Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.751
Filter
1.
Bull World Health Organ ; 102(6): 378-379, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38828056

ABSTRACT

Attention is beginning to focus on the need for vaccines to tackle antimicrobial resistant pathogens. Gary Humphreys reports.


Subject(s)
Drug Resistance, Bacterial , Humans , Bacterial Vaccines , Anti-Bacterial Agents/pharmacology
2.
Front Immunol ; 15: 1392681, 2024.
Article in English | MEDLINE | ID: mdl-38835751

ABSTRACT

Background: Pasteurella multocida is a bacterial pathogen that causes a variety of infections across diverse animal species, with one of the most devastating associated diseases being hemorrhagic septicemia. Outbreaks of hemorrhagic septicemia in cattle and buffaloes are marked by rapid progression and high mortality. These infections have particularly harmful socio-economic impacts on small holder farmers in Africa and Asia who are heavily reliant on a small number of animals kept as a means of subsistence for milk and draft power purposes. A novel vaccine target, PmSLP-3, has been identified on the surface of hemorrhagic septicemia-associated strains of P. multocida and was previously shown to elicit robust protection in cattle against lethal challenge with a serogroup B strain. Methods: Here, we further investigate the protective efficacy of this surface lipoprotein, including evaluating the immunogenicity and protection upon formulation with a variety of adjuvants in both mice and cattle. Results: PmSLP-3 formulated with Montanide ISA 61 elicited the highest level of serum and mucosal IgG, elicited long-lasting serum antibodies, and was fully protective against serogroup B challenge. Studies were then performed to identify the minimum number of doses required and the needed protein quantity to maintain protection. Duration studies were performed in cattle, demonstrating sustained serum IgG titres for 3 years after two doses of vaccine and full protection against lethal serogroup B challenge at 7 months after a single vaccine dose. Finally, a serogroup E challenge study was performed, demonstrating that PmSLP-3 vaccine can provide protection against challenge by the two serogroups responsible for hemorrhagic septicemia. Conclusion: Together, these data indicate that PmSLP-3 formulated with Montanide ISA 61 is an immunogenic and protective vaccine against hemorrhagic septicemia-causing P. multocida strains in cattle.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Cattle Diseases , Hemorrhagic Septicemia , Pasteurella multocida , Animals , Cattle , Pasteurella multocida/immunology , Hemorrhagic Septicemia/prevention & control , Hemorrhagic Septicemia/veterinary , Hemorrhagic Septicemia/immunology , Hemorrhagic Septicemia/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Cattle Diseases/prevention & control , Cattle Diseases/immunology , Cattle Diseases/microbiology , Mice , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Female , Serogroup , Pasteurella Infections/prevention & control , Pasteurella Infections/veterinary , Pasteurella Infections/immunology , Pasteurella Infections/microbiology , Adjuvants, Immunologic/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice, Inbred BALB C , Vaccination
3.
PLoS One ; 19(5): e0294998, 2024.
Article in English | MEDLINE | ID: mdl-38713688

ABSTRACT

Tularemia is a zoonotic disease caused by the facultative intracellular gram-negative bacterium Francisella tularensis. F. tularensis has a very low infection dose by the aerosol route which can result in an acute, and potentially lethal, infection in humans. Consequently, it is classified as a Category A bioterrorism agent by the US Centers for Disease Control (CDC) and is a pathogen of concern for the International Biodefence community. There are currently no licenced tularemia vaccines. In this study we report on the continued assessment of a tularemia subunit vaccine utilising ß-glucan particles (GPs) as a vaccine delivery platform for immunogenic F. tularensis antigens. Using a Fischer 344 rat infection model, we demonstrate that a GP based vaccine comprising the F. tularensis lipopolysaccharide antigen together with the protein antigen FTT0814 provided partial protection of F344 rats against an aerosol challenge with a high virulence strain of F. tularensis, SCHU S4. Inclusion of imiquimod as an adjuvant failed to enhance protective efficacy. Moreover, the level of protection afforded was dependant on the challenge dose. Immunological characterisation of this vaccine demonstrated that it induced strong antibody immunoglobulin responses to both polysaccharide and protein antigens. Furthermore, we demonstrate that the FTT0814 component of the GP vaccine primed CD4+ and CD8+ T-cells from immunised F344 rats to express interferon-γ, and CD4+ cells to express interleukin-17, in an antigen specific manner. These data demonstrate the development potential of this tularemia subunit vaccine and builds on a body of work highlighting GPs as a promising vaccine platform for difficult to treat pathogens including those of concern to the bio-defence community.


