1
|
Elia U, Levy Y, Cohen H, Zauberman A, Gur D, Hazan-Halevy I, Aftalion M, Benarroch S, Bar-Haim E, Redy-Keisar O, Cohen O, Peer D, Mamroud E. Novel Bivalent mRNA-LNP Vaccine for Highly Effective Protection against Pneumonic Plague. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501286. [PMID: 40279638 DOI: 10.1002/advs.202501286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/19/2025] [Indexed: 04/27/2025]
Abstract
Yersinia pestis, the causative agent of plague, remains a significant global health hazard and a potential top-tier biothreat despite modern medical advances. Here, two mRNA constructs encoding different versions of the low-calcium response virulence (LcrV) protective antigen, an essential virulence factor of Y. pestis, are designed and evaluated. Next, the immunogenicity and protective efficacy both independently and in combination is assessed with the previously reported F1-encoding mRNA construct in the well-established mouse model of pneumonic plague. The findings reveal that human Fc-conjugated F1 + LcrV combination mRNA vaccination resulted in significant immune activation and substantial protection against intranasal Y. pestis challenge. Notably, the combined vaccine demonstrates protective efficacy against two highly virulent wild-type Y. pestis strains representing distinct biovars and an atypical, unencapsulated strain. This study represents the first comprehensive evaluation of mRNA constructs encoding innovatively designed versions of LcrV and F1 for pneumonic plague prevention, addressing critical gaps in current vaccination approaches. This study establishes the mRNA-lipid nanoparticle (LNP) platform as a promising tool for addressing bacterial pathogens, including those resistant to antibiotics. By broadening its applicability to diverse threats, this technology represents an innovative approach to tackling some of the most pressing challenges in global health.
Collapse
Affiliation(s)
- Uri Elia
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Yinon Levy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Hila Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Ayelet Zauberman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - David Gur
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Shani Benarroch
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Orit Redy-Keisar
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Ofer Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Emanuelle Mamroud
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, 76100, Israel
| |
Collapse
|
2
|
Tchalla EYI, Betadpur A, Khalil AY, Bhalla M, Bou Ghanem EN. Sex-based difference in immune responses and efficacy of the pneumococcal conjugate vaccine. J Leukoc Biol 2024; 117:qiae177. [PMID: 39141715 PMCID: PMC11684992 DOI: 10.1093/jleuko/qiae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024] Open
Abstract
Vaccine-mediated protection and susceptibility to Streptococcus pneumoniae (pneumococcus) infections are influenced by biological sex. The incidence of invasive pneumococcal disease remains higher in males compared to females even after the introduction of the pneumococcal conjugate vaccine. However, sex-based differences in the immune response to this conjugate vaccine remain unexplored. To investigate those differences, we vaccinated adult male and female mice with pneumococcal conjugate vaccine and assessed cellular and humoral immune responses. Compared to females, male mice displayed lower levels of T follicular helper cells, germinal center B cells, and plasmablasts, which are all required for antibody production following vaccination. This was linked to lower IgG and IgM levels against pneumococci and lower isotype switching to IgG3 in vaccinated males. Due to lower antibody levels, sera of vaccinated male mice had lower efficacy in several anti-pneumococcal functions, including neutralization of bacterial binding to pulmonary epithelial cells as well as direct cytotoxicity against S. pneumoniae. Importantly, while the vaccine was highly protective in females, vaccinated males succumbed to infection more readily and were more susceptible to both lung-localized infection and systemic spread following S. pneumoniae challenge. These findings identify sex-based differences in immune responses to pneumococcal conjugate vaccine that can inform future vaccine strategies.
