Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 500
Filter
1.
Front Immunol ; 15: 1397579, 2024.
Article in English | MEDLINE | ID: mdl-38835755

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.


Subject(s)
Mice, Inbred BALB C , Plague Vaccine , Plague , Yersinia pestis , Animals , Female , Plague/prevention & control , Plague/immunology , Male , Yersinia pestis/immunology , Plague Vaccine/immunology , Plague Vaccine/administration & dosage , Mice , Antibodies, Bacterial/blood , Sex Characteristics , Sex Factors , Disease Models, Animal , Vaccine Efficacy
2.
Front Immunol ; 15: 1277526, 2024.
Article in English | MEDLINE | ID: mdl-38605961

ABSTRACT

This study evaluated a depot-formulated cytokine-based adjuvant to improve the efficacy of the recombinant F1V (rF1V) plague vaccine and examined the protective response following aerosol challenge in a murine model. The results of this study showed that co-formulation of the Alhydrogel-adsorbed rF1V plague fusion vaccine with the depot-formulated cytokines recombinant human interleukin 2 (rhuIL-2) and/or recombinant murine granulocyte macrophage colony-stimulating factor (rmGM-CSF) significantly enhances immunogenicity and significant protection at lower antigen doses against a lethal aerosol challenge. These results provide additional support for the co-application of the depot-formulated IL-2 and/or GM-CSF cytokines to enhance vaccine efficacy.


Subject(s)
Plague Vaccine , Yersinia pestis , Humans , Animals , Mice , Cytokines , Antigens, Bacterial , Vaccines, Synthetic , Aerosols
3.
Int Immunopharmacol ; 132: 111952, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38555818

ABSTRACT

Yersinia pestis, the causative agent of plague, is a gram-negative bacterium that can be fatal if not treated properly. Three types of plague are currently known: bubonic, septicemic, and pneumonic plague, among which the fatality rate of septicemic and pneumonic plague is very high. Bubonic plague can be treated, but only if antibiotics are used at the initial stage of the infection. But unfortunately, Y. pestis has also shown resistance to certain antibiotics such as kanamycin, minocycline, tetracycline, streptomycin, sulfonamides, spectinomycin, and chloramphenicol. Despite tremendous progress in vaccine development against Y. pestis, there is no proper FDA-approved vaccine available to protect people from its infections. Therefore, effective broad-spectrum vaccine development against Y. pestis is indispensable. In this study, vaccinomics-assisted immunoinformatics techniques were used to find possible vaccine candidates by utilizing the core proteome prepared from 58 complete genomes of Y. pestis. Human non-homologous, pathogen-essential, virulent, and extracellular and membrane proteins are potential vaccine targets. Two antigenic proteins were prioritized for the prediction of lead epitopes by utilizing reverse vaccinology approaches. Four vaccine designs were formulated using the selected B- and T-cell epitopes coupled with appropriate linkers and adjuvant sequences capable of inducing potent immune responses. The HLA allele population coverage of the T-cell epitopes selected for vaccine construction was also analyzed. The V2 constructs were top-ranked and selected for further analysis on the basis of immunological, physicochemical, and immune-receptor docking interactions and scores. Docking and molecular dynamic simulations confirmed the stability of construct V2 interactions with the host immune receptors. Immune simulation analysis anticipated the strong immune profile of the prioritized construct. In silico restriction cloning ensured the feasible cloning ability of the V2 construct in the expression system of E. coli strain K12. It is anticipated that the designed vaccine construct may be safe, effective, and able to elicit strong immune responses against Y. pestis infections and may, therefore, merit investigation using in vitro and in vivo assays.


Subject(s)
Plague , Yersinia pestis , Yersinia pestis/immunology , Yersinia pestis/genetics , Humans , Plague/prevention & control , Plague/immunology , Plague Vaccine/immunology , Plague Vaccine/genetics , Genome, Bacterial , Vaccine Development , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Vaccines, Synthetic/immunology , Animals
4.
PLoS Pathog ; 20(3): e1012129, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38547321

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.


