Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.669
Filter
1.
Front Cell Infect Microbiol ; 14: 1455259, 2024.
Article in English | MEDLINE | ID: mdl-39228894

ABSTRACT

Introduction: Tularemia, caused by the bacterium Francisella tularensis, poses health risks to humans and can spread through a variety of routes. It has also been classified as a Tier 1 Select agent by the CDC, highlighting its potential as a bioterrorism agent. Moreover, it is difficult to diagnose in a timely fashion, owing to the non-specific nature of tularemia infections. Rapid, sensitive, and accurate detection methods are required to reduce mortality rates. We aimed to develop antibodies directed against the outer membrane protein A of F. tularensis (FopA) for rapid and accurate diagnosis of tularemia. Methods: We used a baculovirus insect cell expression vector system to produce the FopA antigen and generate anti-FopA antibodies through immunization of BALB/c mice. We then employed hybridoma and phage display technologies to screen for antibodies that could recognize unique epitopes on FopA. Result: Two monoclonal antibodies, 6B12 and 3C1, identified through phage display screening specifically bound to recombinant FopA in a dose-dependent manner. The binding affinity of the anti-FopA 6B12 and 3C1 antibodies was observed to have an equilibrium dissociation constant of 1.76 × 10-10 M and 1.32 × 10-9 M, respectively. These antibodies were used to develop a sandwich ELISA system for the diagnosis of tularemia. This assay was found to be highly specific and sensitive, with detection limits ranging from 0.062 ng/mL in PBS to 0.064 ng/mL in skim milk matrices. Discussion: Our findings demonstrate the feasibility of a novel diagnostic approach for detecting F. tularensis based on targeting FopA, as opposed to existing tests that target the bacterial lipopolysaccharide.


Subject(s)
Antibodies, Bacterial , Antibodies, Monoclonal , Bacterial Outer Membrane Proteins , Francisella tularensis , Mice, Inbred BALB C , Recombinant Proteins , Tularemia , Tularemia/diagnosis , Animals , Francisella tularensis/immunology , Francisella tularensis/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Antibodies, Monoclonal/immunology , Mice , Immunoassay/methods , Sensitivity and Specificity , Female , Cell Surface Display Techniques , Epitopes/immunology , Enzyme-Linked Immunosorbent Assay/methods , Humans , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Hybridomas , Baculoviridae/genetics
2.
Microb Pathog ; 195: 106909, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39218373

ABSTRACT

Brucellosis is a zoonotic disease caused by Brucella, which is difficult to eliminate by conventional drugs. Therefore, a novel multi-epitope vaccine (MEV) was designed to prevent human Brucella infection. Based on the method of "reverse vaccinology", cytotoxic T lymphocyte epitopes (CTLEs), helper T lymphocyte epitopes (HTLEs), linear B-cell epitopes (LBEs) and conformational B-cell epitopes (CBEs) of four Brucella proteins (VirB9, VirB10, Omp 19 and Omp 25) were obtained. In order to keep the correct protein folding, the multiple epitopes was constructed by connecting epitopes through linkers. In view of the significant connection between human leukocyte antigen CTLA-4 and B7 molecules found on antigen presenting cells (APCs), a new vaccine (V_C4MEV) for preventing brucellosis was created by combining CTLA-4 immunoglobulin variable region (IgV_CTLA-4) with MEV protein. Immunoinformatics analysis showed that V_C4MEV has a good secondary and tertiary structure. Additionally, molecular docking and molecular dynamics simulation (MD) revealed a robust binding affinity between IgV_ CTLA-4 and the B7 molecule. Notably, the vaccine V_C4MEV was demonstrated favorable immunogenicity and antigenicity in both in vitro and in vivo experiments. V_C4MEV had the potential to activate defensive cells and immune responses, offering a hopeful approach for developing vaccines against Brucella in the upcoming years.


Subject(s)
Brucella Vaccine , Brucella , Brucellosis , CTLA-4 Antigen , Computational Biology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Molecular Docking Simulation , Molecular Dynamics Simulation , Brucellosis/prevention & control , Brucellosis/immunology , Epitopes, B-Lymphocyte/immunology , CTLA-4 Antigen/immunology , Epitopes, T-Lymphocyte/immunology , Brucella Vaccine/immunology , Animals , Humans , Brucella/immunology , Brucella/genetics , Mice , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Immunoinformatics , Lipoproteins
3.
Virulence ; 15(1): 2395831, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39185619

ABSTRACT

Salmonella enterica serovar Typhi (S. Typhi) causes typhoid fever, a systemic infection that affects millions of people worldwide. S. Typhi can invade and survive within host cells, such as intestinal epithelial cells and macrophages, by modulating their immune responses. However, the immunomodulatory capability of S. Typhi in relation to TolC-facilitated efflux pump function remains unclear. The role of TolC, an outer membrane protein that facilitates efflux pump function, in the invasion and immunomodulation of S. Typhi, was studied in human intestinal epithelial cells and macrophages. The tolC deletion mutant of S. Typhi was compared with the wild-type and its complemented strain in terms of their ability to invade epithelial cells, survive and induce cytotoxicity in macrophages, and elicit proinflammatory cytokine production in macrophages. The tolC mutant, which has a defective outer membrane, was impaired in invading epithelial cells compared to the wild-type strain, but the intracellular presence of the tolC mutant exhibited greater cytotoxicity and induced higher levels of proinflammatory cytokines (IL-1ß and IL-8) in macrophages compared to the wild-type strain. These effects were reversed by complementing the tolC mutant with a functional tolC gene. Our results suggest that TolC plays a role in S. Typhi to efficiently invade epithelial cells and suppress host immune responses during infection. TolC may be a potential target for the development of novel therapeutics against typhoid fever.