Collapse
Affiliation(s)
- Essi Y I Tchalla
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Anagha Betadpur
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Andrew Y Khalil
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Elsa N Bou Ghanem
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| |
Collapse
|
3
|
Hendrix EK, Sha J, Kilgore PB, Neil BH, Verma AK, Chopra AK. The Protective Effect of IL-17A in Pneumonic Plague Can Be Compensated by Effective Vaccines and Immunization Strategies in Mice. Vaccines (Basel) 2024; 12:1361. [PMID: 39772023 PMCID: PMC11680114 DOI: 10.3390/vaccines12121361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Plague, caused by Yersinia pestis, poses a public health threat not only due to sporadic outbreaks across the globe but also due to its potential as a biothreat agent. Ironically, among the seven deadliest pandemics in global history, three were caused by Y. pestis. Pneumonic plague, the more contagious and severe form of the disease, is difficult to contain, requiring either prophylactic antibiotic treatment or vaccination. However, no vaccine (live attenuated or subunit) is currently approved by the Food and Drug Administration, requiring rigorous preclinical studies in different animal models, thus forming the basis of this study. Objectives: The aim of this study was to evaluate the efficacy and immune responses of two live attenuated vaccines (LAVs), LMA and LMP, either alone or in combination with a trivalent adenoviral vector-based vaccine (Ad5-YFV), in IL-17A-depleted and IgG control mice by using an anti-IL-17A monoclonal antibody (mAb) or its matched isotype IgG, respectively. Methods: IL-17A mAb or IgG isotype control was administered to mice twice per week to their respective groups during the course of immunization. Serum, spleens, and broncho-alveolar lavage fluid (BALF) were collected for assessing immunological responses, and another cohort of mice was intranasally challenged with a lethal dose of parental Y. pestis CO92. Results: Robust humoral and cellular immune responses followed by complete protection were observed in all vaccinated animals against highly lethal intranasal challenge doses of parental Y. pestis CO92. Serum IgG titers to YscF and overall mucosal IgA titers to all three antigens of the Ad5-YFV vaccine were significantly lower, with slightly reduced serum LcrV-neutralizing antibodies when IL-17A was depleted compared to IgG control animals during the course of immunization. A remarkable reduction in Th1 (IFNγ or IL-2) and Th17 cell populations was observed in IL-17A-depleted mice compared to IgG controls in response to vaccination. On the other hand, B cell activities in germinal centers, overall activated antigen-specific T cells, and memory B and T cells remained at comparable levels in both vaccinated IL-17A-depleted and IgG control mice. Conclusions: These data demonstrated the effectiveness of our vaccines even under the reduced levels of both Th1 and Th17 responses and thus should be suitable for those individuals associated with certain immune deficiencies.
Collapse
Affiliation(s)
- Emily K. Hendrix
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Jian Sha
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Paul B. Kilgore
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Blake H. Neil
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Atul K. Verma
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Ashok K. Chopra
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Biodefense and Emerging Infectious Diseases, Galveston, TX 77555, USA
| |
Collapse
|
4
|
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
|
5
|
Price SL, Oakes RS, Gonzalez RJ, Edwards C, Brady A, DeMarco JK, von Andrian UH, Jewell CM, Lawrenz MB. Microneedle array delivery of Yersinia pestis recapitulates bubonic plague. iScience 2024; 27:108600. [PMID: 38179062 PMCID: PMC10765063 DOI: 10.1016/j.isci.2023.108600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/25/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024] Open
Abstract
Fleas transmit Yersinia pestis directly within the dermis of mammals to cause bubonic plague. Syringe-mediated inoculation is widely used to recapitulate bubonic plague and study Y. pestis pathogenesis. However, intradermal needle inoculation is tedious, error prone, and poses a significant safety risk for laboratorians. Microneedle arrays (MNAs) are micron-scale polymeric structures that deliver materials to the dermis, while minimizing the risk of needle sticks. We demonstrated that MNA inoculation is a viable strategy to recapitulate bubonic plague and study bacterial virulence by defining the parameters needed to establish a lethal infection in the mouse model and characterizing the course of infection using live-animal optical imaging. Using MNAs, we also demonstrated that Y. pestis must overcome calprotectin-mediated zinc restriction within the dermis and dermal delivery of an attenuated mutant has vaccine potential. Together, these data demonstrate that MNAs are a safe alternative to study Y. pestis pathogenesis in the laboratory.
Collapse
Affiliation(s)
- Sarah L. Price
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Robert S. Oakes
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Rodrigo J. Gonzalez
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Camilla Edwards
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Amanda Brady
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jennifer K. DeMarco
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Ulrich H. von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD 20742, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical School, Baltimore, MD 21201, USA
| | - Matthew B. Lawrenz
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
6
|
Price SL, Thibault D, Garrison TM, Brady A, Guo H, Kehl‐Fie TE, Garneau‐Tsodikova S, Perry RD, van Opijnen T, Lawrenz MB. Droplet Tn-Seq identifies the primary secretion mechanism for yersiniabactin in Yersinia pestis. EMBO Rep 2023; 24:e57369. [PMID: 37501563 PMCID: PMC10561177 DOI: 10.15252/embr.202357369] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Nutritional immunity includes sequestration of transition metals from invading pathogens. Yersinia pestis overcomes nutritional immunity by secreting yersiniabactin to acquire iron and zinc during infection. While the mechanisms for yersiniabactin synthesis and import are well-defined, those responsible for yersiniabactin secretion are unknown. Identification of this mechanism has been difficult because conventional mutagenesis approaches are unable to inhibit trans-complementation by secreted factors between mutants. To overcome this obstacle, we utilized a technique called droplet Tn-seq (dTn-seq), which uses microfluidics to isolate individual transposon mutants in oil droplets, eliminating trans-complementation between bacteria. Using this approach, we first demonstrated the applicability of dTn-seq to identify genes with secreted functions. We then applied dTn-seq to identify an AcrAB efflux system as required for growth in metal-limited conditions. Finally, we showed this efflux system is the primary yersiniabactin secretion mechanism and required for virulence during bubonic and pneumonic plague. Together, these studies have revealed the yersiniabactin secretion mechanism that has eluded researchers for over 30 years and identified a potential therapeutic target for bacteria that use yersiniabactin for metal acquisition.