Subject(s)
Plague Vaccine , Vaccines , Yersinia pestis , Humans , Animals , Mice , Yersinia pestis/genetics , Open Reading Frames , Plague Vaccine/genetics , Cytokines/genetics
5.
Bull Exp Biol Med ; 176(4): 472-476, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38492103

ABSTRACT

Vaccine strains Yersinia pestis EV NIIEG at a dose of 103 CFU and Francisella tularensis 15 NIIEG at a dose of 102 CFU induced changes in the concentration of cyclic nucleotides in the thymus and spleen of white mice. Antigen-induced changes in the cAMP/cGMP ratio in immunocompetent organs had a phase or oscillatory character, which seems to be related to the regulation of postvaccination immunoreactivity in the body. Synthetic organoselenium compound 974zh stimulated an increase in the amplitude of cAMP/cGMP oscillations, indicating its stimulating effect on the immunogenic properties of vaccine strains at doses an order of magnitude below the standard doses.


Subject(s)
Plague , Tularemia , Yersinia pestis , Animals , Mice , Plague/prevention & control , Plague Vaccine , Spleen , Tularemia/prevention & control , Vaccination
6.
Small ; 20(15): e2307066, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38009518

ABSTRACT

A new Yersinia pseudotuberculosis mutant strain, YptbS46, carrying the lpxE insertion and pmrF-J deletion is constructed and shown to exclusively produce monophosphoryl lipid A (MPLA) having adjuvant properties. Outer membrane vesicles (OMVs) isolated from YptbS46 harboring an lcrV expression plasmid, pSMV13, are designated OMV46-LcrV, which contained MPLA and high amounts of LcrV (Low Calcium response V) and displayed low activation of Toll-like receptor 4 (TLR4). Intramuscular prime-boost immunization with 30 µg of of OMV46-LcrV exhibited substantially reduced reactogenicity than the parent OMV44-LcrV and conferred complete protection to mice against a high-dose of respiratory Y. pestis challenge. OMV46-LcrV immunization induced robust adaptive responses in both lung mucosal and systemic compartments and orchestrated innate immunity in the lung, which are correlated with rapid bacterial clearance and unremarkable lung damage during Y. pestis challenge. Additionally, OMV46-LcrV immunization conferred long-term protection. Moreover, immunization with reduced doses of OMV46-LcrV exhibited further lower reactogenicity and still provided great protection against pneumonic plague. The studies strongly demonstrate the feasibility of OMV46-LcrV as a new type of plague vaccine candidate.


Subject(s)
Lipid A/analogs & derivatives , Plague Vaccine , Plague , Yersinia pestis , Mice , Animals , Yersinia , Plague/prevention & control , Antigens, Bacterial
7.
Adv Mater ; 35(51): e2304514, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37784226

ABSTRACT

Bacterial pneumonia is the leading cause of death worldwide among all infectious diseases. However, currently available vaccines against fatal bacterial lung infections, e.g., pneumonic plague, are accompanied by limitations, including insufficient antigen-adjuvant co-delivery and inadequate immune stimulation. Therefore, there is an urgent requirement to develop next-generation vaccines to improve the interaction between antigen and adjuvant, as well as enhance the effects of immune stimulation. This study develops a novel amino-decorated mesoporous manganese silicate nanoparticle (AMMSN) loaded with rF1-V10 (rF1-V10@AMMSN) to prevent pneumonic plague. These results suggest that subcutaneous immunization with rF1-V10@AMMSN in a prime-boost strategy induces robust production of rF1-V10-specific IgG antibodies with a geometric mean titer of 315,844 at day 42 post-primary immunization, which confers complete protection to mice against 50 × LD50 of Yersinia pestis (Y. pestis) challenge via the aerosolized intratracheal route. Mechanistically, rF1-V10@AMMSN can be taken up by dendritic cells (DCs) and promote DCs maturation through activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway and production of type I interferon. This process results in enhanced antigen presentation and promotes rF1-V10-mediated protection against Y. pestis infection. This manganese-based nanoparticle vaccine represents a valuable strategy for combating fatal bacterial pneumonia.