Subject(s)
Bacterial Outer Membrane Proteins , Epithelial Cells , Macrophages , Salmonella typhi , Typhoid Fever , Salmonella typhi/pathogenicity , Salmonella typhi/immunology , Salmonella typhi/genetics , Humans , Macrophages/microbiology , Macrophages/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/immunology , Epithelial Cells/microbiology , Epithelial Cells/immunology , Typhoid Fever/immunology , Typhoid Fever/microbiology , Immunomodulation , Cytokines/metabolism , Cytokines/immunology , Microbial Viability , Interleukin-8/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , Cell Line
4.
Methods Mol Biol ; 2843: 195-216, 2024.
Article in English | MEDLINE | ID: mdl-39141302

ABSTRACT

Engineered outer membrane vesicles (OMVs) derived from Gram-negative bacteria are a promising vaccine technology for developing immunity against diverse pathogens. However, antigen display on OMVs can be challenging to control and highly variable due to bottlenecks in protein expression and localization to the bacterial host cell's outer membrane, especially for bulky and complex antigens. Here, we describe methods related to a universal vaccine technology called AvidVax (avidin-based vaccine antigen crosslinking) for rapid and simplified assembly of antigens on the exterior of OMVs during vaccine development. The AvidVax platform involves remodeling the OMV surface with multiple copies of a synthetic antigen-binding protein (SNAP), which is an engineered fusion protein comprised of an outer membrane scaffold protein linked to a biotin-binding protein. The resulting SNAPs enable efficient decoration of OMVs with a molecularly diverse array of biotinylated subunit antigens, including globular and membrane proteins, glycans and glycoconjugates, haptens, lipids, nucleic acids, and short peptides. We detail the key steps in the AvidVax vaccine production pipeline including preparation and isolation of SNAP-OMVs, biotinylation and enrichment of vaccine antigens, and formulation and characterization of antigen-loaded SNAP-OMVs.


Subject(s)
Antigens, Bacterial , Biotinylation , Extracellular Vesicles , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Vaccines/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Vaccine Development , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/immunology
5.
mSphere ; 9(9): e0033024, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39158304

ABSTRACT

Outer membrane vesicles (OMVs) from Gram-negative bacteria can be used as a vaccine platform to deliver heterologous antigens. Here, the major protective antigens of Yersinia pestis, F1 and LcrV, were fused either with the leader sequence or the transmembrane domain of the outer membrane protein A (OmpA), resulting in chimeric proteins OmpA-ls-F1V and OmpA46-159-F1V, respectively. We show that OmpA-ls-F1V and OmpA46-159-F1V can be successfully delivered into the lumen and membrane of the OMVs of Escherichia coli, respectively. Mutation of ompA but not tolR in E. coli enhanced the delivery efficiency of OmpA-ls-F1V into OMVs. The OmpA-ls-F1V protein comprises up to 20% of the total protein in OMVs derived from the ompA mutant (OMVdA-ALS-F1V), a proportion significantly higher than the 1% observed for OmpA46-159-F1V in OMVs produced by an ompA mutant that expresses OmpA46-159-F1V, referred to as OMVdA-LATM5-F1V. Intramuscular (i.m.) immunization of mice with OMVdA-ALS-F1V induced significantly higher levels of serum anti-LcrV and anti-F1 IgG, and provided higher efficacy in protection against subcutaneous (s.c.) Y. pestis infection compared to OMVdA-LATM5-F1V and the purified recombinant F1V (rF1V) protein adsorbed to aluminum hydroxide. The three-dose i.m. immunization with OMVdA-ALS-F1V, administered at 14-day intervals, provides complete protection to mice against s.c. infection with 130 LD50 of Y. pestis 201 and conferred 80% against intranasal (i.n.) challenge with 11.4 LD50 of Y. pestis 201. Taken together, our findings indicate that the engineered OMVs containing F1V fused with the leader sequence of OmpA provide significantly higher protection than rF1V against both s.c. and i.n. infection of Y. pestis and more balanced Th1/Th2 responses.IMPORTANCEThe two major protective antigens of Y. pestis, LcrV and F1, have demonstrated the ability to elicit systemic and local mucosal immune responses as subunit vaccines. However, these vaccines have failed to provide adequate protection against pneumonic plague in African green monkeys. Here, Y. pestis F1 and LcrV antigens were successfully incorporated into the lumen and the surface of the outer membrane vesicles (OMVs) of E. coli by fusion either with the leader sequence or the transmembrane domain of OmpA. We compared the humoral immune response elicited by these OMV formulations and their protective efficacy in mice against Y. pestis. Our results demonstrate that the plague OMV vaccine candidates can induce robust protective immunity against both s.c. and i.n. Y. pestis infections, surpassing the effectiveness of rF1V. In addition, immunization with OMVs generated a relatively balanced Th1/Th2 immune response compared to rF1V immunization. These findings underscore the potential of OMVs-based plague vaccines for further development.