Subject(s)
Bacterial Vaccines , Disease Models, Animal , Francisella tularensis , Rats, Inbred F344 , Tularemia , Vaccines, Subunit , Animals , Tularemia/prevention & control , Tularemia/immunology , Rats , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Francisella tularensis/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Glucans/immunology , Glucans/pharmacology , T-Lymphocytes/immunology , Female , Antigens, Bacterial/immunology
4.
Genome Med ; 16(1): 67, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711148

ABSTRACT

BACKGROUND: Infections caused by multidrug-resistant gram-negative bacteria present a severe threat to global public health. The WHO defines drug-resistant Klebsiella pneumoniae as a priority pathogen for which alternative treatments are needed given the limited treatment options and the rapid acquisition of novel resistance mechanisms by this species. Longitudinal descriptions of genomic epidemiology of Klebsiella pneumoniae can inform management strategies but data from sub-Saharan Africa are lacking. METHODS: We present a longitudinal analysis of all invasive K. pneumoniae isolates from a single hospital in Blantyre, Malawi, southern Africa, from 1998 to 2020, combining clinical data with genome sequence analysis of the isolates. RESULTS: We show that after a dramatic increase in the number of infections from 2016 K. pneumoniae becomes hyperendemic, driven by an increase in neonatal infections. Genomic data show repeated waves of clonal expansion of different, often ward-restricted, lineages, suggestive of hospital-associated transmission. We describe temporal trends in resistance and surface antigens, of relevance for vaccine development. CONCLUSIONS: Our data highlight a clear need for new interventions to prevent rather than treat K. pneumoniae infections in our setting. Whilst one option may be a vaccine, the majority of cases could be avoided by an increased focus on and investment in infection prevention and control measures, which would reduce all healthcare-associated infections and not just one.


Subject(s)
Klebsiella Infections , Klebsiella pneumoniae , Klebsiella pneumoniae/genetics , Humans , Klebsiella Infections/epidemiology , Klebsiella Infections/microbiology , Longitudinal Studies , Bacterial Vaccines/immunology , Adult , Female , Hospitals , Child , Male , Child, Preschool , Infant , Middle Aged , Africa South of the Sahara/epidemiology , Cross Infection/microbiology , Adolescent , Genome, Bacterial , Drug Resistance, Multiple, Bacterial/genetics , Infant, Newborn , Malawi/epidemiology , Young Adult
5.
BMC Immunol ; 25(1): 27, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38706005

ABSTRACT

BACKGROUND: Due to antibiotic resistance, the Klebsiella genus is linked to morbidity and death, necessitating the development of a universally protective vaccine against Klebsiella pathogens. METHODS: Core sequence analysis prioritized non-redundant host molecules and expected lipid bilayer peptides from fully sequenced Klebsiella genomes. These proteins were refined to identify epitopes, examining their immunogenicity, toxicity, solubility, and interaction with MHC alleles. Epitopes were linked to CPG ODN C274 via EAAAK, HEYGAEALERAG, and GGGS linkers to enhance immunological responses. The vaccine's tertiary structure was modelled and docked with MHC-I and MHC-II. RESULTS: Fifty-five proteins were recognized in the Vaxign collection as having remarkable features. Twenty-three proteins with potential pathogenicity were then identified. Eight options for vaccines emerged after the immunogenicity of proteins was examined. The best antigens were three proteins: MrkD, Iron-regulated lipid membrane polypeptides, and RmpA. These compounds were selected for their sensitivity. The structural protein sequences of K. pneumoniae were utilized to identify seven CTL epitopes, seven HTL epitopes, and seven LBL epitopes, respectively. The produced immunization displayed a stable contact with the receptors, based on molecular dynamic simulations lasting 250 nanoseconds. Intermolecular binding free energies also indicated the dominance of the van der Waals and electrostatic energies. CONCLUSION: In summary, the results of this study might help scientists develop a novel vaccine to prevent K. pneumoniae infections.


Subject(s)
Bacterial Vaccines , Klebsiella Infections , Klebsiella pneumoniae , Klebsiella pneumoniae/immunology , Bacterial Vaccines/immunology , Klebsiella Infections/immunology , Klebsiella Infections/prevention & control , Animals , Epitopes, T-Lymphocyte/immunology , Mice , Humans , Molecular Dynamics Simulation , Antigens, Bacterial/immunology , Oligodeoxyribonucleotides/immunology , Epitopes/immunology , Molecular Docking Simulation
6.
PLoS One ; 19(5): e0301688, 2024.
Article in English | MEDLINE | ID: mdl-38768145

ABSTRACT

Swine atrophic rhinitis is a disease caused by Pasteurella multocida and Bordetella bronchiseptica that affects pigs. Inactivated vaccines containing the toxins produced by Pasteurella multocida and Bordetella bronchiseptica have been widely used for the prevention of swine atrophic rhinitis. The efficacy of a vaccine is correlated with the amount of antigen present; however, the protective toxin of P. multocida bound to aluminum hydroxide, which is used as an adjuvant, can hinder the monitoring of the antigen concentration in the vaccine. This study assessed the applicability of a dot immunoassay as an antigen quantification method using monoclonal antibodies. This quantification method was able to detect the antigen with high specificity and sensitivity even when the antigen was bound to the adjuvant, and its application to vaccine products revealed a correlation between the amount of antigen present in the vaccine and the neutralizing antibody titers induced in pigs. The antigen quantification method presented in this study is a simple and sensitive assay capable of quantifying the amount of antigen present in a vaccine that can be used as an alternative quality control measure.


