702-5011, USA
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702-5011, USA
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702-5011, USA
| | | | - Susan L Welkos
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702-5011, USA.
| |
Collapse
|
11
|
Culbreth MJ, Biryukov SS, Shoe JL, Dankmeyer JL, Hunter M, Klimko CP, Rosario-Acevedo R, Fetterer DP, Moreau AM, Welkos SL, Cote CK. The Use of Analgesics during Vaccination with a Live Attenuated Yersinia pestis Vaccine Alters the Resulting Immune Response in Mice. Vaccines (Basel) 2019; 7:vaccines7040205. [PMID: 31816945 PMCID: PMC6963655 DOI: 10.3390/vaccines7040205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
The administration of antipyretic analgesics prior to, in conjunction with, or due to sequelae associated with vaccination is a common yet somewhat controversial practice. In the context of human vaccination, it is unclear if even short-term analgesic regimens can significantly alter the resulting immune response, as literature exists to support several scenarios including substantial immune interference. In this report, we used a live attenuated Yersinia pestis vaccine to examine the impact of analgesic administration on the immune response elicited by a single dose of a live bacterial vaccine in mice. Mice were assessed by evaluating natural and provoked behavior, as well as food and water consumption. The resulting immune responses were assessed by determining antibody titers against multiple antigens and assaying cellular responses in stimulated splenocytes collected from vaccinated animals. We observed no substantial benefit to the mice associated with the analgesic administration. Splenocytes from both C57BL/6 and BALB/c vaccinated mice receiving acetaminophen have a significantly reduced interferon-gamma (IFN-γ) recall response. Additionally, there is a significantly lower immunoglobulin (Ig)G2a/IgG1 ratio in vaccinated BALB/c mice treated with either acetaminophen or meloxicam and a significantly lower IgG2c/IgG1 ratio in vaccinated C57BL/6 mice treated with acetaminophen. Taken together, our data indicate that the use of analgesics, while possibly ethically warranted, may hinder the accurate characterization and evaluation of novel vaccine strategies with little to no appreciable benefits to the vaccinated mice.
Collapse
Affiliation(s)
- Marilynn J. Culbreth
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Comparative Medicine Division, Fort Detrick, Frederick, MD 21702, USA;
| | - Sergei S. Biryukov
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
| | - Jennifer L. Shoe
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
| | - Jennifer L. Dankmeyer
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
| | - Melissa Hunter
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
| | - Christopher P. Klimko
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
| | - Raysa Rosario-Acevedo
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
| | - David P. Fetterer
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Biostatistics Medicine Division, Fort Detrick, Frederick, MD 21702, USA;
| | - Alicia M. Moreau
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Pathology Division, Fort Detrick, Frederick, MD 21702, USA;
| | - Susan L. Welkos
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
| | - Christopher K. Cote
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Bacteriology Division, Fort Detrick, Frederick, MD 21702, USA; (S.S.B.); (J.L.S.); (J.L.D.); (M.H.); (C.P.K.); (R.R.-A.); (S.L.W.)
- Correspondence:
| |
Collapse
|
12
|
Shift from primary pneumonic to secondary septicemic plague by decreasing the volume of intranasal challenge with Yersinia pestis in the murine model. PLoS One 2019; 14:e0217440. [PMID: 31121001 PMCID: PMC6532925 DOI: 10.1371/journal.pone.0217440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 05/10/2019] [Indexed: 12/17/2022] Open
Abstract
Yersinia pestis is the causative agent of pneumonic plague, a disease involving uncontrolled bacterial growth and host immunopathology. Secondary septicemic plague commonly occurs as a consequence of the host inflammatory response that causes vasodilation and vascular leakage, which facilitates systemic spread of the bacteria and the colonization of secondary tissues. The mortality rates of pneumonic and septicemic plague are high even when antibiotics are administered. In this work, we show that primary pneumonic or secondary septicemic plague can be preferentially modeled in mice by varying the volume used for intranasal delivery of Y. pestis. Low volume intranasal challenge (10μL) of wild type Y. pestis resulted in a high frequency of lethal secondary septicemic plague, with a low degree of primary lung infection and rapid development of sepsis. In contrast, high volume intranasal challenge (30μL) yielded uniform early lung infection and primary disease and a significant increase in lethality. In a commonly used BSL2 model, high volume challenge with Y. pestis lacking the pigmentation locus (pgm-) gave 105-fold greater deposition compared to low volume challenge, yet moribund mice did not develop severe lung disease and there was no detectable difference in lethality. These data indicate the primary cause of death of mice in the BSL2 model is sepsis regardless of intranasal dosing method. Overall, these findings allow for the preferential modeling of pneumonic or septicemic plague by intranasal dosing of mice with Y. pestis.
Collapse
|
13
|
Yersinia pestis Pla Protein Thwarts T Cell Defense against Plague. Infect Immun 2019; 87:IAI.00126-19. [PMID: 30804102 DOI: 10.1128/iai.00126-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 01/08/2023] Open
Abstract
Plague is a rapidly lethal human disease caused by the bacterium Yersinia pestis This study demonstrated that the Y. pestis plasminogen activator Pla, a protease that promotes fibrin degradation, thwarts T cell-mediated defense against fully virulent Y. pestis Introducing a single point mutation into the active site of Pla suffices to render fully virulent Y. pestis susceptible to primed T cells. Mechanistic studies revealed essential roles for fibrin during T cell-mediated defense against Pla-mutant Y. pestis Moreover, the efficacy of T cell-mediated protection against various Y. pestis strains displayed an inverse relationship with their levels of Pla activity. Together, these data indicate that Pla functions to thwart fibrin-dependent T cell-mediated defense against plague. Other important human bacterial pathogens, including staphylococci, streptococci, and borrelia, likewise produce virulence factors that promote fibrin degradation. The discovery that Y. pestis thwarts T cell defense by promoting fibrinolysis suggests novel therapeutic approaches to amplifying T cell responses against human pathogens.
Collapse
|
14
|
Yersinia pestis Exploits Early Activation of MyD88 for Growth in the Lungs during Pneumonic Plague. Infect Immun 2019; 87:IAI.00757-18. [PMID: 30642901 PMCID: PMC6434131 DOI: 10.1128/iai.00757-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
Yersinia pestis causes bubonic, pneumonic, and septicemic plague. Although no longer responsible for pandemic outbreaks, pneumonic plague continues to be a challenge for medical treatment and has been classified as a reemerging disease in some parts of the world. In the early stage of infection, inflammatory responses are believed to be suppressed by Y. pestis virulence factors in order to prevent clearance, while later, the hyperactivation of inflammation contributes to the progression of disease. In this work, we sought to identify the host factors that mediate this process and studied the role of the Toll/interleukin 1 (IL-1) receptor adapter and major inflammatory mediator myeloid differentiation primary response 88 (MyD88) in pneumonic plague. We show that pulmonary challenge of Myd88-/- mice with wild-type (WT) Y. pestis results in significant loss of pro- and anti-inflammatory cytokines and chemokines, especially gamma interferon (IFN-γ) and KC, in the lungs compared to that in WT mice. Bacterial growth in the lungs occurred more rapidly in the WT mice, however, indicating a role for the MyD88 response in facilitating the primary lung infection. Nevertheless, Myd88-/- mice were more sensitive to lethality from secondary septicemic plague. Together these findings indicate a central role for MyD88 during the biphasic inflammatory response to pulmonary Y. pestis infection. In the early phase, low-level MyD88-dependent chemokine expression limits initial growth but facilitates Y. pestis access to a protected replicative niche. The later hyperinflammatory phase is partially MyD88 dependent and ineffective in the lungs but controls systemic infection and reduces the progression of secondary septicemic plague.
Collapse
|
15
|
Verma SK, Gupta A, Batra L, Tuteja U. Escherichia coli expressed flagellin C (FliC) of Salmonella Typhi improved the protective efficacy of YopE against plague infection. Vaccine 2018; 37:19-24. [PMID: 30497835 DOI: 10.1016/j.vaccine.2018.11.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 10/27/2022]
Abstract
In the current antibiotic resistance scenario, vaccines may provide best defense against lethal bacterial diseases. So far, there is no idealvaccine available against plague. Despite providing complete protection in small animal models, F1/LcrV based vaccine failed to provide ideal protection in non human primates. Here, we cloned, expressed and purified YopE of Yersinia pestis and flagellin C (FliC) of Salmonella Typhi. However the best possible protection needs the significant induction of IFN-γ and TNF-α. To determine the protective potential of the recombinant YopE alone or in formulation with FliC, Balb/C mice were immunized subcutaneously. The formulations were prepared with alum, a human compatible adjuvant. In our studies, the combination of YopE + FliC induced significantly strong humoral and cellular immune responses. A combination of YopE + FliC provided 83% protection whereas YopE alone provided only 50% against 100LD50 of Y. pestis in a mouse model.