Subject(s)
Antibodies, Bacterial , Antigens, Bacterial , Bacterial Outer Membrane Proteins , Escherichia coli , Plague Vaccine , Plague , Pore Forming Cytotoxic Proteins , Yersinia pestis , Animals , Plague/prevention & control , Plague/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Escherichia coli/genetics , Escherichia coli/immunology , Yersinia pestis/immunology , Yersinia pestis/genetics , Mice , Pore Forming Cytotoxic Proteins/immunology , Pore Forming Cytotoxic Proteins/genetics , Plague Vaccine/immunology , Plague Vaccine/administration & dosage , Plague Vaccine/genetics , Female , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Bacterial Outer Membrane/immunology , Bacterial Proteins
6.
J Infect Dis ; 230(Supplement_1): S82-S86, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39140718

ABSTRACT

Lyme disease is caused by the spirochete, Borrelia burgdorferi, which is transmitted by Ixodes spp ticks. The rise in Lyme disease cases since its discovery in the 1970s has reinforced the need for a vaccine. A vaccine based on B burgdorferi outer surface protein A (OspA) was approved by the Food and Drug Administration (FDA) several decades ago, but was pulled from the market a few years later, reportedly due to poor sales, despite multiple organizations concluding that it was safe and effective. Newer OspA-based vaccines are being developed and are likely to be available in the coming years. More recently, there has been a push to develop vaccines that target the tick vector instead of the pathogen to inhibit tick feeding and thus prevent transmission of tick-borne pathogens to humans and wildlife reservoirs. This review outlines the history of Lyme disease vaccines and this movement to anti-tick vaccine approaches.


Subject(s)
Borrelia burgdorferi , Ixodes , Lyme Disease Vaccines , Lyme Disease , Lyme Disease/prevention & control , Lyme Disease/immunology , Humans , Animals , Borrelia burgdorferi/immunology , Lyme Disease Vaccines/immunology , Ixodes/microbiology , Vaccination , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Antigens, Surface/immunology , Lipoproteins/immunology
7.
Vaccine ; 42(22): 126205, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39141987

ABSTRACT

BACKGROUND: Group A Streptococcus (Strep A) causes both uncomplicated and severe invasive infections, as well as the post-infection complications acute rheumatic fever and rheumatic heart disease. Despite the high global burden of disease resulting from Strep A infections, there is not a licensed vaccine. A 30-valent M protein-based vaccine has previously been shown to be immunogenic in animal models and in a Phase I clinical trial (NCT02564237). Here, we assessed the immunogenicity of a 30-valent messenger (m)RNA vaccine designed to express the same M peptide targets as the 30-valent protein vaccine and compared it with the protein vaccine. METHODS: Female New Zealand white rabbits were immunized with one of four vaccine formulations (3 doses of each formulation at days 1, 28, and 56): soluble mRNA (100 µg/animal), C-terminal transmembrane mRNA (100 µg/animal), protein vaccine (400 µg/animal), or a non-translatable RNA control (100 µg/animal). Serum was collected one day prior to the first dose and on days 42 and 70. Rabbit serum samples were assayed for antibody levels against synthetic M peptides by ELISA. HL-60 opsonophagocytic killing (OPK) assays were performed to assess functional antibody levels. RESULTS: Serum IgG levels were similar for the mRNA and protein vaccines. The CtTM version of the mRNA vaccine elicited slightly higher antibody levels than the mRNA designed to express soluble proteins. OPK activity was similar for the mRNA and protein vaccines, regardless of M type. CONCLUSIONS: The total antibody responses and functional antibody levels elicited by the 30-valent mRNA Strep A vaccines were similar to those observed following immunization with the analogous protein vaccine. The mRNA vaccine platform provides potential advantages to protein-based vaccines including inherent adjuvant activity, increased production efficiency, lower cost, and the potential to rapidly change epitopes/peptides, all of which are important considerations related to multivalent Strep A vaccine development.


Subject(s)
Antibodies, Bacterial , Antigens, Bacterial , Streptococcal Infections , Streptococcal Vaccines , Streptococcus pyogenes , Animals , Female , Rabbits , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/genetics , Immunogenicity, Vaccine , RNA, Messenger/genetics , RNA, Messenger/immunology , Streptococcal Infections/prevention & control , Streptococcal Infections/immunology , Streptococcal Vaccines/immunology , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/genetics , Streptococcus pyogenes/immunology , Streptococcus pyogenes/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Clinical Trials, Phase I as Topic
8.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000557

ABSTRACT

The effects of intestinal microflora on extraintestinal immune response by intestinal cytokines and metabolites have been documented, but whether intestinal microbes stimulate serum antibody generation is unknown. Here, serum antibodies against 69 outer membrane proteins of Escherichia coli, a dominant bacterium in the human intestine, are detected in 141 healthy individuals of varying ages. Antibodies against E. coli outer membrane proteins are determined in all serum samples tested, and frequencies of antibodies to five outer membrane proteins (OmpA, OmpX, TsX, HlpA, and FepA) are close to 100%. Serum antibodies against E. coli outer membrane proteins are further validated by Western blot and bacterial pull-down. Moreover, the present study shows that OstA, HlpA, Tsx, NlpB, OmpC, YfcU, and OmpA provide specific immune protection against pathogenic E. coli, while HlpA and OmpA also exhibit cross-protection against Staphylococcus aureus infection. These finding indicate that intestinal E. coli activate extraintestinal antibody responses and provide anti-infective immunity.


Subject(s)
Antibodies, Bacterial , Bacterial Outer Membrane Proteins , Escherichia coli , Humans , Escherichia coli/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/immunology , Adult , Female , Staphylococcus aureus/immunology , Male , Antibody Formation/immunology , Middle Aged , Escherichia coli Proteins/immunology , Young Adult , Aged , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Adolescent , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology
9.
Scand J Immunol ; 99(4): e13353, 2024 Apr.
Article in English | MEDLINE | ID: mdl-39007994