Subject(s)
Adjuvants, Immunologic , Aluminum Hydroxide , Antigens, Bacterial , Bacterial Vaccines , Pasteurella multocida , Rhinitis, Atrophic , Swine Diseases , Animals , Pasteurella multocida/immunology , Swine , Rhinitis, Atrophic/immunology , Rhinitis, Atrophic/prevention & control , Rhinitis, Atrophic/microbiology , Bacterial Vaccines/immunology , Antigens, Bacterial/immunology , Swine Diseases/prevention & control , Swine Diseases/microbiology , Swine Diseases/immunology , Bordetella bronchiseptica/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Pasteurella Infections/prevention & control , Pasteurella Infections/veterinary , Pasteurella Infections/immunology , Antibodies, Neutralizing/immunology
7.
J Nanobiotechnology ; 22(1): 273, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773507

ABSTRACT

BACKGROUND: The outer membrane vesicles (OMVs) produced by Gram-negative bacteria can modulate the immune system and have great potentials for bacterial vaccine development. RESULTS: A highly active Acinetobacter baumannii phage lysin, LysP53, can stimulate the production of OMVs after interacting with A. baumannii, Escherichia coli, and Salmonella. The OMVs prepared by the lysin (LOMVs) from A. baumannii showed better homogeneity, higher protein yield, lower endotoxin content, and lower cytotoxicity compared to the naturally produced OMVs (nOMVs). The LOMVs contain a significantly higher number of cytoplasmic and cytoplasmic membrane proteins but a smaller number of periplasmic and extracellular proteins compared to nOMVs. Intramuscular immunization with either LOMVs or nOMVs three times provided robust protection against A. baumannii infections in both pneumonia and bacteremia mouse models. Intranasal immunization offered good protection in the pneumonia model but weaker protection (20-40%) in the bacteremia model. However, with a single immunization, LOMVs demonstrated better protection than the nOMVs in the pneumonia mouse model. CONCLUSIONS: The novel lysin approach provides a superior choice compared to current methods for OMV production, especially for vaccine development.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Bacteriophages , Animals , Acinetobacter Infections/prevention & control , Mice , Female , Mice, Inbred BALB C , Bacterial Vaccines/immunology , Immunization , Extracellular Vesicles , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane Proteins/immunology , Disease Models, Animal , Humans , Administration, Intranasal , Viral Proteins
8.
Front Immunol ; 15: 1392456, 2024.
Article in English | MEDLINE | ID: mdl-38779673

ABSTRACT

In response to the global threat posed by bacterial pathogens, which are the second leading cause of death worldwide, vaccine development is challenged by the diversity of bacterial serotypes and the lack of immunoprotection across serotypes. To address this, we introduce BacScan, a novel genome-wide technology for the rapid discovery of conserved highly immunogenic proteins (HIPs) across serotypes. Using bacterial-specific serum, BacScan combines phage display, immunoprecipitation, and next-generation sequencing to comprehensively identify all the HIPs in a single assay, thereby paving the way for the development of universally protective vaccines. Our validation of this technique with Streptococcus suis, a major pathogenic threat, led to the identification of 19 HIPs, eight of which conferred 20-100% protection against S. suis challenge in animal models. Remarkably, HIP 8455 induced complete immunity, making it an exemplary vaccine target. BacScan's adaptability to any bacterial pathogen positions it as a revolutionary tool that can expedite the development of vaccines with broad efficacy, thus playing a critical role in curbing bacterial transmission and slowing the march of antimicrobial resistance.


Subject(s)
Bacterial Proteins , Animals , Mice , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Streptococcal Infections/immunology , Streptococcal Infections/prevention & control , Streptococcus suis/immunology , Streptococcus suis/genetics , Genome, Bacterial , High-Throughput Nucleotide Sequencing , Female , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Humans , Bacterial Vaccines/immunology
9.
Microb Genom ; 10(5)2024 May.
Article in English | MEDLINE | ID: mdl-38787376