Subject(s)
Plague Vaccine , Plague , Pneumonia, Bacterial , Vaccines , Mice , Animals , Plague/prevention & control , Nanovaccines , Manganese , Antigens, Bacterial/genetics , Pneumonia, Bacterial/prevention & control , Adjuvants, Immunologic , Bacterial Proteins
8.
Hum Vaccin Immunother ; 19(2): 2216085, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37289480

ABSTRACT

Yersinia pestis is a gram-negative bacterium that causes plague in animals and humans. Depending on the route of disease transmission, the bacterium can cause an acute, often fatal disease that has a narrow window for treatment with antibiotics. Additionally, antibiotic resistant strains have been identified, emphasizing the need for novel treatments. Antibody therapy is an appealing option that can direct the immune system to clear bacterial infections. Advances in biotechnology have made both engineering and producing antibodies easier and more affordable. In this study, two screening assays were optimized to evaluate the ability of antibodies to promote phagocytosis of Y. pestis by macrophages and to induce a cytokine signature in vitro that may be predictive of protection in vivo. We evaluated a panel of 21 mouse monoclonal antibodies targeting either the anti-phagocytic capsule F1 protein or the LcrV antigen, which is part of the type 3 secretion system that facilitates translocation of virulence factors into the host cell, using two functional assays. Anti-F1 and anti-LcrV monoclonal antibodies both increased bacterial uptake by macrophages, with greater uptake observed in the presence of antibodies that were protective in the mouse pneumonic plague model. In addition, the protective anti-F1 and anti-LcrV antibodies produced unique cytokine signatures that were also associated with in vivo protection. These antibody-dependent characteristics from in vitro functional assays will be useful in down-selecting efficacious novel antibodies that can be used for treatment of plague.


Subject(s)
Plague Vaccine , Plague , Yersinia pestis , Mice , Humans , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Bacterial , Antibodies, Bacterial , Cytokines , Pore Forming Cytotoxic Proteins
9.
Sci Adv ; 9(10): eadg1036, 2023 03 10.
Article in English | MEDLINE | ID: mdl-36888708

ABSTRACT

Messenger RNA (mRNA) lipid nanoparticle (LNP) vaccines have emerged as an effective vaccination strategy. Although currently applied toward viral pathogens, data concerning the platform's effectiveness against bacterial pathogens are limited. Here, we developed an effective mRNA-LNP vaccine against a lethal bacterial pathogen by optimizing mRNA payload guanine and cytosine content and antigen design. We designed a nucleoside-modified mRNA-LNP vaccine based on the bacterial F1 capsule antigen, a major protective component of Yersinia pestis, the etiological agent of plague. Plague is a rapidly deteriorating contagious disease that has killed millions of people during the history of humankind. Now, the disease is treated effectively with antibiotics; however, in the case of a multiple-antibiotic-resistant strain outbreak, alternative countermeasures are required. Our mRNA-LNP vaccine elicited humoral and cellular immunological responses in C57BL/6 mice and conferred rapid, full protection against lethal Y. pestis infection after a single dose. These data open avenues for urgently needed effective antibacterial vaccines.


Subject(s)
Plague Vaccine , Plague , Yersinia pestis , Mice , Animals , Plague/prevention & control , Plague Vaccine/genetics , Bacterial Proteins/genetics , Mice, Inbred C57BL , Yersinia pestis/genetics , Antigens, Bacterial/genetics
10.
Infect Immun ; 90(8): e0016522, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35900096

ABSTRACT

A newly attenuated Yersinia pseudotuberculosis strain (designated Yptb1) with triple mutation Δasd ΔyopK ΔyopJ and chromosomal insertion of the Y. pestis caf1R-caf1M-caf1A-caf1 operon was constructed as a live vaccine platform. Yptb1 tailored with an Asd+ plasmid (pYA5199) (designated Yptb1[pYA5199]) simultaneously delivers Y. pestis LcrV and F1. The attenuated Yptb1(pYA5199) localized in the Peyer's patches, lung, spleen, and liver for a few weeks after oral immunization without causing any disease symptoms in immunized rodents. An oral prime-boost Yptb1(pYA5199) immunization stimulated potent antibody responses to LcrV, F1, and Y. pestis whole-cell lysate (YPL) in Swiss Webster mice and Brown Norway rats. The prime-boost Yptb1(pYA5199) immunization induced higher antigen-specific humoral and cellular immune responses in mice than a single immunization did, and it provided complete short-term and long-term protection against a high dose of intranasal Y. pestis challenge in mice. Moreover, the prime-boost immunization afforded substantial protection for Brown Norway rats against an aerosolized Y. pestis challenge. Our study highlights that Yptb1(pYA5199) has high potential as an oral vaccine candidate against pneumonic plague.


Subject(s)
Plague Vaccine , Plague , Yersinia pestis , Yersinia pseudotuberculosis Infections , Yersinia pseudotuberculosis , Animals , Antibodies, Bacterial , Antigens, Bacterial/genetics , Mice , Plague/prevention & control , Rats , Vaccination , Yersinia pestis/genetics , Yersinia pseudotuberculosis/genetics
11.
Proc Natl Acad Sci U S A ; 119(11): e2109667119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35275791

ABSTRACT

SignificanceYersinia pestis, the etiologic agent of plague, has been responsible for high mortality in several epidemics throughout human history. This plague bacillus has been used as a biological weapon during human history and is currently one of the deadliest biological threats. Currently, no licensed plague vaccines are available in the Western world. Since an array of immunogens are enclosed in outer membrane vesicles (OMVs), immune responses elicited by OMVs against a diverse range of antigens may reduce the likelihood of antigen circumvention. Therefore, self-adjuvanting OMVs from a remodeled Yersinia pseudotuberculosis strain as a type of plague vaccine could diversify prophylactic choices and solve current vaccine limitations.


Subject(s)
Antigens, Bacterial , Lipid A , Plague Vaccine , Plague , Pore Forming Cytotoxic Proteins , Yersinia pseudotuberculosis , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Lethal Dose 50 , Lipid A/genetics , Lipid A/immunology , Mice , Plague/prevention & control , Plague Vaccine/administration & dosage , Plague Vaccine/genetics , Plague Vaccine/immunology , Plasmids/genetics , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/immunology
12.
Front Immunol ; 13: 793382, 2022.
Article in English | MEDLINE | ID: mdl-35154110

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.


Subject(s)
Plague Vaccine/immunology , Plague/prevention & control , Vaccines, Subunit/immunology , Yersinia pestis/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Disease Models, Animal , Drug Compounding , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Mucosal , Immunization/methods , Mice , Mice, Inbred BALB C , Organ Specificity , Plague/immunology , Plague/mortality , Plague Vaccine/administration & dosage , Plague Vaccine/chemistry , Recombinant Proteins/immunology , Respiratory Aerosols and Droplets , Respiratory Mucosa/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry
13.
mBio ; 12(6): e0322321, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34872353

ABSTRACT

Mice immunized with a combination of an adenovirus vector (Ad5-YFV) and live-attenuated (LMA)-based vaccines were evaluated for protective efficacy against pneumonic plague. While the Ad5-YFV vaccine harbors a fusion cassette of three genes encoding YscF, F1, and LcrV, LMA represents a mutant of parental Yersinia pestis CO92 deleted for genes encoding Lpp, MsbB, and Ail. Ad5-YFV and LMA were either administered simultaneously (1-dose regimen) or 21 days apart in various orders and route of administration combinations (2-dose regimen). The 2-dose regimen induced robust immune responses to provide full protection to animals against parental CO92 and its isogenic F1 deletion mutant (CAF-) challenges during both short- and long-term studies. Mice intranasally (i.n.) immunized with Ad5-YFV first followed by LMA (i.n. or intramuscularly [i.m.]) had higher T- and B-cell proliferative responses and LcrV antibody titers than those in mice vaccinated with LMA (i.n. or i.m.) first ahead of Ad5-YFV (i.n.) during the long-term study. Specifically, the needle- and adjuvant-free vaccine combination (i.n.) is ideal for use in plague regions of endemicity. Conversely, with a 1-dose regimen, mice vaccinated with Ad5-YFV i.n. and LMA by the i.m. route provided complete protection to animals against CO92 and its CAF- mutant challenges and elicited Th1/Th2, as well as Th17 responses, making it suitable for emergency vaccination during a plague outbreak or bioterrorist attack. This is a first study in which a viral vector-based and live-attenuated vaccines were effectively used in combination, representing adjuvant- and/or needle-free immunization, with each vaccine triggering a distinct cellular immune response. IMPORTANCE Yersinia pestis, the causative agent of plague, is a Tier-1 select agent and a reemerging human pathogen. A 2017 outbreak in Madagascar with >75% of cases being pneumonic and 8.6% causalities emphasized the importance of the disease. The World Health Organization has indicated an urgent need to develop new-generation subunit and live-attenuated plague vaccines. We have developed a subunit vaccine, including three components (YscF, F1, and LcrV) using an adenovirus platform (Ad5-YFV). In addition, we have deleted virulence genes of Y. pestis (e.g., lpp, msbB, and ail) to develop a live-attenuated vaccine (LMA). Both of these vaccines generated robust humoral and cellular immunity and were highly efficacious in several animal models. We hypothesized the use of a heterologous prime-boost strategy or administrating both vaccines simultaneously could provide an adjuvant- and/or a needle-free vaccine(s) that has attributes of both vaccines for use in regions of endemicity and during an emergency situation.


Subject(s)
Adenoviridae/immunology , Antigens, Bacterial/administration & dosage , Plague Vaccine/administration & dosage , Plague/prevention & control , Pneumonia/prevention & control , Vaccines, Attenuated/administration & dosage , Yersinia pestis/immunology , Adenoviridae/genetics , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Mice , Plague/immunology , Plague/microbiology , Plague Vaccine/genetics , Plague Vaccine/immunology , Pneumonia/immunology , Pneumonia/microbiology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Yersinia pestis/genetics
14.
Vector Borne Zoonotic Dis ; 21(12): 921-940, 2021 12.
Article in English | MEDLINE | ID: mdl-34757815

ABSTRACT

The plague bacterium Yersinia pestis is lethal to endangered black-footed ferrets (Mustela nigripes, BFF) and the prairie dogs (Cynomys spp., PD) on which they depend for habitat and prey. We assessed the effectiveness of an oral sylvatic plague vaccine delivered in baits to black-tailed PD (Cynomys ludovicianus, BTPD) from 2013 to 2017 on the Charles M. Russell National Wildlife Refuge (CMR) in northcentral Montana. We permanently marked BTPD on four paired vaccine (N = 1,349 individuals) and placebo plots (N = 926; 7,027 total captures). We analyzed capture-recapture data under a Cormack-Jolly-Seber model to estimate annual apparent survival. Overall, survival averaged 0.05 lower on vaccine plots than on paired placebo plots. Immediately before noticeable die-offs and detecting plague on pairs CMR1 and CMR2, 89% of BTPD sampled on vaccine plots had consumed at least one bait and the immune systems (pleural) of 40% were likely boosted by consuming baits over multiple years. Survival to the following year was 0.16 and 0.05 on the vaccine plots and 0.19 and 0.06 on the placebo plots for pairs CMR1 and CMR2, respectively. These rates were markedly lower than 0.63, the overall average estimate on those same plots during the previous 3 years. PD populations subjected to such large die-offs would not be expected to sustain a BFF population. An overriding limitation to achieving sufficient protection rests with vaccine delivery constraints. Late summer/fall bait distribution results in the highest bait uptake rates. However, the PD birth pulse each spring can double the size of populations in most years, greatly reducing the proportion of vaccinates in populations and diminishing potential herd immunity benefits. In addition to nonvaccinated juveniles and PD that do not consume bait, incomplete vaccine protection and time required for immunity to develop leaves a large majority of PD populations vulnerable to plague for 6-7 months or more each year.


Subject(s)
Plague Vaccine , Rodent Diseases , Siphonaptera , Yersinia pestis , Animals , Ferrets , Sciuridae
15.
Front Immunol ; 12: 726416, 2021.
Article in English | MEDLINE | ID: mdl-34512658

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.


Subject(s)
Adjuvants, Immunologic , Antigens, Bacterial/immunology , Plague Vaccine/immunology , Plague/prevention & control , Toll-Like Receptors/physiology , Animals , Female , Mice , Mice, Inbred BALB C , Plague/immunology , Vaccination , Vaccine Efficacy , Vaccines, Synthetic/immunology , Yersinia pestis/immunology
16.
Appl Microbiol Biotechnol ; 105(12): 4931-4941, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34142207

ABSTRACT

As the reality of pandemic threats challenges humanity, exemplified during the ongoing SARS-CoV-2 infections, the development of vaccines targeting these etiological agents of disease has become increasingly critical. Of paramount concern are novel and reemerging pathogens that could trigger such events, including the plague bacterium Yersinia pestis. Y. pestis is responsible for more human deaths than any other known pathogen and exists globally in endemic regions of the world, including the four corners region and Northern California in the USA. Recent cases have been scattered throughout the world, including China and the USA, with serious outbreaks in Madagascar during 2008, 2013-2014, and, most recently, 2017-2018. This review will focus on recent advances in plague vaccine development, a seemingly necessary endeavor, as there is no Food and Drug Administration-licensed vaccine available for human distribution in western nations, and that antibiotic-resistant strains are recovered clinically or intentionally developed. Progress and recent development involving subunit, live-attenuated, and nucleic acid-based plague vaccine candidates will be discussed in this review. KEY POINTS: • Plague vaccine development remains elusive yet critical. • DNA, animal, and live-attenuated vaccine candidates gain traction.