ABSTRACT

Lyme borreliosis, caused by Borrelia burgdorferi sensu lato, is the most common tickborne disease. Its neuronal form, neuroborreliosis, comprises 3 to 38% of borreliosis cases in Europe. Borrelia outer surface proteins and virulence factors, OspE and BBK32, have been previously reported to help cause infection by promoting attachment to human host epithelial cells and evading complement attack. We assessed the serological responses to BBK32 and OspE in 19 individuals diagnosed with neuroborreliosis to see whether antibodies that could both target the bacteria and neutralize the virulence mechanisms on the microbial surface emerge. Results evaluate levels of total protein, IgG and the chemokine CXCL13, a determinant for B-cell recruitment during neuroinflammation, in patients' cerebrospinal fluid samples. Antibody levels against BBK32 and OspE correlated with those against VlsE, a well-characterized diagnostic serological marker of the disease. A dual serological profile of the patients was observed. K-means clustering split the cohort into two discrete groups presenting distinct serological and CNS responses. One group contained young patients with low levels of anti-BBK32 and OspE antibodies. The other group showed stronger responses, possibly following prolonged infections or reinfections. Additionally, we assessed anti-ganglioside antibodies that could cause autoimmunity or complement dysregulation but observed that they did not correlate with neuroborreliosis in our patient cohort. The dual nature of antibody responses against the virulence factors BBK32 and OspE in neuroborreliosis patients may suggest the necessity of repeated exposures for efficient immune responses. Better protection could be achieved if the virulence factors were formulated into vaccines.


Subject(s)
Antibodies, Bacterial , Antigens, Bacterial , Bacterial Outer Membrane Proteins , Borrelia burgdorferi , Lyme Neuroborreliosis , Humans , Lyme Neuroborreliosis/immunology , Lyme Neuroborreliosis/blood , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Middle Aged , Female , Male , Adult , Aged , Borrelia burgdorferi/immunology , Antigens, Bacterial/immunology , Virulence Factors/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Chemokine CXCL13/blood , Chemokine CXCL13/immunology , Bacterial Proteins/immunology , Antibody Formation/immunology
10.
Microb Biotechnol ; 17(7): e14522, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39016683

ABSTRACT

As a Gram-negative anaerobic bacterium, Akkermansia muciniphila (AKK) participates in the immune response in many cancers. Our study focused on the factors and molecular mechanisms of AKK affecting immune escape in lung adenocarcinoma (LUAD). We cultured AKK bacteria, prepared AKK outer membrane protein Amuc_1100 and constructed a subcutaneous graft tumour mouse model. A549, NCI-H1395 cells and mice were respectively treated with inactivated AKK, Amuc_1100, Ruxolitinib (JAK inhibitor) and RO8191 (JAK activator). CD8+ T cells that penetrated the membrane were counted in the Transwell assay. The toxicity of CD8+ T cells was evaluated by lactate dehydrogenase assay. Western blot was applied to determine JAK/STAT-related protein and PD-L1 expression, whilst CCL5, granzyme B and INF-γ expression were assessed through enzyme-linked immunosorbent assay (ELISA). The proportion of tumour-infiltrating CD8+ T cells and the levels of granzyme B and INF-γ were determined by flow cytometry. AKK markedly accelerated A549 and NCI-H1395 recruiting CD8+ T cells and enhanced CD8+ T cell toxicity. Amuc_1100 purified from AKK exerted the same promoting effects. Besides, Amuc_1100 dramatically suppressed PD-L1, p-STAT and p-JAK expression and enhanced CCL5, granzyme B and INF-γ expression. Treatment with Ruxolitinib accelerated A549 and NCI-H1395 cells recruiting CD8+ T cells, enhanced CD8+ T cell toxicity, CCL5, granzyme B and INF-γ expression, and inhibited PD-L1 expression. In contrast, the RO8191 treatment slowed down the changes induced by Amuc_1100. Animal experiments showed that Amuc_1100 was found to increase the number of tumour-infiltrating CD8+ T cells, increase the levels of granzyme B and INF-γ and significantly inhibit the expression of PD-L1, p-STAT and p-JAK, which exerted an antitumour effect in vivo. In conclusion, through inhibiting the JAK/STAT signalling pathway, AKK outer membrane protein facilitated the recruitment of CD8+ T cells in LUAD and suppressed the immune escape of cells.


Subject(s)
Adenocarcinoma of Lung , Akkermansia , Bacterial Outer Membrane Proteins , CD8-Positive T-Lymphocytes , Janus Kinases , Signal Transduction , CD8-Positive T-Lymphocytes/immunology , Animals , Mice , Humans , Janus Kinases/metabolism , Adenocarcinoma of Lung/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/immunology , Lung Neoplasms/immunology , Cell Line, Tumor , STAT Transcription Factors/metabolism , Disease Models, Animal
11.
Appl Microbiol Biotechnol ; 108(1): 424, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037584

ABSTRACT

Leptospirosis, a neglected zoonotic disease, is caused by pathogenic spirochetes belonging to the genus Leptospira and has one of the highest morbidity and mortality rates worldwide. Vaccination stands out as one of the most effective preventive measures for susceptible populations. Within the outer membrane of Leptospira spp., we find the LIC12287, LIC11711, and LIC13259 lipoproteins. These are of interest due to their surface location and potential immunogenicity. Thorough examination revealed the conservation of these proteins among pathogenic Leptospira spp.; we mapped the distribution of T- and B-cell epitopes along their sequences and assessed the 3D structures of each protein. This information aided in selecting immunodominant regions for the development of a chimeric protein. Through gene synthesis, we successfully constructed a chimeric protein, which was subsequently expressed, purified, and characterized. Hamsters were immunized with the chimeric lipoprotein, formulated with adjuvants aluminum hydroxide, EMULSIGEN®-D, Sigma Adjuvant System®, and Montanide™ ISA206VG. Another group was vaccinated with an inactivated Escherichia coli bacterin expressing the chimeric protein. Following vaccination, hamsters were challenged with a virulent L. interrogans strain. Our evaluation of the humoral immune response revealed the production of IgG antibodies, detectable 28 days after the second dose, in contrast to pre-immune samples and control groups. This demonstrates the potential of the chimeric protein to elicit a robust humoral immune response; however, no protection against challenge was achieved. While this study provides valuable insights into the subject, further research is warranted to identify protective antigens that could be utilized in the development of a leptospirosis vaccine. KEY POINTS: • Several T- and B-cell epitopes were identified in all the three proteins. • Four different adjuvants were used in vaccine formulations. • Immunization stimulated significant levels of IgG2/3 in vaccinated animals.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Leptospirosis , Lipoproteins , Animals , Leptospirosis/prevention & control , Leptospirosis/immunology , Lipoproteins/immunology , Lipoproteins/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cricetinae , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Adjuvants, Immunologic/administration & dosage , Immunoglobulin G/blood , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Leptospira interrogans/immunology , Leptospira interrogans/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Vaccination , Immunity, Humoral , Leptospira/immunology , Leptospira/genetics , Immunogenicity, Vaccine
12.
J Microbiol Biotechnol ; 34(7): 1419-1424, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-38955797