ABSTRACT

Lyme disease (LD), caused by spirochete bacteria of the genus Borrelia burgdorferi sensu lato, remains the most common vector-borne disease in the northern hemisphere. Borrelia outer surface protein A (OspA) is an integral surface protein expressed during the tick cycle, and a validated vaccine target. There are at least 20 recognized Borrelia genospecies, that vary in OspA serotype. This study presents a new in silico sequence-based method for OspA typing using next-generation sequence data. Using a compiled database of over 400 Borrelia genomes encompassing the 4 most common disease-causing genospecies, we characterized OspA diversity in a manner that can accommodate existing and new OspA types and then defined boundaries for classification and assignment of OspA types based on the sequence similarity. To accommodate potential novel OspA types, we have developed a new nomenclature: OspA in silico type (IST). Beyond the ISTs that corresponded to existing OspA serotypes 1-8, we identified nine additional ISTs that cover new OspA variants in B. bavariensis (IST9-10), B. garinii (IST11-12), and other Borrelia genospecies (IST13-17). The IST typing scheme and associated OspA variants are available as part of the PubMLST Borrelia spp. database. Compared to traditional OspA serotyping methods, this new computational pipeline provides a more comprehensive and broadly applicable approach for characterization of OspA type and Borrelia genospecies to support vaccine development.


Subject(s)
Antigens, Surface , Bacterial Outer Membrane Proteins , Lipoproteins , Lyme Disease , Bacterial Outer Membrane Proteins/genetics , Lyme Disease/microbiology , Lipoproteins/genetics , Antigens, Surface/genetics , Borrelia burgdorferi/genetics , Borrelia burgdorferi/classification , Computer Simulation , Humans , Genome, Bacterial , Borrelia burgdorferi Group/genetics , Borrelia burgdorferi Group/classification , High-Throughput Nucleotide Sequencing/methods , Serogroup , Phylogeny , Bacterial Vaccines
10.
Front Biosci (Landmark Ed) ; 29(5): 176, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38812301

ABSTRACT

BACKGROUND: Listeria monocytogenes, a Gram-positive bacterium, is a prominent foodborne pathogen that causes listeriosis and poses substantial health hazards worldwide. The continuing risk of listeriosis outbreaks underlies the importance of designing an effective prevention strategy and developing a robust immune response by reverse vaccinology approaches. This study aimed to provide a critical approach for developing a potent multiepitope vaccine against this foodborne disease. METHODS: A chimeric peptide construct containing 5 B-cell epitopes, 16 major histocompatibility complex I (MHC-I) epitopes, and 18 MHC-II epitopes were used to create a subunit vaccination against L. monocytogenes. The vaccine safety was evaluated by several online methods, and molecular docking was performed using ClusPro to determine the binding affinity. Immune simulation was performed using the C-ImmSimm server to demonstrate the immune response. RESULTS: The results validated the antigenicity, non-allergenicity, and nontoxicity of the chimeric peptide construct, confirming its suitability as a subunit vaccine. Molecular docking showed a good score of 1276.5 and molecular dynamics simulations confirmed the construct's efficacy, demonstrating its promise as a good candidate for listeriosis prophylaxis. The population coverage was as high as 91.04% with a good immune response, indicating good antigen presentation with dendritic cells and production of memory cells. CONCLUSIONS: The findings of this study highlight the potential of the designed chimeric peptide construct as an effective subunit vaccine against Listeria, paving the way for future advances in preventive methods and vaccine design.


Subject(s)
Bacterial Vaccines , Computational Biology , Listeria monocytogenes , Listeriosis , Molecular Docking Simulation , Vaccines, Subunit , Listeria monocytogenes/immunology , Bacterial Vaccines/immunology , Vaccines, Subunit/immunology , Listeriosis/prevention & control , Listeriosis/immunology , Listeriosis/microbiology , Computational Biology/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Humans , Epitopes/immunology , Molecular Dynamics Simulation , Animals , Foodborne Diseases/prevention & control , Foodborne Diseases/microbiology , Foodborne Diseases/immunology , Immunoinformatics
11.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791526

ABSTRACT

Antimicrobial resistance (AMR) is one of the most critical threats to global public health in the 21st century, causing a large number of deaths every year in both high-income and low- and middle-income countries. Vaccines and monoclonal antibodies can be exploited to prevent and treat diseases caused by AMR pathogens, thereby reducing antibiotic use and decreasing selective pressure that favors the emergence of resistant strains. Here, differences in the mechanism of action and resistance of vaccines and monoclonal antibodies compared to antibiotics are discussed. The state of the art for vaccine technologies and monoclonal antibodies are reviewed, with a particular focus on approaches validated in clinical studies. By underscoring the scope and limitations of the different emerging technologies, this review points out the complementary of vaccines and monoclonal antibodies in fighting AMR. Gaps in antigen discovery for some pathogens, as well as challenges associated with the clinical development of these therapies against AMR pathogens, are highlighted.