Collapse
Affiliation(s)
- Shailendra K Verma
- Microbiology Division, Defence Research & Development Establishment, Jhansi Road, Gwalior 474002, India.
| | - Ankit Gupta
- Microbiology Division, Defence Research & Development Establishment, Jhansi Road, Gwalior 474002, India
| | - Lalit Batra
- Microbiology Division, Defence Research & Development Establishment, Jhansi Road, Gwalior 474002, India
| | - Urmil Tuteja
- Microbiology Division, Defence Research & Development Establishment, Jhansi Road, Gwalior 474002, India
| |
Collapse
|
16
|
Demeure CE, Derbise A, Guillas C, Gerke C, Cauchemez S, Carniel E, Pizarro-Cerdá J. Humoral and cellular immune correlates of protection against bubonic plague by a live Yersinia pseudotuberculosis vaccine. Vaccine 2018; 37:123-129. [PMID: 30467064 DOI: 10.1016/j.vaccine.2018.11.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/05/2018] [Accepted: 11/11/2018] [Indexed: 11/29/2022]
Abstract
Immunization with the live-attenuated Yersinia pseudotuberculosis VTnF1 strain producing a Yersinia pestis F1 pseudocapsule efficiently protects mice against bubonic and pneumonic plague. In clinical trials, demonstration of a plague vaccine's efficacy in humans will not be feasible, and correlates of protection will be needed to bridge the immune response of protected animals to that of vaccinated humans. Using serum transfer and vaccination of antibody-deficient µMT mice, we established that both humoral and cellular responses elicited by VTnF1 independently conferred protection against bubonic plague. Thus, correlates were searched for in both responses, using blood only. Mice were vaccinated with increasing doses of VTnF1 to provide a range of immune responses and survival outcomes. The cellular response was evaluated using an in vitro IFNγ release assay, and IFNγ levels were significantly associated with protection, although some survivors were negative for IFNγ, so that IFNγ release is not a fully satisfactory correlate. Abundant serum IgG against the F1 capsule, Yop injectable toxins, and also non-F1 Y.pestis antigens were found, but none against the LcrV antigen. All readouts correlated to survival and to each other, confirming that vaccination triggered multiple protective mechanisms developing in parallel. Anti-F1 IgG was the most stringent correlate of protection, in both inbred BALB/c mice and outbred OF1 mice. This indicates that antibodies (Ab) to F1 play a dominant role for protection even in the presence of Ab to many other targets. Easy to measure, the anti-F1 IgG titer will be useful to evaluate the immune response in other animal species and in clinical trials.
Collapse
Affiliation(s)
- Christian E Demeure
- Unité de Recherche Yersinia, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| | - Anne Derbise
- Unité de Recherche Yersinia, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| | - Chloé Guillas
- Unité de Recherche Yersinia, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| | - Christiane Gerke
- Vaccine Programs, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| | - Simon Cauchemez
- Unité de Modélisation Mathématique des Maladies Infectieuses, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France; Centre National de la Recherche Scientifique, URA3012, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France; Center of Bioinformatics, Biostatistics and Integrative Biology, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| | - Elisabeth Carniel
- Unité de Recherche Yersinia, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| | - Javier Pizarro-Cerdá
- Unité de Recherche Yersinia, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
17
|
Leal EA, Moreira JD, Nunes FF, Souza LR, Martins JM, Toledo VPC, Almeida AMP, Guimarães TMP. Humoral and cellular immune response of mice challenged with Yersinia pestis antigenic preparations. Braz J Infect Dis 2017; 21:620-626. [PMID: 29031042 PMCID: PMC9425539 DOI: 10.1016/j.bjid.2017.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/08/2017] [Accepted: 09/26/2017] [Indexed: 11/29/2022] Open
Abstract
Objectives The plague, which is an infectious disease caused by Yersinia pestis, still threatens many populations in several countries. The worldwide increase in human plague cases and the potential use of the bacteria as a biological weapon reinforce the need to study the immunity that is induced by potential vaccine candidates. To determine the immunogenicity of antigenic preparations based on the F1 protein and the total extract from Y. pestis, we assessed the role of these antigens in inducing an immune response. Methods The immunogenicity of antigenic preparations based on the Y. pestis (YP) total extract and the Y. pestis fraction 1 capsular antigen protein (F1) was determined in Swiss-Webster mice immunized with 40 μg or 20 μg for each preparation. Immunophenotyping was performed by flow cytometry. Results Animals immunized with the YP total extract did not elicit detectable anti-F1 antibodies (Ab) in the hemaglutination/inhibition (HA/HI) test. Animals immunized with 40 μg or 20 μg of the F1 protein produced anti-F1 Abs, with titres ranging from 1/16 to 1/8132. The average of CD3+–CD4+ and CD3+–CD8+ T cells did not differ significantly between the groups. Neither YP total extract nor F1 protein induced a significant expression of IFN-γ and IL-10 in CD4+ T lymphocytes. In addition, F1 failed to induce IFN-γ expression in CD8+ T cells, unlike the YP total extract. Conclusion The results showed that F1 protein is not an immunogenic T cell antigen, although the YP total extract (40 μg dose) favoured CD8+ T cell-mediated cellular immunity.
Collapse
Affiliation(s)
- Elida A Leal
- Instituto Octavio Magalhães, Divisão de Epidemologia e Controle de Doenças, Serviço de Doenças Bacterianas e Fúngicas, Belo Horizonte, MG, Brazil.
| | - Josimar D Moreira
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| | - Fernanda F Nunes
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| | - Larissa R Souza
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| | - Janaina M Martins
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| | - Vicente P C Toledo
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| | - Alzira M P Almeida
- Fundação Oswaldo Cruz, Centro de Pesquisa Aggeu Magalhães, Departamento de Microbiologia, Recife, PE, Brazil
| | - Tania M P Guimarães
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| |
Collapse
|
18
|
Perry RD, Bobrov AG, Fetherston JD. The role of transition metal transporters for iron, zinc, manganese, and copper in the pathogenesis of Yersinia pestis. Metallomics 2016; 7:965-78. [PMID: 25891079 DOI: 10.1039/c4mt00332b] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Yersinia pestis, the causative agent of bubonic, septicemic and pneumonic plague, encodes a multitude of Fe transport systems. Some of these are defective due to frameshift or IS element insertions, while others are functional in vitro but have no established role in causing infections. Indeed only 3 Fe transporters (Ybt, Yfe and Feo) have been shown to be important in at least one form of plague. The yersiniabactin (Ybt) system is essential in the early dermal/lymphatic stages of bubonic plague, irrelevant in the septicemic stage, and critical in pneumonic plague. Two Mn transporters have been characterized (Yfe and MntH). These two systems play a role in bubonic plague but the double yfe mntH mutant is fully virulent in a mouse model of pneumonic plague. The same in vivo phenotype occurs with a mutant lacking two (Yfe and Feo) of four ferrous transporters. A role for the Ybt siderophore in Zn acquisition has been revealed. Ybt-dependent Zn acquisition uses a transport system completely independent of the Fe-Ybt uptake system. Together Ybt components and ZnuABC play a critical role in Zn acquisition in vivo. Single mutants in either system retain high virulence in a mouse model of septicemic plague while the double mutant is completely avirulent.
Collapse
Affiliation(s)
- Robert D Perry
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA.