ABSTRACT

Secretin PilQ is an antigenically conserved outer membrane protein that is present in most meningococci and PorA is a major protein that elicits bactericidal immune response in humans following natural disease and immunization. In the present study, BALB/c mice were immunized subcutaneously with rPilQ406-770 or rPorA together with Freund's adjuvant (FA). Serum antibody responses to serogroup A and B Neisseria meningitides whole cells or purified proteins and functional activity of antibodies were determined by ELISA and serum bactericidal assay (SBA), respectively. Serum IgG responses were significantly increased in the immunized group with rPilQ406-770 or rPorA together with FA compared to control groups. IgG antibody response of mice immunized with rPilQ406-770 was significantly more than mice immunized with rPorA (OD at 450 nm was 1.6 versus 0.83). The booster injections were effective in increasing the responses of anti-rPilQ406-770 or anti-rPorA IgG significantly. Antisera produced against rPilQ406-770 or rPorA demonstrated strong surface reactivity to serogroup B N. meningitides in comparison with control groups. Antisera raised against rPorA or rPilQ406-770 and FA demonstrated SBA titers from 1/1024 to 1/2048 against serogroup B. The strongest bactericidal activity was detected in sera from mice immunized with rPilQ406-770 mixed with FA. These results suggest that rPilQ406-770 is a potential vaccine candidate for serogroup B N. meningitidis.


Subject(s)
Antibodies, Bacterial , Bacterial Outer Membrane Proteins , Immunoglobulin G , Meningococcal Vaccines , Mice, Inbred BALB C , Recombinant Proteins , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Recombinant Proteins/immunology , Meningococcal Vaccines/immunology , Meningococcal Vaccines/administration & dosage , Neisseria meningitidis/immunology , Female , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Antibody Formation/immunology , Immunization , Enzyme-Linked Immunosorbent Assay , Serum Bactericidal Antibody Assay , Antigens, Bacterial/immunology
13.
Nanotechnology ; 35(39)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38917779

ABSTRACT

Safe and effective vaccine candidates are needed to address the limitations of existing vaccines against Brucellosis, a disease responsible for substantial economic losses in livestock. The present study aimed to encapsulate recombinant Omp25 and EipB proteins, knowledged antigen properties, into PLGA nanoparticles, characterize synthesized nanoparticles with different methods, and assessed theirin vitro/in vivoimmunostimulatory activities to develop new vaccine candidates. The recombinant Omp25 and EipB proteins produced with recombinant DNA technology were encapsulated into PLGA nanoparticles by double emulsion solvent evaporation technique. The nanoparticles were characterized using FE-SEM, Zeta-sizer, and FT-IR instruments to determine size, morphology, zeta potentials, and polydispersity index values, as well as to analyze functional groups chemically. Additionally, the release profiles and encapsulation efficiencies were assessed using UV-Vis spectroscopy. After loading with recombinant proteins, O-NPs reached sizes of 221.2 ± 5.21 nm, while E-NPs reached sizes of 274.4 ± 9.51 nm. The cumulative release rates of the antigens, monitored until the end of day 14, were determined to be 90.39% for O-NPs and 56.1% for E-NPs. Following the assessment of thein vitrocytotoxicity and immunostimulatory effects of both proteins and nanoparticles on the J774 murine macrophage cells,in vivoimmunization experiments were conducted using concentrations of 16µg ml-1for each protein. Both free antigens and antigen-containing nanoparticles excessively induced humoral immunity by increasing producedBrucella-specific IgG antibody levels for 3 times in contrast to control. Furthermore, it was also demonstrated that vaccine candidates stimulated Th1-mediated cellular immunity as well since they significantly raised IFN-gamma and IL-12 cytokine levels in murine splenocytes rather than IL-4 following to immunization. Additionally, the vaccine candidates conferred higher than 90% protection from the infection according to challenge results. Our findings reveal that PLGA nanoparticles constructed with the encapsulation of recombinant Omp25 or EipB proteins possess great potential to triggerBrucella-specific humoral and cellular immune response.