Subject(s)
Anti-Bacterial Agents , Antibodies, Monoclonal , Humans , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Animals , Drug Resistance, Bacterial/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , Bacterial Infections/immunology , Bacterial Infections/drug therapy
12.
J Microbiol Biotechnol ; 34(5): 987-993, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38719774

ABSTRACT

Campylobacteriosis is a significant foodborne illness caused by Campylobacter bacteria. It is one of the most common bacterial causes of gastroenteritis worldwide, with poultry being a major reservoir and source of infection in humans. In poultry farms, Campylobacters colonize the intestinal tract of chickens and contaminate meat during processing. Vaccines under development against Campylobacters in poultry showed partial or no protection against their cecal colonization. Therefore, this review will elaborate on campylobacteriosis and emphasize the control strategies and recent vaccine trials against Campylobacters in poultry farms. The epidemiology, diagnosis, and treatment of Campylobacter infection, along with specific mention of poultry Campylobacter contamination events in Malaysia, will also be discussed.


Subject(s)
Campylobacter Infections , Campylobacter , Chickens , Farms , Poultry Diseases , Poultry , Animals , Campylobacter Infections/prevention & control , Campylobacter Infections/veterinary , Campylobacter Infections/microbiology , Campylobacter Infections/epidemiology , Campylobacter/isolation & purification , Poultry Diseases/prevention & control , Poultry Diseases/microbiology , Chickens/microbiology , Poultry/microbiology , Humans , Bacterial Vaccines/immunology , Malaysia/epidemiology , Meat/microbiology
13.
Vet Microbiol ; 293: 110093, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692193

ABSTRACT

Mycoplasma gallisepticum causes chronic respiratory disease in poultry. A novel vaccine, Vaxsafe MG304 (the ts-304 strain), has greater protective efficacy in chickens than the Vaxsafe MG (strain ts-11) vaccine when delivered by eye drop at 3 weeks of age. Applying this vaccine in the hatchery to 1-day-old birds, using mass administration methods, would improve animal welfare and reduce labour costs associated with handling individual birds. This study assessed the protection provided by vaccination with Vaxsafe MG304 after administration to 1-day-old chicks. Chicks were administered a single dose of the vaccine to assess the efficacy of either a high dose (107.0 colour changing units, CCU) or a low dose (105.7 CCU) after eye drop or spray (in water or gel) administration against experimental challenge with virulent M. gallisepticum strain Ap3AS at 7 weeks of age. The vaccine was able to colonise the palatine cleft of chicks after vaccination by eye drop (at both doses) or by spray (in water or gel) (at the high dose). The high dose of vaccine, when delivered by eye drop or spray, was shown to be safe and induced a serological response and protective immunity (as measured by tracheal mucosal thickness and air sac lesion scores) against challenge. Vaccination of 1-day-old chicks with Vaxsafe MG304 by eye drop induced protective immunity equivalent to vaccination at 3 weeks of age. Vaxsafe MG304 was also protective when applied by both coarse- and gel spray methods at the higher dose and is therefore a suitable live attenuated vaccine for use in 1-day-old chicks.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Chickens , Mycoplasma Infections , Mycoplasma gallisepticum , Poultry Diseases , Vaccination , Animals , Mycoplasma gallisepticum/immunology , Chickens/immunology , Chickens/microbiology , Poultry Diseases/prevention & control , Poultry Diseases/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Mycoplasma Infections/prevention & control , Mycoplasma Infections/veterinary , Mycoplasma Infections/immunology , Specific Pathogen-Free Organisms , Vaccination/veterinary , Antibodies, Bacterial/blood
14.
Vet Immunol Immunopathol ; 272: 110772, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704989

ABSTRACT

A live, infectious vaccine candidate for epizootic bovine abortion, designated EBAA Vaccine, USDA-APHIS Product code #1544.00, has been reported to be both safe and effective. Previous studies established that a single dose of EBAA vaccine administered to cows at potencies of either 2000 or 500 live P. abortibovis-infected murine spleen cells (P.a.-LIC) induced protective immunity for a minimum of 5 months. The current study employed 19 pregnant cows that were challenged with P. abortibovis in their 2nd trimester of gestation; 9 were vaccinated 17.2-months earlier as 1-year-olds with 2000 P.a.-LIC and 10 served as negative controls. Eighty-nine percent of the vaccinates gave birth to healthy calves as compared to 10% of challenge controls. Vaccine efficacy was significant when analyzed by prevented fractions (87.7%; 95% CI=0.4945-0.9781). Serologic data supports previous findings that pregnant cows with detectable P. abortibovis antibodies are immune to P. abortibovis challenge as demonstrated by the birth of healthy calves.


Subject(s)
Abortion, Veterinary , Animals , Cattle , Female , Pregnancy , Abortion, Veterinary/immunology , Abortion, Veterinary/prevention & control , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Seasons , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage
15.
Int Immunopharmacol ; 135: 112287, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38776850

ABSTRACT

Achromobacter xylosoxidans is an aerobic, catalase-positive, non-pigment-forming, Gram-negative, and motile bacterium. It potentially causes a wide range of human infections in cystic fibrosis and non-cystic fibrosis patients. However, developing a safe preventive or therapeutic solution against A. xylosoxidans remains challenging. This study aimed to construct an epitope-based vaccine candidate using immunoinformatic techniques. A. xylosoxidans was isolated from an auto workshop in Lahore, and its identification was confirmed through 16S rRNA amplification and bioinformatic analysis. Two protein targets with GenBank accession numbers AKP90890.1 and AKP90355.1 were selected for the vaccine construct. Both proteins exhibited antigenicity, with scores of 0.757 and 0.580, respectively and the epitopes were selected based on the IC50 value using the ANN 4.0 and NN-align 2.3 epitope prediction method for MHC I and MHC II epitopes respectively and predicted epitopes were analyzed for antigenicity, allergenicity and pathogenicity. The vaccine construct demonstrated structural stability, thermostability, solubility, and hydrophilicity. The vaccine produced 250 B-memory cells per mm3 and approximately 16,000 IgM + IgG counts, indicating an effective immune response against A. xylosoxidans. Moreover, the vaccine candidate interacted stably with toll-like receptor 5, a pattern recognition receptor, with a confidence score of 0.98. These results highlight the potency of the designed vaccine candidate, suggesting its potential to withstand rigorous in vitro and in vivo clinical trials. This epitope-based vaccine could serve as the first preventive immunotherapy against A. xylosoxidans infections, addressing this bacterium's health and financial burdens. The findings demonstrate the value of employing immunoinformatic tools in vaccine development, paving the way for more precise and tailored approaches to combating microbial threats.


Subject(s)
Achromobacter denitrificans , Bacterial Vaccines , Gram-Negative Bacterial Infections , RNA, Ribosomal, 16S , Achromobacter denitrificans/immunology , Achromobacter denitrificans/genetics , Bacterial Vaccines/immunology , Humans , RNA, Ribosomal, 16S/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/microbiology , Animals , Epitopes/immunology , Computer Simulation , Female , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Mice , Computational Biology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics
16.
Int J Biol Macromol ; 270(Pt 1): 132105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710251

ABSTRACT

In this study, a methodical workflow using subtractive proteomics, vaccine designing, molecular simulation, and agent-based modeling approaches were used to annotate the whole proteome of Burkholderia pseudomallei (strain K96243) for vaccine designing. Among the total 5717 proteins in the whole proteome, 505 were observed to be essential for the pathogen's survival and pathogenesis predicted by the Database of Essential Genes. Among these, 23 vaccine targets were identified, of which fimbrial assembly chaperone (Q63UH5), Outer membrane protein (Q63UH1), and Hemolysin-like protein (Q63UE4) were selected for the subsequent analysis based on the systematic approaches. Using immunoinformatic approaches CTL (cytotoxic T lymphocytes), HTL (helper T lymphocytes), IFN-positive, and B cell epitopes were predicted for these targets. A total of 9 CTL epitopes were added using the GSS linker, 6 HTL epitopes using the GPGPG linker, and 6 B cell epitopes using the KK linker. An adjuvant was added for enhanced antigenicity, an HIV-TAT peptide for improved delivery, and a PADRE sequence was added to form a 466 amino acids long vaccine construct. The construct was classified as non-allergenic, highly antigenic, and experimentally feasible. Molecular docking results validated the robust interaction of MEVC with immune receptors such as TLR2/4. Furthermore, molecular simulation revealed stable dynamics and compact nature of the complexes. The binding free energy results further validated the robust binding. In silico cloning, results revealed GC contents of 50.73 % and a CIA value of 0.978 which shows proper downstream processing. Immune simulation results reported that after the three injections of the vaccine a robust secondary immune response, improved antigen clearance, and effective immune memory generation were observed highlighting its potential for effective and sustained immunity. Future directions should encompass experimental validations, animal model studies, and clinical trials to substantiate the vaccine's efficacy, safety, and immunogenicity.


Subject(s)
Bacterial Vaccines , Burkholderia pseudomallei , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Proteomics , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Proteomics/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Molecular Docking Simulation , Humans , Bacterial Proteins/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Melioidosis/prevention & control , Melioidosis/immunology , Proteome , Molecular Dynamics Simulation
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 441-446, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38645870