Subject(s)
COVID-19 , Plague Vaccine , Plague , Yersinia pestis , Animals , Antibodies, Bacterial , China , Humans , SARS-CoV-2 , Vaccines, Attenuated
17.
J Wildl Dis ; 57(3): 632-636, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33787901

ABSTRACT

The variable response of wild mice to Yersinia pestis infection, the causative agent of plague, has generated much speculation concerning their role in the ecology of this potentially lethal disease. Researchers have questioned the means by which Y. pestis is maintained in nature and also sought methods for managing the disease. Here we assessed the efficacy of a new tool, the sylvatic plague vaccine (SPV), in wild-caught northern grasshopper mice (Onychomys leucogaster) and commercially acquired Sonoran deer mice (Peromyscus maniculatus sonoriensis). More than 40% of the animals survived a subcutaneous Y. pestis challenge of 175,000 colony forming units (over 30,000 times the white mouse 50% lethal dose) in both vaccine-treated and control groups. Our results indicate that SPV distribution is unlikely to protect adult mice from plague infection in field settings and corroborate the heterogeneous response to Y. pestis infection in mice reported by others.


Subject(s)
Mice/microbiology , Peromyscus/microbiology , Plague Vaccine , Plague/veterinary , Rodent Diseases , Yersinia pestis , Animals , Plague/prevention & control , Rodent Diseases/microbiology , Rodent Diseases/prevention & control
18.
Vaccine ; 39(9): 1435-1444, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33531196

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.


Subject(s)
Plague Vaccine , Plague , Sepsis , Yersinia pestis , Animals , Antibodies, Bacterial , Antigens, Bacterial , Bacterial Proteins , Mice , Plague/prevention & control , Pore Forming Cytotoxic Proteins , Vaccines, Subunit
19.
Biomolecules ; 10(12)2020 12 18.
Article in English | MEDLINE | ID: mdl-33353123

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.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Plague/prevention & control , Vaccines , Yersinia pestis/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins , Humans , Immune System , Immunoglobulin G , Mice , Plague Vaccine/immunology , Yersinia pestis/immunology
20.
Biomolecules ; 10(11)2020 11 14.
Article in English | MEDLINE | ID: mdl-33202679

ABSTRACT

The Gram-negative bacterium Yersinia pestis causes plague, a fatal flea-borne anthropozoonosis, which can progress to aerosol-transmitted pneumonia. Y. pestis overcomes the innate immunity of its host thanks to many pathogenicity factors, including plasminogen activator, Pla. This factor is a broad-spectrum outer membrane protease also acting as adhesin and invasin. Y. pestis uses Pla adhesion and proteolytic capacity to manipulate the fibrinolytic cascade and immune system to produce bacteremia necessary for pathogen transmission via fleabite or aerosols. Because of microevolution, Y. pestis invasiveness has increased significantly after a single amino-acid substitution (I259T) in Pla of one of the oldest Y. pestis phylogenetic groups. This mutation caused a better ability to activate plasminogen. In paradox with its fibrinolytic activity, Pla cleaves and inactivates the tissue factor pathway inhibitor (TFPI), a key inhibitor of the coagulation cascade. This function in the plague remains enigmatic. Pla (or pla) had been used as a specific marker of Y. pestis, but its solitary detection is no longer valid as this gene is present in other species of Enterobacteriaceae. Though recovering hosts generate anti-Pla antibodies, Pla is not a good subunit vaccine. However, its deletion increases the safety of attenuated Y. pestis strains, providing a means to generate a safe live plague vaccine.


Subject(s)
Plasminogen Activators/metabolism , Protein Interaction Maps/physiology , Yersinia pestis/metabolism , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Humans , Plague/genetics , Plague/metabolism , Plague/prevention & control , Plague Vaccine/administration & dosage , Plague Vaccine/genetics , Plague Vaccine/metabolism , Plasminogen Activators/chemistry , Plasminogen Activators/genetics , Point Mutation/physiology , Protein Structure, Secondary , Yersinia pestis/classification , Yersinia pestis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...