Subject(s)
Brucellosis , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Recombinant Proteins , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Brucellosis/prevention & control , Brucellosis/immunology , Mice , Nanoparticles/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/chemistry , Mice, Inbred BALB C , Female , Brucella Vaccine/immunology , Brucella Vaccine/genetics , Brucella Vaccine/administration & dosage , Brucella abortus/immunology , Brucella abortus/genetics , Drug Carriers/chemistry , Nanovaccines
14.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38862192

ABSTRACT

To begin to optimize the immunization routes for our reported PLGA-rMOMP nanovaccine [PLGA-encapsulated Chlamydia muridarum (Cm) recombinant major outer membrane protein (rMOMP)], we compared two prime-boost immunization strategies [subcutaneous (SC) and intramuscular (IM-p) prime routes followed by two SC-boosts)] to evaluate the nanovaccine-induced protective efficacy and immunogenicity in female BALB/c mice. Our results showed that mice immunized via the SC and IM-p routes were protected against a Cm genital challenge by a reduction in bacterial burden and with fewer bacteria in the SC mice. Protection of mice correlated with rMOMP-specific Th1 (IL-2 and IFN-γ) and not Th2 (IL-4, IL-9, and IL-13) cytokines, and CD4+ memory (CD44highCD62Lhigh) T-cells, especially in the SC mice. We also observed higher levels of IL-1α, IL-6, IL-17, CCL-2, and G-CSF in SC-immunized mice. Notably, an increase of cytokines/chemokines was seen after the challenge in the SC, IM-p, and control mice (rMOMP and PBS), suggesting a Cm stimulation. In parallel, rMOMP-specific Th1 (IgG2a and IgG2b) and Th2 (IgG1) serum, mucosal, serum avidity, and neutralizing antibodies were more elevated in SC than in IM-p mice. Overall, the homologous SC prime-boost immunization of mice induced enhanced cellular and antibody responses with better protection against a genital challenge compared to the heterologous IM-p.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Chlamydia Infections , Chlamydia muridarum , Cytokines , Mice, Inbred BALB C , Animals , Female , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Chlamydia muridarum/immunology , Cytokines/metabolism , Chlamydia Infections/prevention & control , Chlamydia Infections/immunology , Mice , Antibodies, Bacterial/blood , Injections, Intramuscular , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Immunization, Secondary , Disease Models, Animal , Immunogenicity, Vaccine , Injections, Subcutaneous , Nanoparticles/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/administration & dosage , Vaccine Efficacy , Th1 Cells/immunology , Nanovaccines
15.
Vaccine ; 42(23): 126047, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-38897891

ABSTRACT

Chlamydia is an obligate intracellular bacterial pathogen responsible for disease and infertility across multiple species. Currently vaccines are being studied to help reduce the prevalence of this disease. The main advantage of protein subunit vaccines is their high degree of safety although this is traded off with the requirement for multiple booster doses to achieve complete protection. Although in certain populations the booster dose can be difficult and costly to administer, development of delayed vaccine delivery techniques, such as a vaccine capsule, could be the solution to this problem. One of the main drawbacks in this technology is that the antigen must remain stable at body temperature (37 °C) until release is achieved. Here we elucidate the stability of a recombinant chlamydial major outer membrane protein (MOMP) antigen and assess its antigenic and immunogenic properties after subjecting the antigen to 37 °C for four to six weeks. Through in vitro and in vivo assessment we found that the aged chlamydial MOMP was able to produce equivalent humoral and cell-mediated immune responses when compared with the unaged vaccine. It was also found that vaccines formulated with the aged antigen conferred equivalent protection against a live infection challenge as the unaged antigen. Thus ageing chlamydial MOMP antigens at 37 °C for four to six weeks did not cause any significant structural or antigenic/immunogenic degradation and recombinant C. muridarum MOMP is suitable for use in a delayed vaccine delivery system.


Subject(s)
Antibodies, Bacterial , Antigens, Bacterial , Bacterial Outer Membrane Proteins , Bacterial Vaccines , Chlamydia Infections , Chlamydia muridarum , Chlamydia muridarum/immunology , Animals , Antigens, Bacterial/immunology , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Female , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Mice , Body Temperature , Mice, Inbred BALB C , Protein Stability , Immunity, Cellular , Temperature , Recombinant Proteins/immunology
16.
Vaccine ; 42(24): 126084, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-38937181

ABSTRACT

Lyme disease, caused by the bacterium Borrelia burgdorferi, is the most common tick-borne illness in the United States. Despite the rise in Lyme disease incidence, there is no vaccine against B. burgdorferi approved for human use. Little is known about the immune correlates of protection needed to prevent Lyme disease. In this work, a mouse model was used to characterize the immune response and compare the protection provided by two USDA-approved vaccines for use in canines: Duramune (bacterin vaccine) and Vanguard crLyme (subunit vaccine composed of two outer surface proteins, OspA and OspC). C3H/HeNCrl mice were immunized with two doses of either Duramune or Vanguard, and immune responses and protection against B. burgdorferi were assessed in short (35 days) and long-term (120 days) studies. Flow cytometry, ELISPOT detection of antibody-producing cells, and antibody affinity studies were performed to identify correlates of vaccine-mediated protection. Both vaccines induced humoral responses, with high IgG titers against B. burgdorferi. However, the levels of anti-B. burgdorferi antibodies decayed over time in Vanguard-vaccinated mice. While both vaccines triggered the production of antibodies against both OspA and OspC, antibody levels against these proteins were also lower in Vanguard-vaccinated mice 120 days post-vaccination. Both vaccines only provided partial protection against B. burgdorferi at the dose used in this model. The protection provided by Duramune was superior to Vanguard 120 days post-vaccination, and was characterized by higher antibody titers, higher abundance of long-lived plasma cells, and higher avidity antibodies than Vanguard. Overall, these studies provide insights into the importance of the humoral memory response to veterinary vaccines against Lyme disease and will help inform the development of future human vaccines.