ABSTRACT

Objective: To study the immunoadjuvant effects of chitosan oligosaccharide (COS), including the immune activation and the triggering of lysosomal escape, and to explore whether COS can be used as an adjuvant for attenuated live bacteria vector vaccines. Methods: 1) Mouse macrophages RAW264.7 cells were cultured with COS at 0 mg/mL (the control group) and 0.1-4 mg/mL for 24 h and the effect on cell viability was measured by CCK8 assay. Mouse macrophages RAW264.7 were treated with COS at 0 (the control group), 1, 2, and 4 mg/mL for 24 h. Then, the mRNA expression levels of the cytokines, including IFN-γ, IL-10, TGF-ß, and TLR4, were determined by RT-qPCR assay. 2) RAW264.7 cells were treated with 1 mL of PBS containing different components, including calcein at 50 µg/mL, COS at 2 mg/mL, and bafilomycin A1, an inhibitor, at 1 µmol/mL, for culturing. The cells were divided into the Calcein group, Calcein+COS group, and Calcein+COS+Bafilomycin A1 group accordingly. Laser scanning confocal microscopy was used to observe the phagocytosis and the intracellular fluorescence distribution of calcein, a fluorescent dye, in RAW264.7 cells in the presence or absence of COS intervention to determine whether COS was able to trigger lysosomal escape. 3) LM∆E6E7 and LI∆E6E7, the attenuated Listeria vector candidate therapeutic vaccines for cervical cancer, were encapsulated with COS at the mass concentrations of 0.5 mg/mL, 1 mg/mL, 2 mg/mL , 4 mg/mL, and 8 mg/mL. Then, the changes in zeta potential were measured to select the concentration of COS that successfully encapsulated the bacteria. Phagocytosis of the vaccine strains by RAW264.7 cells was measured before and after LM∆E6E7 and LI∆E6E7 were coated with COS at 2 mg/mL. Results: 1) CCK8 assays showed that, compared with the findings for the control group, the intervention of RAW264.7 cells with COS at different concentrations for 24 h was not toxic to the cells and promoted cell proliferation, with the difference being statistically significant (P<0.05). According to the RT-qPCR results, compared with those of the control group, the COS intervention up-regulated the mRNA levels of TLR4 and IFN-γ in RAW264.7 cells, while it inhibited the mRNA expression levels of TGF-ß and IL-10, with the most prominent effect being observed in the 4 mg/mL COS group (P<0.05). 2) Laser scanning confocal microscopy revealed that the amount of fluorescent dye released from lysosomes into the cells was greater in the Calcein+COS group than that in the Calcein group. In other words, a greater amount of fluorescent dye was released from lysosomes into the cells under COS intervention. Furthermore, this process could be blocked by bafilomycin A1. 3) The zeta potential results showed that COS could successfully encapsulate the surface of bacteria when its mass concentration reached 2 mg/mL. Before and after the vaccine strain was encapsulated by COS, the phagocytosis of LM∆E6E7 by RAW264.7 cells was 5.70% and 22.00%, respectively, showing statistically significant differences (P<0.05); the phagocytosis of LI∆E6E7 by RAW264.7 cells was 1.55% and 6.12%, respectively, showing statistically significant differences (P<0.05). Conclusion: COS has the effect of activating the immune response of macrophages and triggering lysosomal escape. The candidates strains of coated live attenuated bacterial vector vaccines can promote the phagocytosis of bacteria by macrophages. Further research is warranted to develop COS into an adjuvant for bacterial vector vaccine.


Subject(s)
Adjuvants, Immunologic , Bacterial Vaccines , Chitosan , Oligosaccharides , Animals , Mice , RAW 264.7 Cells , Oligosaccharides/pharmacology , Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/immunology , Macrophages/metabolism , Macrophages/immunology , Macrophages/drug effects , Vaccines, Attenuated/immunology , Cytokines/metabolism , Cell Survival/drug effects
18.
Front Immunol ; 15: 1373411, 2024.
Article in English | MEDLINE | ID: mdl-38646535

ABSTRACT

Introduction: Veterinary vaccines against Clostridium perfringens type C need to be tested for absence of toxicity, as mandated by pharmacopoeias worldwide. This toxicity testing is required at multiple manufacturing steps and relies on outdated mouse tests that involve severe animal suffering. Clostridium perfringens type C produces several toxins of which the ß-toxin is the primary component responsible for causing disease. Here, we describe the successful development of a new cell-based in vitro assay that can address the specific toxicity of the ß-toxin. Methods: Development of the cell-based assay followed the principle of in vitro testing developed for Cl. septicum vaccines, which is based on Vero cells. We screened four cell lines and selected the THP-1 cell line, which was shown to be the most specific and sensitive for ß-toxin activity, in combination with a commercially available method to determine cell viability (MTS assay) as a readout. Results: The current animal test is estimated to detect 100 - 1000-fold dilutions of the Cl. perfringens type C non-inactivated antigen. When tested with an active Cl. perfringens type C antigen preparation, derived from a commercial vaccine manufacturing process, our THP-1 cell-based assay was able to detect toxin activity from undiluted to over 10000-fold dilution, showing a linear range between approximately 1000- and 10000-fold dilutions. Assay specificity for the ß-toxin was confirmed with neutralizing antibodies and lack of reaction to Cl. perfringens culture medium. In addition, assay parameters demonstrated good repeatability. Conclusions: Here, we have shown proof of concept for a THP-1 cell-based assay for toxicity testing of veterinary Cl. perfringens type C vaccines that is suitable for all vaccine production steps. This result represents a significant step towards the replacement of animal-based toxicity testing of this veterinary clostridial antigen. As a next step, assessment of the assay's sensitivity and repeatability and validation of the method will have to be performed in a commercial manufacturing context in order to formally implement the assay in vaccine quality control.


Subject(s)
Bacterial Toxins , Clostridium perfringens , Animals , Clostridium perfringens/immunology , Bacterial Toxins/immunology , Bacterial Toxins/toxicity , Humans , Vero Cells , Chlorocebus aethiops , Toxicity Tests/methods , Clostridium Infections/veterinary , Clostridium Infections/immunology , Clostridium Infections/diagnosis , THP-1 Cells , Mice , Cell Survival/drug effects , Cell Line , Bacterial Vaccines/immunology , Animal Testing Alternatives/methods
19.
Curr Microbiol ; 81(5): 125, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38558085

ABSTRACT

More than half of the world's population is infected with Helicobacter pylori (H. pylori), which may lead to chronic gastritis, peptic ulcers, and stomach cancer. LeoA, a conserved antigen of H. pylori, aids in preventing this infection by triggering specific CD3+ T-cell responses. In this study, recombinant plasmids containing the LeoA gene of H. pylori are created and conjugated with chitosan nanoparticle (CSNP) to immunize BALB/c mice against the H. pylori infection. We used the online Vaxign tool to analyze the genomes of five distinct strains of H. pylori, and we chose the outer membrane as a prospective vaccine candidate. Afterward, the proteins' immunogenicity was evaluated. The DNA vaccine was constructed and then encapsulated in CSNPs. The effectiveness of the vaccine's immunoprotective effects was evaluated in BALB/c mice. Purified activated splenic CD3+ T cells are used to test the anticancer effects in vitro. Nanovaccines had apparent spherical forms, were small (mean size, 150-250 nm), and positively charged (41.3 ± 3.11 mV). A consistently delayed release pattern and an entrapment efficiency (73.35 ± 3.48%) could be established. Compared to the non-encapsulated DNA vaccine, vaccinated BALB/c mice produced higher amounts of LeoA-specific IgG in plasma and TNF-α in splenocyte lysate. Moreover, BALB/c mice inoculated with nanovaccine demonstrated considerable immunity (87.5%) against the H. pylori challenge and reduced stomach injury and bacterial burdens in the stomach. The immunological state in individuals with GC with chronic infection with H. pylori is mimicked by the H. pylori DNA nanovaccines by inducing a shift from Th1 to Th2 in the response. In vitro human GC cell development is inhibited by activated CD3+ T lymphocytes. According to our findings, the H. pylori vaccine-activated CD3+ has potential immunotherapeutic benefits.


Subject(s)
Chitosan , Helicobacter Infections , Helicobacter pylori , Nanoparticles , Vaccines, DNA , Humans , Animals , Mice , Helicobacter pylori/genetics , Vaccines, DNA/genetics , DNA , Vaccination , Helicobacter Infections/prevention & control , Helicobacter Infections/microbiology , Bacterial Vaccines/genetics , Mice, Inbred BALB C , Antibodies, Bacterial
20.
Vaccine ; 42(12): 3075-3083, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38584060

ABSTRACT

As the major outer membrane protein (OMP) presents in the Pasteurella multocida envelope, OmpH was frequently expressed for laboratory assessments of its immunogenicity against P. multocida infections, but the results are not good. In this study, we modified OmpH with dendritic cell targeting peptide (Depeps) and/or Salmonella FliCd flagellin, and expressed three types of recombinant proteins with the MBP tag (rDepeps-FliC-OmpH-MBP, rDepeps-OmpH-MBP, rFliC-OmpH-MBP). Assessments in mouse models revealed that vaccination with rDepeps-FliC-OmpH-MBP, rDepeps-OmpH-MBP, or rFliC-OmpH-MBP induced significant higher level of antibodies as well as IFN-γ and IL-4 in murine sera than vaccination with rOmpH-MBP (P < 0.5). Vaccination with the three modified proteins also provided increased protection (rDepeps-FliC-OmpH-MBP, 70 %; rDepeps-OmpH-MBP, 50 %; rFliC-OmpH-MBP, 60 %) against P. multocida serotype D compared to vaccination with rOmpH-MBP (30 %). In mice vaccinated with different types of modified OmpHs, a significantly decreased bacterial strains were recovered from bloods, lungs, and spleens compared to rOmpH-MBP-vaccinated mice (P < 0.5). Notably, our assessments also demonstrated that vaccination with rDepeps-FliC-OmpH-MBP provided good protection against infections caused by a heterogeneous group of P. multocida serotypes (A, B, D). Our above findings indicate that modification with DCpep and Salmonella flagellin could be used as a promising strategy to improve vaccine effectiveness.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Animals , Mice , Serogroup , Pasteurella Infections/prevention & control , Flagellin/metabolism , Bacterial Outer Membrane Proteins , Peptides/metabolism , Dendritic Cells , Bacterial Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL
...