Collapse
Affiliation(s)
- Sarah L Price
- Department of Microbiology and ImmunologyUniversity of LouisvilleLouisvilleKYUSA
| | | | - Taylor M Garrison
- Department of Microbiology and ImmunologyUniversity of LouisvilleLouisvilleKYUSA
| | - Amanda Brady
- Department of Microbiology and ImmunologyUniversity of LouisvilleLouisvilleKYUSA
| | - Haixun Guo
- Center for Predictive Medicine for Biodefense and Emerging Infectious DiseasesUniversity of LouisvilleLouisvilleKYUSA
- Department of RadiologyUniversity of LouisvilleLouisvilleKYUSA
| | - Thomas E Kehl‐Fie
- Department of MicrobiologyUniversity of Illinois Urbana‐ChampaignChampaignILUSA
- Carl R Woese Institute for Genomic BiologyUrbanaILUSA
| | | | - Robert D Perry
- Department of Microbiology, Immunology and Molecular GeneticsUniversity of KentuckyLexingtonKYUSA
| | | | - Matthew B Lawrenz
- Department of Microbiology and ImmunologyUniversity of LouisvilleLouisvilleKYUSA
- Center for Predictive Medicine for Biodefense and Emerging Infectious DiseasesUniversity of LouisvilleLouisvilleKYUSA
| |
Collapse
|
7
|
Patnaik A, Rai SK, Dhaked RK. Recent Advancements and Novel Approaches Contributing to the Present Arsenal of Prophylaxis and Treatment Strategies Against Category A Bacterial Biothreat Agents. Indian J Microbiol 2023; 63:161-172. [PMID: 37325016 PMCID: PMC10220334 DOI: 10.1007/s12088-023-01075-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/04/2023] [Indexed: 06/17/2023] Open
Abstract
Bacterial pathogens have always been a part of the ecosystem in which we thrive. Some pathogens have caused deadly outbreaks in the past and have been exploited as an agent of threat. Natural hotspots for these biological pathogens are widely distributed throughout the world and hence they remain clinically important. Technological advancement and change in general lifestyle has driven the evolution of these pathogens into more virulent and resistant variants. There has been a growing concern over the development of multidrug-resistant bacterial strains that could be used as bioweapons. This rapid change in pathogens also propels the field of science to develop and innovate new strategies and methodologies which are superior and safer to the existing ones. Some bacterial agents like-Bacillus anthracis, Yersinia pestis, Francisella tularensis and toxins produced by strains of Clostridium botulinum, have been segregated as Category A substances as they pose imminent threat to public health with a history of life threatening and catastrophic disease. This review highlights some encouraging developments and value additions in the current plan of action for protection against these select biothreat bacterial pathogens.
Collapse
Affiliation(s)
- Abhinandan Patnaik
- Biotechnology Division, Defence Research and Development Establishment, Jhansi Road, Gwalior, MP 474002 India
| | - Sharad Kumar Rai
- Biotechnology Division, Defence Research and Development Establishment, Jhansi Road, Gwalior, MP 474002 India
| | - Ram Kumar Dhaked
- Biotechnology Division, Defence Research and Development Establishment, Jhansi Road, Gwalior, MP 474002 India
| |
Collapse
|
8
|
Park KS, Svennerholm K, Crescitelli R, Lässer C, Gribonika I, Andersson M, Boström J, Alalam H, Harandi AM, Farewell A, Lötvall J. Detoxified synthetic bacterial membrane vesicles as a vaccine platform against bacteria and SARS-CoV-2. J Nanobiotechnology 2023; 21:156. [PMID: 37208676 DOI: 10.1186/s12951-023-01928-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/13/2023] [Indexed: 05/21/2023] Open
Abstract
The development of vaccines based on outer membrane vesicles (OMV) that naturally bud off from bacteria is an evolving field in infectious diseases. However, the inherent inflammatory nature of OMV limits their use as human vaccines. This study employed an engineered vesicle technology to develop synthetic bacterial vesicles (SyBV) that activate the immune system without the severe immunotoxicity of OMV. SyBV were generated from bacterial membranes through treatment with detergent and ionic stress. SyBV induced less inflammatory responses in macrophages and in mice compared to natural OMV. Immunization with SyBV or OMV induced comparable antigen-specific adaptive immunity. Specifically, immunization with Pseudomonas aeruginosa-derived SyBV protected mice against bacterial challenge, and this was accompanied by significant reduction in lung cell infiltration and inflammatory cytokines. Further, immunization with Escherichia coli-derived SyBV protected mice against E. coli sepsis, comparable to OMV-immunized group. The protective activity of SyBV was driven by the stimulation of B-cell and T-cell immunity. Also, SyBV were engineered to display the SARS-CoV-2 S1 protein on their surface, and these vesicles induced specific S1 protein antibody and T-cell responses. Collectively, these results demonstrate that SyBV may be a safe and efficient vaccine platform for the prevention of bacterial and viral infections.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Kristina Svennerholm
- Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rossella Crescitelli
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Inta Gribonika
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mickael Andersson
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Jonas Boström
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Hanna Alalam
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- BC Children's Hospital Research Institute, Vaccine Evaluation Center, University of British Columbia, Columbia, Canada
| | - Anne Farewell
- Department of Chemistry and Molecular Biology, Centre for Antibiotic Resistance, University of Gothenburg, Gothenburg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
9
|
Galloway DR, Nguyen NX, Li J, Houston N, Gregersen G, Williamson ED, Falkenberg FW, Herron JN, Hale JS. The magnitude of the germinal center B cell and T follicular helper cell response predicts long-lasting antibody titers to plague vaccination. Front Immunol 2022; 13:1017385. [PMID: 36389793 PMCID: PMC9650111 DOI: 10.3389/fimmu.2022.1017385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/13/2022] [Indexed: 11/27/2022] Open
Abstract
The development of a safe and effective vaccine against Yersinia pestis, the causative organism for plague disease, remains an important global health priority. Studies have demonstrated effective immune-based protection against plague challenge that is induced by plague antigen subunit vaccination in an aqueous alhydrogel formulation; however, whether these candidate vaccines in this formulation and presentation, induce long-lasting immunological memory in the form of durable cellular and antibody recall responses has not been fully demonstrated. In this study, we analyzed germinal center T follicular helper and germinal center B cell responses following F1V and F1 + V plague subunit immunization of mice with vaccines formulated in various adjuvants. Our data demonstrate that recombinant plague protein immunization formulated with IL-2/GM-CSF cytokines bound to alhydrogel adjuvant drive an increase in the magnitude of the germinal center T follicular helper and germinal center B cell responses following primary immunization, compared to vaccines formulated with Alhydrogel adjuvant alone. In contrast, plague protein subunit immunization combined with CpG ODN bound to alhydrogel increased the magnitude and duration of the germinal center Tfh and B cell responses following booster immunization. Importantly, enhanced germinal center Tfh and B cell responses correlated with long-lasting and high F1V-specific antibody titers and more robust antibody recall responses to F1V re-exposure. These findings indicate that vaccine formulations that drive enhancement of the germinal center Tfh and B cell responses are critical for inducing durable plague-specific humoral immunity.
Collapse
Affiliation(s)
- Darrell R. Galloway
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - Nguyen X. Nguyen
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - Jiahui Li
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - Nicholas Houston
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - Gage Gregersen
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - E. Diane Williamson
- Chemical Biological Radiological Division, Defense Science and Technology Laboratory (DSTL) Porton Down, Salisbury, United Kingdom
| | | | - James N. Herron
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - J. Scott Hale
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
10
|
BAP31 affects macrophage polarization through regulating helper T cells activation. J Mol Histol 2022; 53:843-855. [PMID: 36018529 DOI: 10.1007/s10735-022-10095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/28/2022] [Indexed: 10/15/2022]
Abstract
Previously, we reported that B cell receptor associated protein 31 (BAP31) is a positive regulator on T-cells activation. Helper T cells [cluster of differentiation 4+ (CD4+) T cells] can regulate macrophage activation in adaptive immune response against pathogens. In this study, we elucidate that M1 and M2 macrophages polarization is significantly suppressed in Lck Cre-BAP31flox/flox mice or the co-culture system of CD4+ T cells from Lck Cre-BAP31flox/flox mice and macrophages from WT mice. It means that BAP31 may affect the regulation of CD4+ T cells on macrophages. Further studies suggest that BAP31 deficiency significantly reduce the expressions of T helper 1 (Th1)/ Th2/ Th17/ Th9/ Th22/ Treg cells-related cytokines and transcription factors. The inhibition of macrophages activation caused by BAP31 knockdown is due to the reduction of IFN-γ and IL-4 secreted by Th1 and Th2 cells. BAP31 also affects the levels of early activation markers (CD69 and CD25) of CD4+ T cells. Moreover, BAP31 deficiency downregulates the expression of TCRαβ-CD3 complex, and the adaptor proteins p-Zap70, p-Lck, and p-Lat in TCR signaling pathway. These results demonstrate that BAP31 deficiency inhibits TCR/CD3-mediated activation in CD4+ T cells and adversely affects macrophages polarization. These findings establish a theoretical foundation for the study of BAP31 in immunotherapy.