| | | | | |
Collapse
|
19
|
Singh AK, Kingston JJ, Gupta SK, Batra HV. Recombinant Bivalent Fusion Protein rVE Induces CD4+ and CD8+ T-Cell Mediated Memory Immune Response for Protection Against Yersinia enterocolitica Infection. Front Microbiol 2015; 6:1407. [PMID: 26733956 PMCID: PMC4679870 DOI: 10.3389/fmicb.2015.01407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/25/2015] [Indexed: 11/16/2022] Open
Abstract
Studies investigating the correlates of immune protection against Yersinia infection have established that both humoral and cell mediated immune responses are required for the comprehensive protection. In our previous study, we established that the bivalent fusion protein (rVE) comprising immunologically active regions of Y. pestis LcrV (100–270 aa) and YopE (50–213 aa) proteins conferred complete passive and active protection against lethal Y. enterocolitica 8081 challenge. In the present study, cohort of BALB/c mice immunized with rVE or its component proteins rV, rE were assessed for cell mediated immune responses and memory immune protection against Y. enterocolitica 8081. rVE immunization resulted in extensive proliferation of both CD4 and CD8 T cell subsets; significantly high antibody titer with balanced IgG1: IgG2a/IgG2b isotypes (1:1 ratio) and up-regulation of both Th1 (TNF-α, IFN-γ, IL-2, and IL-12) and Th2 (IL-4) cytokines. On the other hand, rV immunization resulted in Th2 biased IgG response (11:1 ratio) and proliferation of CD4+ T-cell; rE group of mice exhibited considerably lower serum antibody titer with predominant Th1 response (1:3 ratio) and CD8+ T-cell proliferation. Comprehensive protection with superior survival (100%) was observed among rVE immunized mice when compared to the significantly lower survival rates among rE (37.5%) and rV (25%) groups when IP challenged with Y. enterocolitica 8081 after 120 days of immunization. Findings in this and our earlier studies define the bivalent fusion protein rVE as a potent candidate vaccine molecule with the capability to concurrently stimulate humoral and cell mediated immune responses and a proof of concept for developing efficient subunit vaccines against Gram negative facultative intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Amit K Singh
- Department of Microbiology, Defence Food Research Laboratory, Defence Research and Development Organisation Mysore, India
| | - Joseph J Kingston
- Department of Microbiology, Defence Food Research Laboratory, Defence Research and Development Organisation Mysore, India
| | - Shishir K Gupta
- Department of Bioinformatics, Biocenter, University of Würzburg Würzburg, Germany
| | - Harsh V Batra
- Department of Microbiology, Defence Food Research Laboratory, Defence Research and Development Organisation Mysore, India
| |
Collapse
|
20
|
Evseeva VV, Platonov ME, Kopylov PK, Dentovskaya SV, Anisimov AP. PLASMINOGEN ACTIVATOR OF YERSINIA PESTIS. ACTA ACUST UNITED AC 2015. [DOI: 10.15789/2220-7619-2015-1-27-36] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
21
|
Alem F, Yao K, Lane D, Calvert V, Petricoin EF, Kramer L, Hale ML, Bavari S, Panchal RG, Hakami RM. Host response during Yersinia pestis infection of human bronchial epithelial cells involves negative regulation of autophagy and suggests a modulation of survival-related and cellular growth pathways. Front Microbiol 2015; 6:50. [PMID: 25762983 PMCID: PMC4327736 DOI: 10.3389/fmicb.2015.00050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/14/2015] [Indexed: 12/16/2022] Open
Abstract
Yersinia pestis (Yp) causes the re-emerging disease plague, and is classified by the CDC and NIAID as a highest priority (Category A) pathogen. Currently, there is no approved human vaccine available and advances in early diagnostics and effective therapeutics are urgently needed. A deep understanding of the mechanisms of host response to Yp infection can significantly advance these three areas. We employed the Reverse Phase Protein Microarray (RPMA) technology to reveal the dynamic states of either protein level changes or phosphorylation changes associated with kinase-driven signaling pathways during host cell response to Yp infection. RPMA allowed quantitative profiling of changes in the intracellular communication network of human lung epithelial cells at different times post infection and in response to different treatment conditions, which included infection with the virulent Yp strain CO92, infection with a derivative avirulent strain CO92 (Pgm-, Pst-), treatment with heat inactivated CO92, and treatment with LPS. Responses to a total of 111 validated antibodies were profiled, leading to discovery of 12 novel protein hits. The RPMA analysis also identified several protein hits previously reported in the context of Yp infection. Furthermore, the results validated several proteins previously reported in the context of infection with other Yersinia species or implicated for potential relevance through recombinant protein and cell transfection studies. The RPMA results point to strong modulation of survival/apoptosis and cell growth pathways during early host response and also suggest a model of negative regulation of the autophagy pathway. We find significant cytoplasmic localization of p53 and reduced LC3-I to LC3-II conversion in response to Yp infection, consistent with negative regulation of autophagy. These studies allow for a deeper understanding of the pathogenesis mechanisms and the discovery of innovative approaches for prevention, early diagnosis, and treatment of plague.
Collapse
Affiliation(s)
- Farhang Alem
- National Center for Biodefense and Infectious Diseases and School of Systems Biology, George Mason University Manassas, VA, USA
| | - Kuan Yao
- National Center for Biodefense and Infectious Diseases and School of Systems Biology, George Mason University Manassas, VA, USA
| | - Douglas Lane
- U.S. Army Medical Research Institute of Infectious Diseases Frederick, MD, USA
| | - Valerie Calvert
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason University Manassas, VA, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason University Manassas, VA, USA
| | - Liana Kramer
- National Center for Biodefense and Infectious Diseases and School of Systems Biology, George Mason University Manassas, VA, USA
| | - Martha L Hale
- U.S. Army Medical Research Institute of Infectious Diseases Frederick, MD, USA
| | - Sina Bavari
- U.S. Army Medical Research Institute of Infectious Diseases Frederick, MD, USA
| | - Rekha G Panchal
- U.S. Army Medical Research Institute of Infectious Diseases Frederick, MD, USA
| | - Ramin M Hakami
- National Center for Biodefense and Infectious Diseases and School of Systems Biology, George Mason University Manassas, VA, USA
| |
Collapse
|
22
|
Batra L, Verma SK, Nagar DP, Saxena N, Pathak P, Pant SC, Tuteja U. HSP70 domain II of Mycobacterium tuberculosis modulates immune response and protective potential of F1 and LcrV antigens of Yersinia pestis in a mouse model. PLoS Negl Trop Dis 2014; 8:e3322. [PMID: 25474358 PMCID: PMC4256173 DOI: 10.1371/journal.pntd.0003322] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/07/2014] [Indexed: 02/04/2023] Open
Abstract
No ideal vaccine exists to control plague, a deadly dangerous disease caused by Yersinia pestis. In this context, we cloned, expressed and purified recombinant F1, LcrV antigens of Y. pestis and heat shock protein70 (HSP70) domain II of M. tuberculosis in E. coli. To evaluate the protective potential of each purified protein alone or in combination, Balb/C mice were immunized. Humoral and cell mediated immune responses were evaluated. Immunized animals were challenged with 100 LD50 of Y. pestis via intra-peritoneal route. Vaccine candidates i.e., F1 and LcrV generated highly significant titres of anti-F1 and anti-LcrV IgG antibodies. A significant difference was noticed in the expression level of IL-2, IFN-γ and TNF-α in splenocytes of immunized animals. Significantly increased percentages of CD4+ and CD8+ T cells producing IFN-γ in spleen of vaccinated animals were observed in comparison to control group by flow cytometric analysis. We investigated whether the F1, LcrV and HSP70(II) antigens alone or in combination can effectively protect immunized animals from any histopathological changes. Signs of histopathological lesions noticed in lung, liver, kidney and spleen of immunized animals on 3rd day post challenge whereas no lesions in animals that survived to day 20 post-infection were observed. Immunohistochemistry showed bacteria in lung, liver, spleen and kidney on 3rd day post-infection whereas no bacteria was observed on day 20 post-infection in surviving animals in LcrV, LcrV+HSP70(II), F1+LcrV, and F1+LcrV+HSP70(II) vaccinated groups. A significant difference was observed in the expression of IL-2, IFN-γ, TNF-α, and CD4+/CD8+ T cells secreting IFN-γ in the F1+LcrV+HSP70(II) vaccinated group in comparison to the F1+LcrV vaccinated group. Three combinations that included LcrV+HSP70(II), F1+LcrV or F1+LcrV+HSP70(II) provided 100% protection, whereas LcrV alone provided only 75% protection. These findings suggest that HSP70(II) of M. tuberculosis can be a potent immunomodulator for F1 and LcrV containing vaccine candidates against plague.
Collapse
Affiliation(s)
- Lalit Batra
- Microbiology Division, Defence Research & Development Establishment, Gwalior, India
| | - Shailendra K. Verma
- Microbiology Division, Defence Research & Development Establishment, Gwalior, India
| | - Durgesh P. Nagar
- Pharmacology and Toxicology Division, Defence Research & Development Establishment, Gwalior, India
| | - Nandita Saxena
- Pharmacology and Toxicology Division, Defence Research & Development Establishment, Gwalior, India
| | - Prachi Pathak
- Microbiology Division, Defence Research & Development Establishment, Gwalior, India
| | - Satish C. Pant
- Pharmacology and Toxicology Division, Defence Research & Development Establishment, Gwalior, India
| | - Urmil Tuteja
- Microbiology Division, Defence Research & Development Establishment, Gwalior, India
| |
Collapse
|
23
|
Gaddy CE, Cuevas PF, Hartman LJ, Howe GB, Worsham PL, Minogue TD. Development of real-time PCR assays for specific detection of hmsH, hmsF, hmsR, and irp2 located within the 102-kb pgm locus of Yersinia pestis. Mol Cell Probes 2014; 28:288-95. [PMID: 25261118 DOI: 10.1016/j.mcp.2014.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 01/20/2023]
Abstract
Virulent isolates of three pathogenic Yersinia species (Yersinia pestis, Yersinia pseudotuberculosis, and Yersinia enterocolitica) harbor a 102-kb chromosomal region which encodes elements critical for virulence. A 35-kb high pathogenicity island is contained in this region, is a known virulence determinant, contains irp1 and irp2 iron-regulating genes. An additional segment, the 68-kb high pathogenicity island, contains genetic elements responsible for conferring the Y. pestis pigmentation phenotype on Congo red agar at 28 °C. Collectively, these contiguous segments are referred to as the pigmentation (pgm) locus, the absence of which results in strain attenuation and exemption from CDC Select Agent status. In this study, we developed a set of four real-time PCR assays to detect the presence or absence of multiple virulence genes located within this region. Specifically, we designed TaqMan(®) PCR assays to individually detect three hemin storage genes (hmsH, hmsF, and hmsR) which are genetic elements that confer the pigmentation phenotype, as well as the iron-regulating status of 25 Y. pestis isolates (representing 23 different strains), thus establishing a molecular based assay capable of determining the pgm status of candidate Y. pestis isolates. Included in the validation process, was a comparison of these real-time PCR assays and newly developed conventional PCR assays targeting much larger areas of the 102-kb region (including one assay spanning hmsR and hmsF, one spanning hmsH and hsmF, one targeting hmsF, and one targeting irp2). There was high concordance between the conventional and real-time PCR assays for all Y. pestis strains tested. The results from the comparative analysis document the specificity and sensitivity of the real-time PCR assays and further solidify the ostensible benefits of real-time PCR over conventional PCR.
Collapse
Affiliation(s)
- Charla E Gaddy
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Pedro F Cuevas
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Laurie J Hartman
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA; ClinicalRM, Inc., Hinckley, OH, USA
| | - Gerald B Howe
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Patricia L Worsham
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Timothy D Minogue
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA.
| |
Collapse
|
24
|
GRAHAM CHRISTINEB, WOODS MICHAELE, VETTER SARAM, PETERSEN JEANNINEM, MONTENIERI JOHNA, HOLMES JENNIFERL, MAES SARAHE, BEARDEN SCOTTW, GAGE KENNETHL, EISEN REBECCAJ. Evaluation of the effect of host immune status on short-term Yersinia pestis infection in fleas with implications for the enzootic host model for maintenance of Y. pestis during interepizootic periods. JOURNAL OF MEDICAL ENTOMOLOGY 2014; 51:1079-86. [PMID: 25276941 PMCID: PMC4636331 DOI: 10.1603/me14080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Plague, a primarily flea-borne disease caused by Yersinia pestis, is characterized by rapidly spreading epizootics separated by periods of quiescence. Little is known about how and where Y. pestis persists between epizootics. It is commonly proposed, however, that Y pestis is maintained during interepizootic periods in enzootic cycles involving flea vectors and relatively resistant host populations. According to this model, while susceptible individuals serve as infectious sources for feeding fleas and subsequently die of infection, resistant hosts survive infection, develop antibodies to the plague bacterium, and continue to provide bloodmeals to infected fleas. For Y. pestis to persist under this scenario, fleas must remain infected after feeding on hosts carrying antibodies to Y. pestis. Studies of other vector-borne pathogens suggest that host immunity may negatively impact pathogen survival in the vector. Here, we report infection rates and bacterial loads for fleas (both Xenopsylla cheopis (Rothschild) and Oropsylla montana (Baker)) that consumed an infectious bloodmeal and subsequently fed on an immunized or age-matched naive mouse. We demonstrate that neither the proportion of infected fleas nor the bacterial loads in infected fleas were significantly lower within 3 d of feeding on immunized versus naive mice. Our findings thus provide support for one assumption underlying the enzootic host model of interepizootic maintenance of Y. pestis.
Collapse
Affiliation(s)
- CHRISTINE B. GRAHAM
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - MICHAEL E. WOODS
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - SARA M. VETTER
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - JEANNINE M. PETERSEN
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - JOHN A. MONTENIERI
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - JENNIFER L. HOLMES
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - SARAH E. MAES
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - SCOTT W. BEARDEN
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - KENNETH L. GAGE
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| | - REBECCA J. EISEN
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Road, Fort Collins, CO 80521
| |
Collapse
|
25
|
Bozue J, Cote CK, Chance T, Kugelman J, Kern SJ, Kijek TK, Jenkins A, Mou S, Moody K, Fritz D, Robinson CG, Bell T, Worsham P. A Yersinia pestis tat mutant is attenuated in bubonic and small-aerosol pneumonic challenge models of infection but not as attenuated by intranasal challenge. PLoS One 2014; 9:e104524. [PMID: 25101850 PMCID: PMC4125294 DOI: 10.1371/journal.pone.0104524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 07/11/2014] [Indexed: 01/01/2023] Open
Abstract
Bacterial proteins destined for the Tat pathway are folded before crossing the inner membrane and are typically identified by an N-terminal signal peptide containing a twin arginine motif. Translocation by the Tat pathway is dependent on the products of genes which encode proteins possessing the binding site of the signal peptide and mediating the actual translocation event. In the fully virulent CO92 strain of Yersinia pestis, the tatA gene was deleted. The mutant was assayed for loss of virulence through various in vitro and in vivo assays. Deletion of the tatA gene resulted in several consequences for the mutant as compared to wild-type. Cell morphology of the mutant bacteria was altered and demonstrated a more elongated form. In addition, while cultures of the mutant strain were able to produce a biofilm, we observed a loss of adhesion of the mutant biofilm structure compared to the biofilm produced by the wild-type strain. Immuno-electron microscopy revealed a partial disruption of the F1 antigen on the surface of the mutant. The virulence of the ΔtatA mutant was assessed in various murine models of plague. The mutant was severely attenuated in the bubonic model with full virulence restored by complementation with the native gene. After small-particle aerosol challenge in a pneumonic model of infection, the mutant was also shown to be attenuated. In contrast, when mice were challenged intranasally with the mutant, very little difference in the LD50 was observed between wild-type and mutant strains. However, an increased time-to-death and delay in bacterial dissemination was observed in mice infected with the ΔtatA mutant as compared to the parent strain. Collectively, these findings demonstrate an essential role for the Tat pathway in the virulence of Y. pestis in bubonic and small-aerosol pneumonic infection but less important role for intranasal challenge.
Collapse
Affiliation(s)
- Joel Bozue
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
- * E-mail:
| | - Christopher K. Cote
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Taylor Chance
- Pathology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Jeffrey Kugelman
- Center for Genome Sciences, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Steven J. Kern
- Office of Research Support, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Todd K. Kijek
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Amy Jenkins
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Sherry Mou
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Krishna Moody
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - David Fritz
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Camenzind G. Robinson
- Pathology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Todd Bell
- Pathology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Patricia Worsham
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| |
Collapse
|
26
|
Cui Y, Yang X, Xiao X, Anisimov AP, Li D, Yan Y, Zhou D, Rajerison M, Carniel E, Achtman M, Yang R, Song Y. Genetic variations of live attenuated plague vaccine strains (Yersinia pestis EV76 lineage) during laboratory passages in different countries. INFECTION GENETICS AND EVOLUTION 2014; 26:172-9. [PMID: 24905600 DOI: 10.1016/j.meegid.2014.05.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 05/13/2014] [Accepted: 05/21/2014] [Indexed: 12/20/2022]
Abstract
Plague, one of the most devastating infectious diseases in human history, is caused by the bacterial species Yersinia pestis. A live attenuated Y. pestis strain (EV76) has been widely used as a plague vaccine in various countries around the world. Here we compared the whole genome sequence of an EV76 strain used in China (EV76-CN) with the genomes of Y. pestis wild isolates to identify genetic variations specific to the EV76 lineage. We identified 6 SNPs and 6 Indels (insertions and deletions) differentiating EV76-CN from its counterparts. Then, we screened these polymorphic sites in 28 other strains of EV76 lineage that were stored in different countries. Based on the profiles of SNPs and Indels, we reconstructed the parsimonious dissemination history of EV76 lineage. This analysis revealed that there have been at least three independent imports of EV76 strains into China. Additionally, we observed that the pyrE gene is a mutation hotspot in EV76 lineages. The fine comparison results based on whole genome sequence in this study provide better understanding of the effects of laboratory passages on the accumulation of genetic polymorphisms in plague vaccine strains. These variations identified here will also be helpful in discriminating different EV76 derivatives.
Collapse
Affiliation(s)
- Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xianwei Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiao Xiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Andrey P Anisimov
- State Research Center for Applied Microbiology and Biotechnology, Obolensk, Moscow Region, Russia
| | | | - Yanfeng Yan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | - Elisabeth Carniel
- Yersinia Research Unit, National Reference Laboratory, Institut Pasteur, Paris, France
| | - Mark Achtman
- Environmental Research Institute, University College Cork, Cork, Ireland, United Kingdom; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Yajun Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Environmental Research Institute, University College Cork, Cork, Ireland, United Kingdom.
| |
Collapse
|
27
|
Wang X, Zhang X, Zhou D, Yang R. Live-attenuatedYersinia pestisvaccines. Expert Rev Vaccines 2014; 12:677-86. [DOI: 10.1586/erv.13.42] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Zhang Q, Wang Q, Tian G, Qi Z, Zhang X, Wu X, Qiu Y, Bi Y, Yang X, Xin Y, He J, Zhou J, Zeng L, Yang R, Wang X. Yersinia pestis biovar Microtus strain 201, an avirulent strain to humans, provides protection against bubonic plague in rhesus macaques. Hum Vaccin Immunother 2013; 10:368-77. [PMID: 24225642 DOI: 10.4161/hv.27060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Yersinia pestis biovar Microtus is considered to be a virulent to larger mammals, including guinea pigs, rabbits and humans. It may be used as live attenuated plague vaccine candidates in terms of its low virulence. However, the Microtus strain's protection against plague has yet to be demonstrated in larger mammals. In this study, we evaluated the protective efficacy of the Microtus strain 201 as a live attenuated plague vaccine candidate. Our results show that this strain is highly attenuated by subcutaneous route, elicits an F1-specific antibody titer similar to the EV and provides a protective efficacy similar to the EV against bubonic plague in Chinese-origin rhesus macaques. The Microtus strain 201 could induce elevated secretion of both Th1-associated cytokines (IFN-γ, IL-2 and TNF-α) and Th2-associated cytokines (IL-4, IL-5, and IL-6), as well as chemokines MCP-1 and IL-8. However, the protected animals developed skin ulcer at challenge site with different severity in most of the immunized and some of the EV-immunized monkeys. Generally, the Microtus strain 201 represented a good plague vaccine candidate based on its ability to generate strong humoral and cell-mediated immune responses as well as its good protection against high dose of subcutaneous virulent Y. pestis challenge.
Collapse
Affiliation(s)
- Qingwen Zhang
- Anhui Medical University; Hefei, Anhui PR China; Qinghai Institute for Endemic Disease Prevention and Control of Qinghai Province; Xining, PR China
| | - Qiong Wang
- Anhui Medical University; Hefei, Anhui PR China; Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| | - Guang Tian
- Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| | - Zhizhen Qi
- Qinghai Institute for Endemic Disease Prevention and Control of Qinghai Province; Xining, PR China
| | - Xuecan Zhang
- Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| | - Xiaohong Wu
- Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| | - Yefeng Qiu
- Laboratory Animal Research Center; Academy of Military Medical Science; Beijing; PR China
| | - Yujing Bi
- Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| | - Xiaoyan Yang
- Qinghai Institute for Endemic Disease Prevention and Control of Qinghai Province; Xining, PR China
| | - Youquan Xin
- Qinghai Institute for Endemic Disease Prevention and Control of Qinghai Province; Xining, PR China
| | - Jian He
- Qinghai Institute for Endemic Disease Prevention and Control of Qinghai Province; Xining, PR China
| | - Jiyuan Zhou
- Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| | - Lin Zeng
- Laboratory Animal Research Center; Academy of Military Medical Science; Beijing; PR China
| | - Ruifu Yang
- Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| | - Xiaoyi Wang
- Anhui Medical University; Hefei, Anhui PR China; Laboratory of Analytical Microbiology; State Key Laboratory of Pathogen and Biosecurity; Beijing Institute of Microbiology and Epidemiology; Beijing, PR China
| |
Collapse
|
29
|
Luo D, Lin JS, Parent MA, Mullarky-Kanevsky I, Szaba FM, Kummer LW, Duso DK, Tighe M, Hill J, Gruber A, Mackman N, Gailani D, Smiley ST. Fibrin facilitates both innate and T cell-mediated defense against Yersinia pestis. THE JOURNAL OF IMMUNOLOGY 2013; 190:4149-61. [PMID: 23487423 DOI: 10.4049/jimmunol.1203253] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The Gram-negative bacterium Yersinia pestis causes plague, a rapidly progressing and often fatal disease. The formation of fibrin at sites of Y. pestis infection supports innate host defense against plague, perhaps by providing a nondiffusible spatial cue that promotes the accumulation of inflammatory cells expressing fibrin-binding integrins. This report demonstrates that fibrin is an essential component of T cell-mediated defense against plague but can be dispensable for Ab-mediated defense. Genetic or pharmacologic depletion of fibrin abrogated innate and T cell-mediated defense in mice challenged intranasally with Y. pestis. The fibrin-deficient mice displayed reduced survival, increased bacterial burden, and exacerbated hemorrhagic pathology. They also showed fewer neutrophils within infected lung tissue and reduced neutrophil viability at sites of liver infection. Depletion of neutrophils from wild-type mice weakened T cell-mediated defense against plague. The data suggest that T cells combat plague in conjunction with neutrophils, which require help from fibrin to withstand Y. pestis encounters and effectively clear bacteria.
Collapse
Affiliation(s)
- Deyan Luo
- Trudeau Institute, Saranac Lake, NY 12983, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kota KP, Eaton B, Lane D, Ulrich M, Ulrich R, Peyser BD, Robinson CG, Jaissle JG, Pegoraro G, Bavari S, Panchal RG. Integrating high-content imaging and chemical genetics to probe host cellular pathways critical for Yersinia pestis infection. PLoS One 2013; 8:e55167. [PMID: 23383093 PMCID: PMC3559335 DOI: 10.1371/journal.pone.0055167] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 12/19/2012] [Indexed: 01/14/2023] Open
Abstract
The molecular machinery that regulates the entry and survival of Yersinia pestis in host macrophages is poorly understood. Here, we report the development of automated high-content imaging assays to quantitate the internalization of virulent Y. pestis CO92 by macrophages and the subsequent activation of host NF-κB. Implementation of these assays in a focused chemical screen identified kinase inhibitors that inhibited both of these processes. Rac-2-ethoxy-3 octadecanamido-1-propylphosphocholine (a protein Kinase C inhibitor), wortmannin (a PI3K inhibitor), and parthenolide (an IκB kinase inhibitor), inhibited pathogen-induced NF-κB activation and reduced bacterial entry and survival within macrophages. Parthenolide inhibited NF-κB activation in response to stimulation with Pam3CSK4 (a TLR2 agonist), E. coli LPS (a TLR4 agonist) or Y. pestis infection, while the PI3K and PKC inhibitors were selective only for Y. pestis infection. Together, our results suggest that phagocytosis is the major stimulus for NF-κB activation in response to Y. pestis infection, and that Y. pestis entry into macrophages may involve the participation of protein kinases such as PI3K and PKC. More importantly, the automated image-based screening platform described here can be applied to the study of other bacteria in general and, in combination with chemical genetic screening, can be used to identify host cell functions facilitating the identification of novel antibacterial therapeutics.
Collapse
Affiliation(s)
- Krishna P. Kota
- Perkin Elmer, Waltham, Massachusetts, United States of America
| | - Brett Eaton
- Department of Target Discovery and Cellular Microbiology, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Douglas Lane
- Target Structure Based Drug Discovery Group, SAIC-Frederick, NCI-Frederick, Frederick, Maryland, United States of America
| | - Melanie Ulrich
- Department of Target Discovery and Cellular Microbiology, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Ricky Ulrich
- Department of Target Discovery and Cellular Microbiology, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Brian D. Peyser
- Target Structure Based Drug Discovery Group, SAIC-Frederick, NCI-Frederick, Frederick, Maryland, United States of America
| | - Camenzind G. Robinson
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - James G. Jaissle
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | | | - Sina Bavari
- Department of Target Discovery and Cellular Microbiology, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Rekha G. Panchal
- Department of Target Discovery and Cellular Microbiology, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
31
|
Abstract
Synthesis of Escherichia coli LpxL, which transfers a secondary laurate chain to the 2' position of lipid A, in Yersinia pestis produced bisphosphoryl hexa-acylated lipid A at 37°C, leading to significant attenuation of virulence. Our previous observations also indicated that strain χ10015(pCD1Ap) (ΔlpxP32::P(lpxL) lpxL) stimulated a strong inflammatory reaction but sickened mice before recovery and retained virulence via intranasal (i.n.) infection. The development of live, attenuated Y. pestis vaccines may be facilitated by detoxification of its lipopolysaccharide (LPS). Heterologous expression of the lipid A 1-phosphatase, LpxE, from Francisella tularensis in Y. pestis yields predominantly 1-dephosphorylated lipid A, as confirmed by mass spectrometry. Results indicated that expression of LpxE on top of LpxL provided no significant reduction in virulence of Y. pestis in mice when it was administered i.n. but actually reduced the 50% lethal dose (LD(50)) by 3 orders of magnitude when the strain was administered subcutaneously (s.c.). Additionally, LpxE synthesis in wild-type Y. pestis KIM6+(pCD1Ap) led to slight attenuation by s.c. inoculation but no virulence change by i.n. inoculation in mice. In contrast to Salmonella enterica, expression of LpxE does not attenuate the virulence of Y. pestis.
Collapse
|
32
|
Skarjinskaia M, Ruby K, Araujo A, Taylor K, Gopalasamy-Raju V, Musiychuk K, Chichester JA, Palmer GA, de la Rosa P, Mett V, Ugulava N, Streatfield SJ, Yusibov V. Hairy Roots as a Vaccine Production and Delivery System. BIOTECHNOLOGY OF HAIRY ROOT SYSTEMS 2013; 134:115-34. [DOI: 10.1007/10_2013_184] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
33
|
Gater ST, Peters KN, Kocsis AG, Dhariwala MO, Anderson DM, Anderson PE. Host stress and immune responses during aerosol challenge of Brown Norway rats with Yersinia pestis. Front Cell Infect Microbiol 2012; 2:147. [PMID: 23226684 PMCID: PMC3510485 DOI: 10.3389/fcimb.2012.00147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 11/08/2012] [Indexed: 01/25/2023] Open
Abstract
Inhalation exposure models are becoming the preferred method for the comparative study of respiratory infectious diseases due to their resemblance to the natural route of infection. To enable precise delivery of pathogen to the lower respiratory tract in a manner that imposes minimal biosafety risk, nose-only exposure systems have been developed. Early inhalation exposure technology for infectious disease research grew out of technology used in asthma research where predominantly the Collison nebulizer is used to generate an aerosol by beating a liquid sample against glass. Although infectious aerosol droplets of 1–5 μm in size can be generated, the Collison often causes loss of viability. In this work, we evaluate a gentler method for aerosolization of living cells and describe the use of the Sparging Liquid Aerosol Generator (SLAG) in a rat pneumonic plague model. The SLAG creates aerosols by continuous dripping of liquid sample on a porous metal disc. We show the generation of 0.5–1 μm Yersinia pestis aerosol particles using the SLAG with spray factors typically ranging from 10−7 to 10−8 with no detectable loss of bacterial viability. Delivery of these infectious particles via nose-only exposure led to the rapid development of lethal pneumonic plague. Further, we evaluated the effect of restraint-stress imposed by the nose-only exposure chamber on early inflammatory responses and bacterial deposition. Elevated serum corticosterone which peaked at 2 h post-procedure indicated the animals experienced stress as a result of restraint in the nose-only chamber. However, we observed no correlation between elevated corticosterone and the amount of bacterial deposition or inflammation in the lungs. Together these data demonstrate the utility of the SLAG and the nose-only chamber for aerosol challenge of rodents by Y. pestis.
Collapse
Affiliation(s)
- Susan T Gater
- Laboratory for Infectious Disease Research, University of Missouri Columbia, MO, USA
| | | | | | | | | | | |
Collapse
|
34
|
Mellado-Sanchez G, Ramirez K, Drachenberg CB, Diaz-McNair J, Rodriguez AL, Galen JE, Nataro JP, Pasetti MF. Characterization of systemic and pneumonic murine models of plague infection using a conditionally virulent strain. Comp Immunol Microbiol Infect Dis 2012. [PMID: 23195858 DOI: 10.1016/j.cimid.2012.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Yersinia pestis causes bubonic and pneumonic plague in humans. The pneumonic infection is the most severe and invariably fatal if untreated. Because of its high virulence, ease of delivery and precedent of use in warfare, Y. pestis is considered as a potential bioterror agent. No licensed plague vaccine is currently available in the US. Laboratory research with virulent strains requires appropriate biocontainment (i.e., Biosafety Level 3 (BSL-3) for procedures that generate aerosol/droplets) and secure facilities that comply with federal select agent regulations. To assist in the identification of promising vaccine candidates during the early phases of development, we characterized mouse models of systemic and pneumonic plague infection using the Y. pestis strain EV76, an attenuated human vaccine strain that can be rendered virulent in mice under in vivo iron supplementation. Mice inoculated intranasally or intravenously with Y. pestis EV76 in the presence of iron developed a systemic and pneumonic plague infection that resulted in disease and lethality. Bacteria replicated and severely compromised the spleen, liver and lungs. Susceptibility was age dependent, with younger mice being more vulnerable to pneumonic infection. We used these models of infection to assess the protective capacity of newly developed Salmonella-based plague vaccines. The protective outcome varied depending on the route and dose of infection. Protection was associated with the induction of specific immunological effectors in systemic/mucosal compartments. The models of infection described could serve as safe and practical tools for identifying promising vaccine candidates that warrant further potency evaluation using fully virulent strains in BSL-3 settings.
Collapse
Affiliation(s)
- Gabriela Mellado-Sanchez
- Department of Pediatrics, Center for Vaccine Development, University of Maryland School of Medicine, 685 West Baltimore St., Room 480, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Wilson CN, Vance CO, Doyle TM, Brink DS, Matuschak GM, Lechner AJ. A novel post-exposure medical countermeasure L-97-1 improves survival and acute lung injury following intratracheal infection with Yersinia pestis. Innate Immun 2012; 18:373-89. [PMID: 21862597 PMCID: PMC3362682 DOI: 10.1177/1753425911411595] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Yersinia pestis, a Gram-negative bacillus causing plague and Centers for Disease Control and Prevention (CDC) classified Category A pathogen, has high potential as a bioweapon. Lipopolysaccharide, a virulence factor for Y. pestis, binds to and activates A(1) adenosine receptor (AR)s and, in animals, A(1)AR antagonists block induced acute lung injury (ALI) and increase survival following cecal ligation and perforation. In this study, rats were infected intratracheally with viable Y. pestis [CO99 (pCD1( + )/Δpgm) 1 × 10( 8 ) CFU/animal] and treated daily for 3 d with ciprofloxacin (cipro), the A(1)AR antagonist L-97-1, or cipro plus L-97-1 starting at 0, 6, 24, 48, or 72 h post-Y. pestis. At 72 h post-Y. pestis, cipro plus L-97-1 significantly improved 6-d survival to 60-70% vs 28% for cipro plus H(2)O and 33% for untreated Y. pestis controls (P = 0.02, logrank test). Lung edema, hemorrhage and leukocyte infiltration index (LII) were evaluated histologically to produce ALI scores. Cipro plus L-97-1 significantly reduced lung edema, as well as aggregate lung injury scores vs controls or cipro plus H(2)O, and LII vs controls (P < 0.05, Student's unpaired t test). These results support efficacy for L-97-1 as a post-exposure medical countermeasure, adjunctive therapy to antibiotics for Y. pestis.
Collapse
|
36
|
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: 1.3] [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
|
37
|
Sergueev KV, Nikolich MP, Filippov AA. Field and clinical applications of advanced bacteriophage-based detection of Yersinia pestis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 954:135-41. [PMID: 22782756 DOI: 10.1007/978-1-4614-3561-7_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kirill V Sergueev
- Department of Emerging Bacterial Infections, Division of Bacterial and Rickettsial Diseases, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA.
| | | | | |
Collapse
|
38
|
Filippov AA, Sergueev KV, He Y, Huang XZ, Gnade BT, Mueller AJ, Fernandez-Prada CM, Nikolich MP. Bacteriophage therapy of experimental bubonic plague in mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 954:337-48. [PMID: 22782780 DOI: 10.1007/978-1-4614-3561-7_41] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Andrey A Filippov
- Department of Emerging Bacterial Infections, Division of Bacterial and Rickettsial Diseases, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bacteriophages capable of lysing Yersinia pestis and Yersinia pseudotuberculosis: efficiency of plating tests and identification of receptors in escherichia coli K-12. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 954:123-34. [PMID: 22782755 DOI: 10.1007/978-1-4614-3561-7_16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Humoral and cellular immune responses to Yersinia pestis infection in long-term recovered plague patients. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 19:228-34. [PMID: 22190397 DOI: 10.1128/cvi.05559-11] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Plague is one of the most dangerous diseases and is caused by Yersinia pestis. Effective vaccine development requires understanding of immune protective mechanisms against the bacterium in humans. In this study, the humoral and memory cellular immune responses in plague patients (n = 65) recovered from Y. pestis infection during the past 16 years were investigated using a protein microarray and an enzyme-linked immunosorbent spot assay (ELISpot). The seroprevalence to the F1 antigen in all recovered patients is 78.5%. In patients infected more than a decade ago, the antibody-positive rate still remains 69.5%. There is no difference in the antibody presence between gender, age, and infected years, but it seems to be associated with the F1 antibody titers during infection (r = 0.821; P < 0.05). Except F1 antibody, the antibodies against LcrV and YopD were detected in most of the patients, suggesting they could be the potential diagnostic markers for detecting the infection of F1-negative strains. Regarding cellular immunity, the cell number producing gamma interferon (IFN-γ), stimulated by F1 and LcrV, respectively, in vitro to the peripheral blood mononuclear cells of 7 plague patients and 4 negative controls, showed no significant difference, indicating F1 and LcrV are not dominant T cell antigens against plague for a longer time in humans. Our findings have direct implications for the future design and development of effective vaccines against Y. pestis infection and the development of new target-based diagnostics.
Collapse
|
41
|
Filippov AA, Sergueev KV, He Y, Huang XZ, Gnade BT, Mueller AJ, Fernandez-Prada CM, Nikolich MP. Bacteriophage-resistant mutants in Yersinia pestis: identification of phage receptors and attenuation for mice. PLoS One 2011; 6:e25486. [PMID: 21980477 PMCID: PMC3182234 DOI: 10.1371/journal.pone.0025486] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 09/06/2011] [Indexed: 01/21/2023] Open
Abstract
Background Bacteriophages specific for Yersinia pestis are routinely used for plague diagnostics and could be an alternative to antibiotics in case of drug-resistant plague. A major concern of bacteriophage therapy is the emergence of phage-resistant mutants. The use of phage cocktails can overcome this problem but only if the phages exploit different receptors. Some phage-resistant mutants lose virulence and therefore should not complicate bacteriophage therapy. Methodology/Principal Findings The purpose of this work was to identify Y. pestis phage receptors using site-directed mutagenesis and trans-complementation and to determine potential attenuation of phage-resistant mutants for mice. Six receptors for eight phages were found in different parts of the lipopolysaccharide (LPS) inner and outer core. The receptor for R phage was localized beyond the LPS core. Most spontaneous and defined phage-resistant mutants of Y. pestis were attenuated, showing increase in LD50 and time to death. The loss of different LPS core biosynthesis enzymes resulted in the reduction of Y. pestis virulence and there was a correlation between the degree of core truncation and the impact on virulence. The yrbH and waaA mutants completely lost their virulence. Conclusions/Significance We identified Y. pestis receptors for eight bacteriophages. Nine phages together use at least seven different Y. pestis receptors that makes some of them promising for formulation of plague therapeutic cocktails. Most phage-resistant Y. pestis mutants become attenuated and thus should not pose a serious problem for bacteriophage therapy of plague. LPS is a critical virulence factor of Y. pestis.
Collapse
Affiliation(s)
- Andrey A Filippov
- Division of Bacterial and Rickettsial Diseases, Department of Emerging Bacterial Infections, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Sun W, Roland KL, Curtiss R. Developing live vaccines against plague. J Infect Dev Ctries 2011; 5:614-27. [PMID: 21918302 PMCID: PMC3932668 DOI: 10.3855/jidc.2030] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 05/20/2011] [Accepted: 05/22/2011] [Indexed: 12/13/2022] Open
Abstract
Three great plague pandemics caused by the gram-negative bacterium Yersinia pestis have killed nearly 200 million people and it has been linked to biowarfare in the past. Plague is endemic in many parts of the world. In addition, the risk of plague as a bioweapon has prompted increased research to develop plague vaccines against this disease. Injectable subunit vaccines are being developed in the United States and United Kingdom. However, the live attenuated Y. pestis-EV NIIEG strain has been used as a vaccine for more than 70 years in the former Soviet Union and in some parts of Asia and provides a high degree of efficacy against plague. This vaccine has not gained general acceptance because of safety concerns. In recent years, modern molecular biological techniques have been applied to Y. pestis to construct strains with specific defined mutations designed to create safe, immunogenic vaccines with potential for use in humans and as bait vaccines to reduce the load of Y. pestis in the environment. In addition, a number of live, vectored vaccines have been reported using attenuated viral vectors or attenuated Salmonella strains to deliver plague antigens. Here we summarize the progress of live attenuated vaccines against plagu.
Collapse
Affiliation(s)
- Wei Sun
- Center for Infectious Disease and Vaccinology, The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85287-5401 , USA
| | | | | |
Collapse
|
43
|
Rosenzweig JA, Jejelowo O, Sha J, Erova TE, Brackman SM, Kirtley ML, van Lier CJ, Chopra AK. Progress on plague vaccine development. Appl Microbiol Biotechnol 2011; 91:265-86. [PMID: 21670978 DOI: 10.1007/s00253-011-3380-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 05/11/2011] [Accepted: 05/11/2011] [Indexed: 12/15/2022]
Abstract
Yersinia pestis (YP), the gram-negative plague bacterium, has shaped human history unlike any other pathogen known to mankind. YP (transmitted by the bite of an infected flea) diverged only recently from the related enteric pathogen Yersinia pseudotuberculosis but causes radically different diseases. Three forms of plague exist in humans: bubonic (swollen lymph nodes or bubos), septicemic (spread of YP through the lymphatics or bloodstream from the bubos to other organs), and contagious, pneumonic plague which can be communicated via YP-charged respiratory droplets resulting in person-person transmission and rapid death if left untreated (50-90% mortality). Despite the potential threat of weaponized YP being employed in bioterrorism and YP infections remaining prevalent in endemic regions of the world where rodent populations are high (including the four corner regions of the USA), an efficacious vaccine that confers immunoprotection has yet to be developed. This review article will describe the current vaccine candidates being evaluated in various model systems and provide an overall summary on the progress of this important endeavor.
Collapse
Affiliation(s)
- Jason A Rosenzweig
- Department of Biology, Center for Bionanotechnology and Environmental Research (CBER), Texas Southern University, 3100 Cleburne Street, Houston, TX 77004, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lin JS, Szaba FM, Kummer LW, Chromy BA, Smiley ST. Yersinia pestis YopE contains a dominant CD8 T cell epitope that confers protection in a mouse model of pneumonic plague. THE JOURNAL OF IMMUNOLOGY 2011; 187:897-904. [PMID: 21653834 DOI: 10.4049/jimmunol.1100174] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Septic bacterial pneumonias are a major cause of death worldwide. Several of the highest priority bioterror concerns, including anthrax, tularemia, and plague, are caused by bacteria that acutely infect the lung. Bacterial resistance to multiple antibiotics is increasingly common. Although vaccines may be our best defense against antibiotic-resistant bacteria, there has been little progress in the development of safe and effective vaccines for pulmonary bacterial pathogens. The Gram-negative bacterium Yersinia pestis causes pneumonic plague, an acutely lethal septic pneumonia. Historic pandemics of plague caused millions of deaths, and the plague bacilli's potential for weaponization sustains an ongoing quest for effective countermeasures. Subunit vaccines have failed, to date, to fully protect nonhuman primates. In mice, they induce the production of Abs that act in concert with type 1 cytokines to deliver high-level protection; however, the Y. pestis Ags recognized by cytokine-producing T cells have yet to be defined. In this study, we report that Y. pestis YopE is a dominant Ag recognized by CD8 T cells in C57BL/6 mice. After vaccinating with live attenuated Y. pestis and challenging intranasally with virulent plague, nearly 20% of pulmonary CD8 T cells recognize this single, highly conserved Ag. Moreover, immunizing mice with a single peptide, YopE(69-77), suffices to confer significant protection from lethal pulmonary challenge. These findings suggest YopE could be a valuable addition to subunit plague vaccines and provide a new animal model in which sensitive, pathogen-specific assays can be used to study CD8 T cell-mediated defense against acutely lethal bacterial infections of the lung.
Collapse
|
45
|
Bahta M, Lountos GT, Dyas B, Kim SE, Ulrich RG, Waugh DS, Burke TR. Utilization of nitrophenylphosphates and oxime-based ligation for the development of nanomolar affinity inhibitors of the Yersinia pestis outer protein H (YopH) phosphatase. J Med Chem 2011; 54:2933-43. [PMID: 21443195 PMCID: PMC3085962 DOI: 10.1021/jm200022g] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our current study reports the first K(M) optimization of a library of nitrophenylphosphate-containing substrates for generating an inhibitor lead against the Yersinia pestis outer protein phosphatase (YopH). A high activity substrate identified by this method (K(M) = 80 μM) was converted from a substrate into an inhibitor by replacement of its phosphate group with difluoromethylphosphonic acid and by attachment of an aminooxy handle for further structural optimization by oxime ligation. A cocrystal structure of this aminooxy-containing platform in complex with YopH allowed the identification of a conserved water molecule proximal to the aminooxy group that was subsequently employed for the design of furanyl-based oxime derivatives. By this process, a potent (IC(50) = 190 nM) and nonpromiscuous inhibitor was developed with good YopH selectivity relative to a panel of phosphatases. The inhibitor showed significant inhibition of intracellular Y. pestis replication at a noncytotoxic concentration. The current work presents general approaches to PTP inhibitor development that may be useful beyond YopH.