Subject(s)
Antibodies, Bacterial , Borrelia burgdorferi , Immunoglobulin G , Immunologic Memory , Lyme Disease Vaccines , Lyme Disease , Mice, Inbred C3H , Animals , Lyme Disease/prevention & control , Lyme Disease/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Borrelia burgdorferi/immunology , Lyme Disease Vaccines/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Bacterial Outer Membrane Proteins/immunology , Mice , Female , Lipoproteins/immunology , Disease Models, Animal , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Antibody Affinity , Antigens, Surface/immunology , Enzyme-Linked Immunospot Assay , Antigens, Bacterial/immunology , Bacterial Vaccines
17.
Lancet Infect Dis ; 24(9): 1045-1058, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38830375

ABSTRACT

BACKGROUND: Rising Lyme borreliosis incidence rates, potential for severe outcomes, and limitations in accurate and timely diagnosis for treatment initiation suggest the need for a preventive vaccine; however, no vaccine is currently available for human use. We performed two studies in adults to optimise the dose level and vaccination schedule for VLA15, an investigational Lyme borreliosis vaccine targeting outer surface protein A (OspA) serotypes 1-6, which are associated with the most common pathogenic Borrelia species in Europe and North America. METHODS: Both randomised, observer-blind, placebo-controlled, multicentre phase 2 studies included participants aged 18-65 years without recent history of Lyme borreliosis or tick bites. Study one was conducted at nine clinical research and study centre sites in the USA (n=6), Germany (n=2), and Belgium (n=1); study two was conducted at five of the study one US sites. Based on a randomisation list created by an unmasked statistician for each study, participants were randomly assigned via an electronic case report form randomisation module to receive 90 µg (study one only), 135 µg, or 180 µg VLA15 or placebo by intramuscular injection at months 0, 1, and 2 (study one) or 0, 2, and 6 (study two). Study one began with a run-in phase to confirm safety, after which the Data Safety Monitoring Board recommended the removal of the 90 µg group and continuation of the study. In the study one run-in phase, randomisation was stratified by study site, whereas in the study one main phase and in study two, randomisation was stratified by study site, age group, and baseline B burgdorferi (sensu lato) serostatus. All individuals were masked, other than staff involved in randomisation, vaccine preparation or administration, or safety data monitoring. The primary endpoint for both studies was OspA-specific IgG geometric mean titres (GMTs) at 1 month after the third vaccination and was evaluated in the per-protocol population. Safety endpoints were evaluated in the safety population: all participants who received at least one vaccination. Both studies are registered at ClinicalTrials.gov (study one NCT03769194 and study two NCT03970733) and are completed. FINDINGS: For study one, 573 participants were screened and randomly assigned to treatment groups between Dec 21, 2018, and Sept, 26, 2019. For study two, 248 participants were screened and randomly assigned between June 26 and Sept 3, 2019. In study one, 29 participants were assigned to receive 90 µg VLA15, 215 to 135 µg, 205 to 180 µg, and 124 to placebo. In study two, 97 participants were assigned to receive 135 µg VLA15, 100 to 180 µg, and 51 to placebo. At 1 month after the third vaccination (ie, month 3), OspA-specific IgG GMTs in study one ranged from 74·3 (serotype 1; 95% CI 46·4-119·0) to 267·4 units per mL (serotype 3; 194·8-367·1) for 90 µg VLA15, 101·9 (serotype 1; 87·1-119·4) to 283·2 units per mL (serotype 3; 248·2-323·1) for 135 µg, and 115·8 (serotype 1; 98·8-135·7) to 308·6 units per mL (serotype 3; 266·8-356·8) for 180 µg. In study two, ranges at 1 month after the third vaccination (ie, month 7) were 278·5 (serotype 1; 214·9-361·0) to 545·2 units per mL (serotype 2; 431·8-688·4) for 135 µg VLA15 and 274·7 (serotype 1; 209·4-360·4) to 596·8 units per mL (serotype 3; 471·9-754·8) for 180 µg. Relative to placebo, the VLA15 groups had more frequent reports of solicited local adverse events (study one: 94%, 95% CI 91-96 vs 26%, 19-34; study two: 96%, 93-98 vs 35%, 24-49 after any vaccination) and solicited systemic adverse events (study one: 69%, 65-73 vs 43%, 34-52; study two: 74%, 67-80 vs 51%, 38-64); most were mild or moderate. In study one, unsolicited adverse events were reported by 52% (48-57) of participants in the VLA15 groups and 52% (43-60) of those in the placebo groups; for study two these were 65% (58-71) and 69% (55-80), respectively. Percentages of participants reporting serious unsolicited adverse events (study one: 2%, 1-4; study two: 4%, 2-7) and adverse events of special interest (study one: 1%, 0-2; study two: 1%, 0-3) were low across all groups. A single severe, possibly related unsolicited adverse event was reported (worsening of pre-existing ventricular extrasystoles, which resolved after change of relevant concomitant medication); no related serious adverse events or deaths were reported. INTERPRETATION: VLA15 was safe, well tolerated, and elicited robust antibody responses to all six OspA serotypes. These findings support further clinical development of VLA15 using the 180 µg dose and 0-2-6-month schedule, which was associated with the greatest immune responses. FUNDING: Valneva.