Collapse
|
11
|
Zhang W, Song X, Zhai L, Guo J, Zheng X, Zhang L, Lv M, Hu L, Zhou D, Xiong X, Yang W. Complete Protection Against Yersinia pestis in BALB/c Mouse Model Elicited by Immunization With Inhalable Formulations of rF1-V10 Fusion Protein via Aerosolized Intratracheal Inoculation. Front Immunol 2022; 13:793382. [PMID: 35154110 PMCID: PMC8825376 DOI: 10.3389/fimmu.2022.793382] [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: 10/12/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022] Open
Abstract
Pneumonic plague, caused by Yersinia pestis, is an infectious disease with high mortality rates unless treated early with antibiotics. Currently, no FDA-approved vaccine against plague is available for human use. The capsular antigen F1, the low-calcium-response V antigen (LcrV), and the recombinant fusion protein (rF1-LcrV) of Y. pestis are leading subunit vaccine candidates under intense investigation; however, the inability of recombinant antigens to provide complete protection against pneumonic plague in animal models remains a significant concern. In this study, we compared immunoprotection against pneumonic plague provided by rF1, rV10 (a truncation of LcrV), and rF1-V10, and vaccinations delivered via aerosolized intratracheal (i.t.) inoculation or subcutaneous (s.c.) injection. We further considered three vaccine formulations: conventional liquid, dry powder produced by spray freeze drying, or dry powder reconstituted in PBS. The main findings are: (i) rF1-V10 immunization with any formulation via i.t. or s.c. routes conferred 100% protection against Y. pestis i.t. infection; (ii) rF1 or rV10 immunization using i.t. delivery provided significantly stronger protection than rF1 or rV10 immunization via s.c. delivery; and (iii) powder formulations of subunit vaccines induced immune responses and provided protection equivalent to those elicited by unprocessed liquid formulations of vaccines. Our data indicate that immunization with a powder formulation of rF1-V10 vaccines via an i.t. route may be a promising vaccination strategy for providing protective immunity against pneumonic plague.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaolin Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lina Zhai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jianshu Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xinying Zheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lili Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Meng Lv
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lingfei Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Dongsheng Zhou
- 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
| | - Wenhui Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
12
|
Price SL, Vadyvaloo V, DeMarco JK, Brady A, Gray PA, Kehl-Fie TE, Garneau-Tsodikova S, Perry RD, Lawrenz MB. Yersiniabactin contributes to overcoming zinc restriction during Yersinia pestis infection of mammalian and insect hosts. Proc Natl Acad Sci U S A 2021; 118:e2104073118. [PMID: 34716262 PMCID: PMC8612365 DOI: 10.1073/pnas.2104073118] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 09/09/2021] [Indexed: 02/04/2023] Open
Abstract
Yersinia pestis causes human plague and colonizes both a mammalian host and a flea vector during its transmission cycle. A key barrier to bacterial infection is the host's ability to actively sequester key biometals (e.g., iron, zinc, and manganese) required for bacterial growth. This is referred to as nutritional immunity. Mechanisms to overcome nutritional immunity are essential virulence factors for bacterial pathogens. Y. pestis produces an iron-scavenging siderophore called yersiniabactin (Ybt) that is required to overcome iron-mediated nutritional immunity and cause lethal infection. Recently, Ybt has been shown to bind to zinc, and in the absence of the zinc transporter ZnuABC, Ybt improves Y. pestis growth in zinc-limited medium. These data suggest that, in addition to iron acquisition, Ybt may also contribute to overcoming zinc-mediated nutritional immunity. To test this hypothesis, we used a mouse model defective in iron-mediated nutritional immunity to demonstrate that Ybt contributes to virulence in an iron-independent manner. Furthermore, using a combination of bacterial mutants and mice defective in zinc-mediated nutritional immunity, we identified calprotectin as the primary barrier for Y. pestis to acquire zinc during infection and that Y. pestis uses Ybt to compete with calprotectin for zinc. Finally, we discovered that Y. pestis encounters zinc limitation within the flea midgut, and Ybt contributes to overcoming this limitation. Together, these results demonstrate that Ybt is a bona fide zinc acquisition mechanism used by Y. pestis to surmount zinc limitation during the infection of both the mammalian and insect hosts.
Collapse
Affiliation(s)
- Sarah L Price
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202
| | - Viveka Vadyvaloo
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA 99164
| | - Jennifer K DeMarco
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY 40292
| | - Amanda Brady
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202
| | - Phoenix A Gray
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202
| | - Thomas E Kehl-Fie
- Department of Microbiology and Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Champaign, IL 61820
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY 40536
| | - Robert D Perry
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky School of Medicine, Lexington, KY 40506
| | - Matthew B Lawrenz
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202;
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY 40292
| |
Collapse
|
13
|
Biryukov S, Dankmeyer JL, Shamsuddin Z, Velez I, Rill NO, Rosario-Acevedo R, Klimko CP, Shoe JL, Hunter M, Ward MD, Cazares LH, Fetterer DP, Bozue JA, Worsham PL, Cote CK, Amemiya K. Impact of Toll-Like Receptor-Specific Agonists on the Host Immune Response to the Yersinia pestis Plague rF1V Vaccine. Front Immunol 2021; 12:726416. [PMID: 34512658 PMCID: PMC8430260 DOI: 10.3389/fimmu.2021.726416] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
Relatively recent advances in plague vaccinology have produced the recombinant fusion protein F1-V plague vaccine. This vaccine has been shown to readily protect mice from both bubonic and pneumonic plague. The protection afforded by this vaccine is solely based upon the immune response elicited by the F1 or V epitopes expressed on the F1-V fusion protein. Accordingly, questions remain surrounding its efficacy against infection with non-encapsulated (F1-negative) strains. In an attempt to further optimize the F1-V elicited immune response and address efficacy concerns, we examined the inclusion of multiple toll-like receptor agonists into vaccine regimens. We examined the resulting immune responses and also any protection afforded to mice that were exposed to aerosolized Yersinia pestis. Our data demonstrate that it is possible to further augment the F1-V vaccine strategy in order to optimize and augment vaccine efficacy.
Collapse
Affiliation(s)
- Sergei Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Zain Shamsuddin
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Ivan Velez
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Nathaniel O. Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Raysa Rosario-Acevedo
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jennifer L. Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Michael D. Ward
- Molecular Biology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Lisa H. Cazares
- Molecular Biology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - David P. Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Joel A. Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Patricia L. Worsham
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Kei Amemiya
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| |
Collapse
|
14
|
D'Arco C, McCormick AA, Arnaboldi PM. Single-dose intranasal subunit vaccine rapidly clears secondary sepsis in a high-dose pneumonic plague infection. Vaccine 2021; 39:1435-1444. [PMID: 33531196 DOI: 10.1016/j.vaccine.2021.01.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023]
Abstract
Yersinia pestis, the causative agent of plague, has killed millions throughout human history. Though public health initiatives have reduced the number of plague cases, it remains endemic in many areas of the world. It also remains a significant threat for use as a biological weapon. Naturally occurring multi-drug antibiotic resistance has been observed in Y. pestis, and resistant strains have been engineered for use as a biological weapon. Vaccines represent our best means of protection against the threat of antibiotic resistant plague. We have developed a vaccine consisting of two Y. pestis virulence factors, LcrV (V) and F1, conjugated to Tobacco Mosaic Virus (TMV), a safe, non-replicating plant virus that can be administered mucosally, providing complete protection against pneumonic plague, the deadliest form of the disease and the one most likely to be seen in a biological attack. A single intranasal (i.n.) dose of TMV-F1 + TMV-V (TMV-F1/V) protected 88% of mice against lethal challenge with 100 LD50 of Y. pestis CO92pgm-, while immunization with rF1 + rV without TMV was not protective. Serum and tissues were collected at various timepoints after challenge to assess bacterial clearance, histopathology, cytokine production, and antibody production. Overall, TMV-F1/V immunized mice showed a significant reduction in histopathology, bacterial burden, and inflammatory cytokine production following challenge compared to rF1 + rV vaccinated and unvaccinated mice. Pneumonic challenge resulted in systemic dissemination of the bacteria in all groups, but only TMV-F1/V immunized mice rapidly cleared bacteria from the spleen and liver. There was a direct correlation between pre-challenge serum F1 titers and recovery in all immunized mice, strongly suggesting a role for antibody in the neutralization and/or opsonization of Y. pestis in this model. Mucosal administration of a single dose of a Y. pestis TMV-based subunit vaccine, without any additional adjuvant, can effectively protect mice from lethal infection.
Collapse
Affiliation(s)
- Christina D'Arco
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, United States
| | - Alison A McCormick
- Department of Biology and Pharmaceutical Sciences, College of Pharmacy, Touro University California, Vallejo, CA 94592, United States
| | - Paul M Arnaboldi
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, United States; Biopeptides, Corp., East Setauket, NY 11733, United States.
| |
Collapse
|
15
|
Kilgore PB, Sha J, Andersson JA, Motin VL, Chopra AK. A new generation needle- and adjuvant-free trivalent plague vaccine utilizing adenovirus-5 nanoparticle platform. NPJ Vaccines 2021; 6:21. [PMID: 33514747 PMCID: PMC7846801 DOI: 10.1038/s41541-020-00275-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
A plague vaccine with a fusion cassette of YscF, F1, and LcrV encoding genes in an adenovirus-5 vector (rAd5-YFV) is evaluated for efficacy and immune responses in mice. Two doses of the vaccine provides 100% protection when administered intranasally against challenge with Yersinia pestis CO92 or its isogenic F1 mutant in short- or long- term immunization in pneumonic/bubonic plague models. The corresponding protection rates drop in rAd5-LcrV monovalent vaccinated mice in plague models. The rAd5-YFV vaccine induces superior humoral, mucosal and cell-mediated immunity, with clearance of the pathogen. Immunization of mice with rAd5-YFV followed by CO92 infection dampens proinflammatory cytokines and neutrophil chemoattractant production, while increasing Th1- and Th2-cytokine responses as well as macrophage/monocyte chemo-attractants when compared to the challenge control animals. This is a first study showing complete protection of mice from pneumonic/bubonic plague with a viral vector-based vaccine without the use of needles and the adjuvant.
Collapse
Affiliation(s)
- Paul B. Kilgore
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA
| | - Jian Sha
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| | - Jourdan A. Andersson
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA
| | - Vladimir L. Motin
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Department of Pathology, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX USA
| | - Ashok K. Chopra
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX USA
| |
Collapse
|
16
|
Byvalov AA, Konyshev IV, Uversky VN, Dentovskaya SV, Anisimov AP. Yersinia Outer Membrane Vesicles as Potential Vaccine Candidates in Protecting against Plague. Biomolecules 2020; 10:E1694. [PMID: 33353123 PMCID: PMC7766529 DOI: 10.3390/biom10121694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/03/2020] [Accepted: 12/16/2020] [Indexed: 11/18/2022] Open
Abstract
Despite the relatively low incidence of plague, its etiological agent, Yersinia pestis, is an exceptional epidemic danger due to the high infectivity and mortality of this infectious disease. Reports on the isolation of drug-resistant Y. pestis strains indicate the advisability of using asymmetric responses, such as phage therapy and vaccine prophylaxis in the fight against this problem. The current relatively effective live plague vaccine is not approved for use in most countries because of its ability to cause heavy local and system reactions and even a generalized infectious process in people with a repressed immune status or metabolic disorders, as well as lethal infection in some species of nonhuman primates. Therefore, developing alternative vaccines is of high priority and importance. However, until now, work on the development of plague vaccines has mainly focused on screening for the potential immunogens. Several investigators have identified the protective potency of bacterial outer membrane vesicles (OMVs) as a promising basis for bacterial vaccine candidates. This review is aimed at presenting these candidates of plague vaccine and the results of their analysis in animal models.
Collapse
Affiliation(s)
- Andrey A. Byvalov
- Komi Research Center, Laboratory of Microbial Physiology, Institute of Physiology, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia;
- Department of Biotechnology, Vyatka State University, 610000 Kirov, Russia
| | - Ilya V. Konyshev
- Komi Research Center, Laboratory of Microbial Physiology, Institute of Physiology, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia;
- Department of Biotechnology, Vyatka State University, 610000 Kirov, Russia
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| | - Svetlana V. Dentovskaya
- Laboratory for Plague Microbiology, Especially Dangerous Infections Department, State Research Center for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia;
| | - Andrey P. Anisimov
- Laboratory for Plague Microbiology, Especially Dangerous Infections Department, State Research Center for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia;
| |
Collapse
|
17
|
Kumar D, Batra L, Malik MT. Insights of Novel Coronavirus (SARS-CoV-2) disease outbreak, management and treatment. AIMS Microbiol 2020; 6:183-203. [PMID: 33134740 PMCID: PMC7595841 DOI: 10.3934/microbiol.2020013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022] Open
Abstract
Emerging and re-emerging viral diseases poses a threat to living organisms, and led to serious concern to humankind and public health. The last two decades, viral epidemics such as the severe acute respiratory syndrome (SARS-CoV) reported in the years 2002-2003, and H1N1 influenza (Swine flu) in 2009, middle east respiratory syndrome (MERS-CoV) from Saudi Arabia in 2012, Ebola virus in 2014-2016, and Zika virus in 2015. The recent outbreak of 2019-CoV-2 or severe acute respiratory syndrome-2 (SARS-CoV-2), novel coronavirus (2019-nCoV, or 2019 disease, COVID-19) in Dec 2019, from, Wuhan city of China, has severe implications of health concerns to the whole world, due to global spread and high health risk. More than 423349 deaths had occurred globally and is still increasing every day. The whole world is under a health emergency, and people are advised to stay at their homes to avoid the spread of person-to-person infection, and advised to maintain social distancing. The advancement in clinical diagnosis techniques like Real-Time PCR (RT-PCR), immunological, microscopy, and geographic information system (GIS) mapping technology helped in tacking the rapid diagnosis and tracking viral infection in a short period. In the same way, artificial intelligence (AI), combinatorial chemistry, and deep learning approaches help to find novel therapeutics in less time and wide applicability in biomedical research. National Institute of Allergy and Infectious Diseases (NIAID) has started the clinical trials of investigation COVID-19 vaccine. Therefore, we can expect vaccines to be available for this deadly disease in the coming few months.
Collapse
Affiliation(s)
- Dharmender Kumar
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal-131039, Sonepat, Haryana India
| | - Lalit Batra
- Institute for Cellular Therapeutics,Departments of Microbiology and Immunology, University of Louisville, Louisville, Kentucky-40202, USA
| | - Mohammad Tariq Malik
- Institute for Cellular Therapeutics,Departments of Microbiology and Immunology, University of Louisville, Louisville, Kentucky-40202, USA
- Departments of Microbiology and Immunology, Regenerative Medicine and Stem Cell Biology.and James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky-40202, USA
| |
Collapse
|
18
|
Gupta A, Narayan B, Kumar S, Verma SK. Vaccine Potential of a Recombinant Bivalent Fusion Protein LcrV-HSP70 Against Plague and Yersiniosis. Front Immunol 2020; 11:988. [PMID: 32595634 PMCID: PMC7303293 DOI: 10.3389/fimmu.2020.00988] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/27/2020] [Indexed: 01/31/2023] Open
Abstract
To counteract the deadly pathogens, i.e., Y. pestis, Y. enetrocolitica, and Y. pseudotuberculosis, we prepared a recombinant DNA construct lcrV-hsp70 encoding the bivalent fusion protein LcrV-HSP70. The lcrV gene of Y. pestis and hsp70 domain II DNA fragment of M. tuberculosis were amplified by PCR. The lcrV amplicon was first ligated in the pET vector using NcoI and BamHI restriction sites. Just downstream to the lcrV gene, the hsp70 domain II was ligated using BamHI and Hind III restriction sites. The in-frame and the orientation of cloned lcrV-hsp70 were checked by restriction analysis and nucleotide sequencing. The recombinant bivalent fusion protein LcrV-HSP70 was expressed in E. coli and purified by affinity chromatography. The vaccine potential of LcrV-HSP70 fusion protein was evaluated in formulation with alum. BALB/c mice were vaccinated, and the humoral and cellular immune responses were studied. The fusion protein LcrV-HSP70 induced a strong and significant humoral immune response in comparison to control animals. We also observed a significant difference in the expression levels of IFN-γ and TNF-α in LcrV–HSP70-immunized mice in comparison to control, HSP70, and LcrV groups. To test the protective efficacy of the LcrV–HSP70 fusion protein against plague and Yersiniosis, the vaccinated mice were challenged with Y. pestis, Y. enterocolitica, and Y. pseudotuberculosis separately. The bivalent fusion protein LcrV–HSP70 imparted 100% protection against the plague. In the case of Yersiniosis, on day 2 post challenge, there was a significant reduction in the number of CFU of Y. enterocolitica and Y. pseudotuberculosis in the blood (CFU/ml) and the spleen (CFU/g) of vaccinated animals in comparison to the LcrV, HSP70, and control group animals.
Collapse
Affiliation(s)
- Ankit Gupta
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Bineet Narayan
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Subodh Kumar
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | | |
Collapse
|
19
|
The Impact of Age and Sex on Mouse Models of Melioidosis. Pathogens 2020; 9:pathogens9020113. [PMID: 32054106 PMCID: PMC7168040 DOI: 10.3390/pathogens9020113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022] Open
Abstract
Mouse models have been used to generate critical data for many infectious diseases. In the case of Burkholderia pseudomallei, mouse models have been invaluable for bacterial pathogenesis studies as well as for testing novel medical countermeasures including both vaccines and therapeutics. Mouse models of melioidosis have also provided a possible way forward to better understand the chronicity associated with this infection, as it appears that BALB/c mice develop an acute infection with B. pseudomallei, whereas the C57BL/6 model is potentially more suggestive of a chronic infection. Several unanswered questions, however, persist around this model. In particular, little attention has been paid to the effect of age or sex on the disease outcome in these animal models. In this report, we determined the LD50 of the B. pseudomallei K96243 strain in both female and male BALB/c and C57BL/6 mice in three distinct age groups. Our data demonstrated a modest increase in susceptibility associated with sex in this model, and we documented important histopathological differences associated with the reproductive systems of each sex. There was a statistically significant inverse correlation between age and susceptibility. The older mice, in most cases, were more susceptible to the infection. Additionally, our retrospective analyses suggested that the impact of animal supplier on disease outcome in mice may be minimal. These observations were consistent regardless of whether the mice were injected with bacteria intraperitoneally or if they were exposed to aerosolized bacteria. All of these factors should be considered when designing experiments using mouse models of melioidosis.
Collapse
|
20
|
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: 0.7] [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
|