Collapse
Affiliation(s)
- Medhanit Bahta
- Chemical Biology Laboratory, Molecular Discovery Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NCI-Frederick, Frederick, MD 21702, U.S.A
| | - George T. Lountos
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NCI-Frederick, Frederick, MD 21702, U.S.A
| | - Beverly Dyas
- Laboratory of Molecular Immunology, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702, U.S.A
| | - Sung-Eun Kim
- Chemical Biology Laboratory, Molecular Discovery Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NCI-Frederick, Frederick, MD 21702, U.S.A
| | - Robert G. Ulrich
- Laboratory of Molecular Immunology, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702, U.S.A
| | - David S. Waugh
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NCI-Frederick, Frederick, MD 21702, U.S.A
| | - Terrence R. Burke
- Chemical Biology Laboratory, Molecular Discovery Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NCI-Frederick, Frederick, MD 21702, U.S.A
| |
Collapse
|
46
|
Bozue J, Mou S, Moody KL, Cote CK, Trevino S, Fritz D, Worsham P. The role of the phoPQ operon in the pathogenesis of the fully virulent CO92 strain of Yersinia pestis and the IP32953 strain of Yersinia pseudotuberculosis. Microb Pathog 2011; 50:314-21. [PMID: 21320584 DOI: 10.1016/j.micpath.2011.02.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 01/31/2011] [Accepted: 02/04/2011] [Indexed: 11/30/2022]
Abstract
At the genomic level, Yersinia pestis and Yersinia pseudotuberculosis are nearly identical but cause very different diseases. Y. pestis is the etiologic agent of plague; whereas Y. pseudotuberculosis causes a gastrointestinal infection primarily after the consumption of contaminated food. In many gram-negative pathogenic bacteria, PhoP is part of a two-component global regulatory system in which PhoQ serves as the sensor kinase, and PhoP is the response regulator. PhoP is known to activate a number of genes in many bacteria related to virulence. To determine the role of the PhoPQ proteins in Yersinia infections, primarily using aerosol challenge models, the phoP gene was deleted from the chromosome of the CO92 strain of Y. pestis and the IP32953 strain of Y. pseudotuberculosis, leading to a polar mutation of the phoPQ operon. We demonstrated that loss of phoPQ from both strains leads to a defect in intracellular growth and/or survival within macrophages. These in vitro data would suggest that the phoPQ mutants would be attenuated in vivo. However, the LD(50) for the Y. pestis mutant did not differ from the calculated LD(50) for the wild-type CO92 strain for either the bubonic or pneumonic murine models of infection. In contrast, mice challenged by aerosol with the Y. pseudotuberculosis mutant had a LD(50) value 40× higher than the wild-type strain. These results demonstrate that phoPQ are necessary for full virulence by aerosol infection with the IP32953 strain of Y. pseudotuberculosis. However, the PhoPQ proteins do not play a significant role in infection with a fully virulent strain of Y. pestis.
Collapse
Affiliation(s)
- Joel Bozue
- Bacteriology Division, The United States Army of Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD 21702, United States.
| | | | | | | | | | | | | |
Collapse
|
47
|
Do Y, Koh H, Park CG, Dudziak D, Seo P, Mehandru S, Choi JH, Cheong C, Park S, Perlin DS, Powell BS, Steinman RM. Targeting of LcrV virulence protein from Yersinia pestis to dendritic cells protects mice against pneumonic plague. Eur J Immunol 2010; 40:2791-6. [PMID: 20812236 DOI: 10.1002/eji.201040511] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To help design needed new vaccines for pneumonic plague, we targeted the Yersinia pestis LcrV protein directly to CD8α(+) DEC-205(+) or CD8α(-) DCIR2(+) DC along with a clinically feasible adjuvant, poly IC. By studying Y. pestis in mice, we could evaluate the capacity of this targeting approach to protect against a human pathogen. The DEC-targeted LcrV induced polarized Th1 immunity, whereas DCIR2-targeted LcrV induced fewer CD4(+) T cells secreting IFN-γ, but higher IL-4, IL-5, IL-10, and IL-13 production. DCIR-2 targeting elicited higher anti-LcrV Ab titers than DEC targeting, which were comparable to a protein vaccine given in alhydrogel adjuvant, but the latter did not induce detectable T-cell immunity. When DEC- and DCIR2-targeted and F1-V+ alhydrogel-vaccinated mice were challenged 6 wk after vaccination with the virulent CO92 Y. pestis, the protection level and Ab titers induced by DCIR2 targeting were similar to those induced by F1-V protein with alhydrogel vaccination. Therefore, LcrV targeting to DC elicits combined humoral and cellular immunity, and for the first time with this approach, also induces protection in a mouse model for a human pathogen.
Collapse
Affiliation(s)
- Yoonkyung Do
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center, The Rockefeller University, New York, NY 10065-6399, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Qiu Y, Liu Y, Qi Z, Wang W, Kou Z, Zhang Q, Liu G, Liu T, Yang Y, Yang X, Xin Y, Li C, Cui B, Huang S, Liu H, Zeng L, Wang Z, Yang R, Wang H, Wang X. Comparison of Immunological Responses of Plague Vaccines F1 + rV270 and EV76 in Chinese-Origin Rhesus Macaque, Macaca mulatta. Scand J Immunol 2010; 72:425-33. [DOI: 10.1111/j.1365-3083.2010.02456.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
49
|
Fetherston JD, Kirillina O, Bobrov AG, Paulley JT, Perry RD. The yersiniabactin transport system is critical for the pathogenesis of bubonic and pneumonic plague. Infect Immun 2010; 78:2045-52. [PMID: 20160020 PMCID: PMC2863531 DOI: 10.1128/iai.01236-09] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 11/30/2009] [Accepted: 02/04/2010] [Indexed: 01/18/2023] Open
Abstract
Iron acquisition from the host is an important step in the pathogenic process. While Yersinia pestis has multiple iron transporters, the yersiniabactin (Ybt) siderophore-dependent system plays a major role in iron acquisition in vitro and in vivo. In this study, we determined that the Ybt system is required for the use of iron bound by transferrin and lactoferrin and examined the importance of the Ybt system for virulence in mouse models of bubonic and pneumonic plague. Y. pestis mutants unable to either transport Ybt or synthesize the siderophore were both essentially avirulent via subcutaneous injection (bubonic plague model). Surprisingly, via intranasal instillation (pneumonic plague model), we saw a difference in the virulence of Ybt biosynthetic and transport mutants. Ybt biosynthetic mutants displayed an approximately 24-fold-higher 50% lethal dose (LD(50)) than transport mutants. In contrast, under iron-restricted conditions in vitro, a Ybt transport mutant had a more severe growth defect than the Ybt biosynthetic mutant. Finally, a Delta pgm mutant had a greater loss of virulence than the Ybt biosynthetic mutant, indicating that the 102-kb pgm locus encodes a virulence factor, in addition to Ybt, that plays a role in the pathogenesis of pneumonic plague.
Collapse
Affiliation(s)
- Jacqueline D. Fetherston
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - Olga Kirillina
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - Alexander G. Bobrov
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - James T. Paulley
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - Robert D. Perry
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| |
Collapse
|
50
|
Abstract
The potential application of Yersinia pestis for bioterrorism emphasizes the urgent need to develop more effective vaccines against airborne infection. The current status of plague vaccines has been reviewed. The present emphasis is on subunit vaccines based on the F1 and LcrV antigens. These provide good protection in animal models but may not protect against F1 strains with modifications to the type III secretion system. The duration of protection against pneumonic infection is also uncertain. Other strategies under investigation include defined live-attenuated vaccines, DNA vaccines, mucosal delivery systems and heterologous immunization. The live-attenuated strain Y. pestis EV NIIEG protects against aerosol challenge in animal models and, with further modification to reduce residual virulence and to optimize respiratory protection, it could provide a shortcut to improved vaccines. The regulatory problems inherent in licensing vaccines for which efficacy data are unavailable and their possible solutions are discussed herein.
Collapse
Affiliation(s)
- Valentina A Feodorova
- Scientific and Research Institute for Medical and Veterinary Biotechnologies, Russia-Switzerland, Branch in Saratov, 9 Proviantskaya Street, Box 1580, Saratov 410028, Russia.
| | | |
Collapse
|