Subject(s)
Immunization Schedule , Lyme Disease Vaccines , Lyme Disease , Humans , Adult , Male , Middle Aged , Female , Lyme Disease/prevention & control , Young Adult , Lyme Disease Vaccines/immunology , Lyme Disease Vaccines/administration & dosage , Adolescent , Aged , Lipoproteins/immunology , Lipoproteins/administration & dosage , Antibodies, Bacterial/blood , Borrelia burgdorferi/immunology , Belgium , United States , Bacterial Outer Membrane Proteins/immunology , Single-Blind Method , Antigens, Surface/immunology , Antigens, Surface/administration & dosage , Germany , Vaccination/methods , Healthy Volunteers , Bacterial Vaccines
18.
World J Microbiol Biotechnol ; 40(8): 250, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38910219

ABSTRACT

Aeromonas hydrophila, an opportunistic warm water pathogen, has always been a threat to aquaculture, leading to substantial economic losses. Vaccination of the cultured fish would effectively prevent Aeromoniasis, and recent advancements in nanotechnology show promise for efficacious vaccines. Oral delivery would be the most practical and convenient method of vaccine delivery in a grow-out pond. This study studied the immunogenicity and protective efficacy of a nanoparticle-loaded outer membrane protein A from A. hydrophila in the zebrafish model. The protein was over-expressed, purified, and encapsulated using poly lactic-co-glycolic acid (PLGA) nanoparticles via the double emulsion method. The PLGA nanoparticles loaded with recombinant OmpA (rOmpA) exhibited a size of 295 ± 15.1 nm, an encapsulation efficiency of 72.52%, and a polydispersity index of 0.292 ± 0.07. Scanning electron microscopy confirmed the spherical and isolated nature of the PLGA-rOmpA nanoparticles. The protective efficacy in A. hydrophila-infected zebrafish after oral administration of the nanovaccine resulted in relative percentage survival of 77.7. Gene expression studies showed significant upregulation of immune genes in the vaccinated fish. The results demonstrate the usefulness of oral administration of nanovaccine-loaded rOmpA as a potential vaccine since it induced a robust immune response and conferred adequate protection against A. hydrophila in zebrafish, Danio rerio.


Subject(s)
Aeromonas hydrophila , Bacterial Outer Membrane Proteins , Bacterial Vaccines , Fish Diseases , Gram-Negative Bacterial Infections , Nanoparticles , Recombinant Proteins , Zebrafish , Animals , Zebrafish/immunology , Aeromonas hydrophila/immunology , Aeromonas hydrophila/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Fish Diseases/prevention & control , Fish Diseases/immunology , Fish Diseases/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Administration, Oral , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Vaccination , Nanovaccines
19.
Fish Shellfish Immunol ; 151: 109665, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38830521

ABSTRACT

Bacterial septicemia in freshwater fish is mainly caused by Aeromonas hydrophila infection, which affects the development of aquaculture industry. In the context of sustainable aquaculture, subunit vaccines are of great values because they play positive roles in reducing the overuse of antibiotics and protecting aquatic animals against bacterial infection. In this study, the recombinant outer membrane protein OmpTS of A. hydrophila were used as subunit vaccine to immunize Megalobrama amblycephala, and its immunoprotective effect and host immune responses were evaluated. The survival rates of the vaccinated groups after bacterial infection were significantly higher than that of the control group, especially of the OmpTS high-dose vaccinated group. The better protective effects of vaccinated groups might be attributed to the increased levels of serum IgM-specific antibody titer, the reduced relative abundance of A. hydrophila in various tissues, the increased number of immune-positive cells with different epitopes, the up-regulated expression levels of immune-related genes, and the enhanced activities of antibacterial enzymes. In conclusion, OmpTS subunit vaccine could strongly induce host immune responses in M. amblycephala, thereby enhancing both cellular and humoral immunity, which exhibited excellent and effective immunoprotective efficacy.


Subject(s)
Aeromonas hydrophila , Bacterial Vaccines , Cyprinidae , Fish Diseases , Gram-Negative Bacterial Infections , Vaccines, Subunit , Aeromonas hydrophila/immunology , Animals , Fish Diseases/prevention & control , Fish Diseases/immunology , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/prevention & control , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Cyprinidae/immunology , Bacterial Outer Membrane Proteins/immunology , Immunity, Humoral
20.
Fish Shellfish Immunol ; 151: 109701, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38878911

ABSTRACT

In the field of aquaculture, the enhancement of animal health and disease prevention is progressively being tackled using alternatives to antibiotics, including vaccines and probiotics. This study was designed to evaluate the potential of a recombinant Bacillus methylotrophicus, engineered to express the outer membrane channel protein TolC of Aeromonas hydrophila AH3 and the green fluorescent protein GFP, as an oral vaccine. Initially, the genes encoding tolC and GFP were cloned into a prokaryotic expression system, and anti-TolC mouse antiserum was generated. Subsequently, the tolC gene was subcloned into a modified pMDGFP plasmid, which was transformed into B. methylotrophicus WM-1 for protein expression. The recombinant B. methylotrophicus BmT was then administered to grass carp via co-feeding, and its efficacy as an oral vaccine was assessed. Our findings demonstrated successful expression of the 55 kDa TolC and 28 kDa GFP proteins, and the preparation of polyclonal antibodies with high specificity. The BmT exhibited stable expression of the GFP-TolC fusion protein and excellent genetic stability. Following oral immunization, significant elevations were observed in serum-specific IgM levels and the activities of acid phosphatase (ACP), alkaline phosphatase (AKP), superoxide dismutase (SOD), and lysozyme (LZM) in grass carp. Concurrently, significant upregulation of immune-related genes, including IFN-I, IL-10, IL-1ß, TNF-α, and IgT, was noted in the intestines, head kidney, and spleen of the grass carp. Colonization tests further revealed that the BmT persisted in the gut of immunized fish even after a fasting period of 7 days. Notably, oral administration of BmT enhanced the survival rate of grass carp following A. hydrophila infection. These results suggest that the oral BmT vaccine developed in this study holds promise for future applications in aquaculture.


Subject(s)
Aeromonas hydrophila , Bacterial Vaccines , Carps , Fish Diseases , Gram-Negative Bacterial Infections , Animals , Fish Diseases/immunology , Fish Diseases/prevention & control , Carps/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/immunology , Aeromonas hydrophila/immunology , Administration, Oral , Vaccination/veterinary , Bacillus , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL