Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
BMC Genomics ; 25(1): 791, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160492

ABSTRACT

Acinetobacter baumannii is a gram-negative bacillus prevalent in nature, capable of thriving under various environmental conditions. As an opportunistic pathogen, it frequently causes nosocomial infections such as urinary tract infections, bacteremia, and pneumonia, contributing to increased morbidity and mortality in clinical settings. Consequently, developing novel vaccines against Acinetobacter baumannii is of utmost importance. In our study, we identified 10 highly conserved antigenic proteins from the NCBI and UniProt databases for epitope mapping. We subsequently screened and selected 8 CTL, HTL, and LBL epitopes, integrating them into three distinct vaccines constructed with adjuvants. Following comprehensive evaluations of immunological and physicochemical parameters, we conducted molecular docking and molecular dynamics simulations to assess the efficacy and stability of these vaccines. Our findings indicate that all three multi-epitope mRNA vaccines designed against Acinetobacter baumannii are promising; however, further animal studies are required to confirm their reliability and effectiveness.


Subject(s)
Acinetobacter baumannii , Bacterial Vaccines , Computational Biology , Acinetobacter baumannii/immunology , Acinetobacter baumannii/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Molecular Docking Simulation , Acinetobacter Infections/prevention & control , Acinetobacter Infections/immunology , Epitope Mapping , mRNA Vaccines , Molecular Dynamics Simulation , Humans , RNA, Messenger/genetics , RNA, Messenger/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/chemistry
2.
J Appl Microbiol ; 135(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39020252

ABSTRACT

AIMS: Currently, immunoinformatic approaches have shown promise in rapidly and cost-effectively identifying new antigens from the Leptospira proteome. Chimeric multiepitope proteins offer a strategy with significant potential for implementation in diagnosis and vaccines development. METHODS AND RESULTS: In this study, we detail the immunoinformatic analyses and design of a new recombinant chimeric protein constructed with epitopes identified from the sequences of ErpY-like and LemA proteins, previously identified as potential antigens for controlling leptospirosis. We expressed the chimeric protein using Escherichia coli heterologous systems, evaluated its antigenicity using serum from naturally infected patients, and its immunogenicity in mice as an animal model, with Freund as an adjuvant. The resulting recombinant chimeric protein, named rErpY-LemA, was successfully expressed and purified using a prokaryotic system, with an expected mass of 35 kDa. Serologic assays using serum samples from naturally infected patients demonstrated recognition of the chimera protein by antibodies present in sera. Animals immunized with the chimera exhibited a significant IgG antibody response from the 7th day (P < 0.001), persisting until day 49 of experimentation, with a titer of 1:12,800 (P < 0.05). Notably, significant production of IgA, IgM, and IgG subclasses was observed in animals immunized with the chimera. CONCLUSIONS: These results highlight the promising role of immunoinformatics in rapidly identifying antigens and the potential of chimeric multiepitope proteins in developing effective strategies for leptospirosis control.


Subject(s)
Antigens, Bacterial , Leptospirosis , Recombinant Fusion Proteins , Leptospirosis/immunology , Leptospirosis/prevention & control , Animals , Mice , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Humans , Antibodies, Bacterial/blood , Leptospira/immunology , Leptospira/genetics , Computational Biology , Epitopes/immunology , Epitopes/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Escherichia coli/genetics , Immunoglobulin G/blood , Immunoglobulin G/immunology , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics
3.
Microb Pathog ; 193: 106768, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960217

ABSTRACT

Fowl cholera is an infectious disease that affects both poultry and wild birds, characterized by hemorrhagic and septicemic symptoms, caused by Pasteurella multocida (P. multocida), and leading to substantial economic losses in the poultry sector. The development of genetic engineering vaccines against avian P. multocida encountered early-stage challenges due to the limited availability of effective gene editing tools. Presently, NgAgoDM-enhanced homologous recombination stands as a potent technique for achieving efficient gene knockout in avian P. multocida. Hence, this study employed NgAgoDM-enhanced homologous recombination to target and knockout hyaE (239-359aa), hyaD, hexABC, and hexD, denoted as ΔhyaE (239-359aa), ΔhyaD, ΔhexABC, and ΔhexD, respectively. Additionally, we generated a hyaD recovery strain with two point mutations, designated as mhyaD. Thus, this study systematically examined the impact of capsular synthetic gene clusters on the pathogenicity of P. multocida. Moreover, the study demonstrated the critical role of hyaD activity in the virulence of avian P. multocida. This study offers novel insights for enhancing attenuated vaccines further.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Poultry Diseases , Pasteurella multocida/genetics , Pasteurella multocida/pathogenicity , Animals , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Virulence/genetics , Poultry Diseases/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Homologous Recombination , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Gene Knockout Techniques , Chickens/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Birds/microbiology , Multigene Family , Virulence Factors/genetics , Poultry/microbiology
4.
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
5.
Commun Biol ; 7(1): 779, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38942984

ABSTRACT

The Mycoplasma Immunoglobulin Binding/Protease (MIB-MIP) system is a candidate 'virulence factor present in multiple pathogenic species of the Mollicutes, including the fast-growing species Mycoplasma feriruminatoris. The MIB-MIP system cleaves the heavy chain of host immunoglobulins, hence affecting antigen-antibody interactions and potentially facilitating immune evasion. In this work, using -omics technologies and 5'RACE, we show that the four copies of the M. feriruminatoris MIB-MIP system have different expression levels and are transcribed as operons controlled by four different promoters. Individual MIB-MIP gene pairs of M. feriruminatoris and other Mollicutes were introduced in an engineered M. feriruminatoris strain devoid of MIB-MIP genes and were tested for their functionality using newly developed oriC-based plasmids. The two proteins are functionally expressed at the surface of M. feriruminatoris, which confirms the possibility to display large membrane-associated proteins in this bacterium. However, functional expression of heterologous MIB-MIP systems introduced in this engineered strain from phylogenetically distant porcine Mollicutes like Mesomycoplasma hyorhinis or Mesomycoplasma hyopneumoniae could not be achieved. Finally, since M. feriruminatoris is a candidate for biomedical applications such as drug delivery, we confirmed its safety in vivo in domestic goats, which are the closest livestock relatives to its native host the Alpine ibex.


Subject(s)
Bacterial Vaccines , Mycoplasma , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Mycoplasma/genetics , Mycoplasma/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Immunoglobulins/immunology , Gene Expression Regulation, Bacterial , Mycoplasma Infections/veterinary , Mycoplasma Infections/microbiology , Mycoplasma Infections/immunology , Mycoplasma Infections/prevention & control , Goats
6.
Virulence ; 15(1): 2367783, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38937901

ABSTRACT

Helicobacter pylori causes globally prevalent infections that are highly related to chronic gastritis and even development of gastric carcinomas. With the increase of antibiotic resistance, scientists have begun to search for better vaccine design strategies to eradicate H. pylori colonization. However, while current strategies prefer to formulate vaccines with a single H. pylori antigen, their potential has not yet been fully realized. Outer membrane vesicles (OMVs) are a potential platform since they could deliver multiple antigens. In this study, we engineered three crucial H. pylori antigen proteins (UreB, CagA, and VacA) onto the surface of OMVs derived from Salmonella enterica serovar Typhimurium (S. Typhimurium) mutant strains using the hemoglobin protease (Hbp) autotransporter system. In various knockout strategies, we found that OMVs isolated from the ΔrfbP ΔfliC ΔfljB ΔompA mutants could cause distinct increases in immunoglobulin G (IgG) and A (IgA) levels and effectively trigger T helper 1- and 17-biased cellular immune responses, which perform a vital role in protecting against H. pylori. Next, OMVs derived from ΔrfbP ΔfliC ΔfljB ΔompA mutants were used as a vector to deliver different combinations of H. pylori antigens. The antibody and cytokine levels and challenge experiments in mice model indicated that co-delivering UreB and CagA could protect against H. pylori and antigen-specific T cell responses. In summary, OMVs derived from the S. Typhimurium ΔrfbP ΔfliC ΔfljB ΔompA mutant strain as the vector while importing H. pylori UreB and CagA as antigenic proteins using the Hbp autotransporter system would greatly benefit controlling H. pylori infection.


Outer membrane vesicles (OMVs), as a novel antigen delivery platform, has been used in vaccine design for various pathogens and even tumors. Salmonella enterica serovar Typhimurium (S. Typhimurium), as a bacterium that is easy to engineer and has both adjuvant efficacy and immune stimulation capacity, has become the preferred bacterial vector for purifying OMVs after Escherichia coli. This study focuses on the design of Helicobacter pylori ;(H. pylori) vaccines, utilizing genetically modified Salmonella OMVs to present several major antigens of H. pylori, including UreB, VacA and CagA. The optimal Salmonella OMV delivery vector and antigen combinations are screened and identified, providing new ideas for the development of H. pylori vaccines and an integrated antigen delivery platform for other difficult to develop vaccines for bacteria, viruses, and even tumors.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Helicobacter Infections , Helicobacter pylori , Salmonella typhimurium , Animals , Helicobacter Infections/prevention & control , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Helicobacter pylori/immunology , Helicobacter pylori/genetics , Mice , Salmonella typhimurium/immunology , Salmonella typhimurium/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Female , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Immunoglobulin G , Genetic Engineering , Urease/immunology , Urease/genetics , Disease Models, Animal
7.
Helicobacter ; 29(3): e13104, 2024.
Article in English | MEDLINE | ID: mdl-38923222

ABSTRACT

Helicobacter pylori (H. pylori) strain is the most genetically diverse pathogenic bacterium and now alarming serious human health concern ranging from chronic gastritis to gastric cancer and human death all over the world. Currently, the majority of commercially available diagnostic assays for H. pylori is a challenging task due to the heterogeneity of virulence factors in various geographical regions. In this concern, designing of universal multi-epitope immunogenic biomarker targeted for all H. pylori strains would be crucial to successfully immunodiagnosis assay and vaccine development for H. pylori infection. Hence, the present study aimed to explore the potential immunogenic epitopes of PSA D15 and Cag11 proteins of H. pylori, using immunoinformatics web tools in order to design novel immune-reactive multi-epitope antigens for enhanced immunodiagnosis in humans. Through an in silico immunoinformatics approach, high-ranked B-cell, MHC-I, and MHC-II epitopes of PSA D15 and Cag11 proteins were predicted, screened, and selected. Subsequently, a novel multi-epitope PSA D15 and Cag11 antigens were designed by fused the high-ranked B-cell, MHC-I, and MHC-II epitopes and 50S ribosomal protein L7/L12 adjuvant using linkers. The antigenicity, solubility, physicochemical properties, secondary and tertiary structures, 3D model refinement, and validations were carried. Furthermore, the designed multi-epitope antigens were subjected to codon adaptation and in silico cloning, immune response simulation, and molecular docking with receptor molecules. A novel, stable multi-epitope PSA D15 and Cag11 H. pylori antigens were developed and immune simulation of the designed antigens showed desirable levels of immunological response. Molecular docking of designed antigens with immune receptors (B-cell, MHC-I, MHC-II, and TLR-2/4) revealed robust interactions and stable binding affinity to the receptors. The codon optimized and in silico cloned showed that the designed antigens were successfully expressed (CAI value of 0.95 for PSA D15 and 1.0 for Cag11) after inserted into pET-32ba (+) plasmid of the E. coli K12 strain. In conclusion, this study revealed that the designed multi-epitope antigens have a huge immunological potential candidate biomarker and useful in developing immunodiagnostic assays and vaccines for H. pylori infection.


Subject(s)
Antigens, Bacterial , Computational Biology , Helicobacter pylori , Helicobacter pylori/immunology , Helicobacter pylori/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/chemistry , Humans , Helicobacter Infections/diagnosis , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Epitopes/immunology , Immunologic Tests/methods , Molecular Docking Simulation , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Immunoinformatics
8.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38889932

ABSTRACT

Chlamydia trachomatis (Ct) is the most common sexually transmitted bacterial infection worldwide, potentially leading to severe pathologies including pelvic inflammatory disease, ectopic pregnancy, and tubal infertility if left untreated. Current strategies, including screening and antibiotics, have limited effectiveness due to high rates of asymptomatic cases and logistical challenges. A multiepitope prophylactic vaccine could afford long-term protection against infection. Immunoinformatic analyses were employed to design a multiepitope Chlamydia vaccine antigen. B- and T-cell epitopes from five highly conserved and immunogenic Ct antigens were predicted and selected for the vaccine design. The final construct, adjuvanted with cholera toxin A1 subunit (CTA1), was further screened for immunogenicity. CTA1-MECA (multiepitope Chlamydia trachomatis antigen) was identified as antigenic and nonallergenic. A tertiary structure was predicted, refined, and validated as a good quality model. Molecular docking exhibited strong interactions between the vaccine and toll-like receptor 4 (TLR4). Additionally, immune responses consistent with protection including IFN-γ, IgG + IgM antibodies, and T- and B-cell responses were predicted following vaccination in an immune simulation. Expression of the construct in an Escherichia coli expression vector proved efficient. To further validate the vaccine efficacy, we assessed its immunogenicity in mice. Immunization with CTA1-MECA elicited high levels of Chlamydia-specific antibodies in mucosal and systemic compartments.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Chlamydia Infections , Chlamydia trachomatis , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Molecular Docking Simulation , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Chlamydia Infections/prevention & control , Chlamydia Infections/immunology , Animals , Chlamydia trachomatis/immunology , Epitopes, T-Lymphocyte/immunology , Mice , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Female , Antigens, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Computer Simulation , Epitopes/immunology , Humans , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Cholera Toxin/immunology , Cholera Toxin/genetics , Disease Models, Animal
9.
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
10.
Microb Pathog ; 192: 106685, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750774

ABSTRACT

QseC is a membrane sensor kinase that enables bacteria to perceive autoinducers -3, adrenaline, and norepinephrine to initiate downstream gene transcription. In this study, we found that the QseC protein of Glaesserella parasuis can serve as an effective antigen to activate the host's immune response. Therefore, we investigated the immunogenicity and host protective effect of this protein. ELISA and indirect immunofluorescence results showed that QseC protein can induce high titer levels of humoral immunity in mice and regularly generate specific serum antibodies. We used MTS reagents to detect lymphocyte proliferation levels and found that QseC protein can cause splenic lymphocyte proliferation with memory and specificity. Further immunological analysis of the spleen cell supernatant revealed significant upregulation of levels of IL-1ß, IL-4 and IFN-γ in the QseC + adjuvant group. In the mouse challenge experiment, it was found that QseC + adjuvant can provide effective protection. The results of this study demonstrate that QseC protein provides effective protection in a mouse model and has the potential to serve as a candidate antigen for a novel subunit vaccine for further research.


Subject(s)
Antibodies, Bacterial , Haemophilus Infections , Interferon-gamma , Interleukin-4 , Animals , Mice , Interleukin-4/metabolism , Interleukin-4/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Haemophilus Infections/immunology , Haemophilus Infections/prevention & control , Haemophilus Infections/microbiology , Interferon-gamma/metabolism , Histidine Kinase/genetics , Histidine Kinase/metabolism , Histidine Kinase/immunology , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Immunity, Humoral , Mice, Inbred BALB C , Spleen/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Cell Proliferation , Female , Adjuvants, Immunologic , Haemophilus parasuis/immunology , Haemophilus parasuis/genetics , Cytokines/metabolism , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Disease Models, Animal , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Lymphocytes/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics
11.
Vaccine ; 42(18): 3883-3898, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38777697

ABSTRACT

BACKGROUND: Community-acquired pneumonia often stems from the macrolide-resistant strain of Mycoplasma pneumoniae, yet no effective vaccine exists against it. METHODS: This study proposes a vaccine-immunoinformatics strategy for Mycoplasma pneumoniae and other pathogenic microbes. Specifically, dominant B and T cell epitopes of the Mycoplasma pneumoniae P30 adhesion protein were identified through immunoinformatics method. The vaccine sequence was then constructed by coupling with CTLA-4 extracellular region, a novel molecular adjuvant for antigen-presenting cells. Subsequently, the vaccine's physicochemical properties, antigenicity, and allergenicity were verified. Molecular dynamics modeling was employed to confirm interaction with TLR-2, TLR-4, B7-1, and B7-2. Finally, the vaccine underwent in silico cloning for expression. RESULTS: The vaccine exhibited both antigenicity and non-allergenicity. Molecular dynamics simulation, post-docking with TLR-2, TLR-4, B7-1, and B7-2, demonstrated stable interaction between the vaccine and these molecules. In silico cloning confirmed effective expression of the vaccine gene in insect baculovirus vectors. CONCLUSION: This vaccine-immunoinformatics approach holds promise for the development of vaccines against Mycoplasma pneumoniae and other pathogenic non-viral and non-bacterial microbes.


Subject(s)
Bacterial Vaccines , CTLA-4 Antigen , Computational Biology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Mycoplasma pneumoniae , Pneumonia, Mycoplasma , Mycoplasma pneumoniae/immunology , Mycoplasma pneumoniae/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Humans , Computational Biology/methods , Pneumonia, Mycoplasma/prevention & control , Pneumonia, Mycoplasma/immunology , CTLA-4 Antigen/immunology , Molecular Dynamics Simulation , Molecular Docking Simulation , Toll-Like Receptor 2/immunology , Immunoinformatics
12.
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
13.
Virulence ; 15(1): 2345019, 2024 12.
Article in English | MEDLINE | ID: mdl-38656137

ABSTRACT

Klebsiella pneumoniae is an important gram-negative bacterium that causes severe respiratory and healthcare-associated infections. Although antibiotic therapy is applied to treat severe infections caused by K. pneumoniae, drug-resistant isolates pose a huge challenge to clinical practices owing to adverse reactions and the mismanagement of antibiotics. Several studies have attempted to develop vaccines against K. pneumoniae, but there are no licensed vaccines available for the control of K. pneumoniae infection. In the current study, we constructed a novel DNA vaccine, pVAX1-YidR, which encodes a highly conserved virulence factor YidR and a recombinant expression plasmid pVAX1-IL-17 encoding Interleukin-17 (IL-17) as a molecular adjuvant. Adaptive immune responses were assessed in immunized mice to compare the immunogenicity of the different vaccine schemes. The results showed that the targeted antigen gene was expressed in HEK293T cells using an immunofluorescence assay. Mice immunized with pVAX1-YidR elicited a high level of antibodies, induced strong cellular immune responses, and protected mice from K. pneumoniae challenge. Notably, co-immunization with pVAX1-YidR and pVAX1-IL-17 significantly augmented host adaptive immune responses and provided better protection against K. pneumoniae infections in vaccinated mice. Our study demonstrates that combined DNA vaccines and molecular adjuvants is a promising strategy to develop efficacious antibacterial vaccines against K. pneumoniae infections.


Subject(s)
Bacterial Vaccines , Interleukin-17 , Klebsiella Infections , Klebsiella pneumoniae , Vaccines, DNA , Animals , Female , Humans , Mice , Adaptive Immunity , Adjuvants, Immunologic/administration & dosage , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/administration & dosage , Disease Models, Animal , HEK293 Cells , Immunity, Cellular , Immunization , Interleukin-17/immunology , Interleukin-17/genetics , Klebsiella Infections/prevention & control , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Klebsiella pneumoniae/genetics , Mice, Inbred BALB C , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Vaccines, DNA/administration & dosage , Virulence Factors/immunology , Virulence Factors/genetics
14.
Microb Biotechnol ; 17(3): e14446, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38536702

ABSTRACT

Developing protein-based vaccines against bacteria has proved much more challenging than producing similar immunisations against viruses. Currently, anti-bacterial vaccines are designed using methods based on reverse vaccinology. These identify broadly conserved, immunogenic proteins using a combination of genomic and high-throughput laboratory data. While this approach has successfully generated multiple rationally designed formulations that show promising immunogenicity in animal models, few have been licensed. The difficulty of inducing protective immunity in humans with such vaccines mirrors the ability of many bacteria to recolonise individuals despite recognition by natural polyvalent antibody repertoires. As bacteria express too many antigens to evade all adaptive immune responses through mutation, they must instead inhibit the efficacy of such host defences through expressing surface structures that interface with the immune system. Therefore, 'immune interface interference' (I3) vaccines that target these features should synergistically directly target bacteria and prevent them from inhibiting responses to other surface antigens. This approach may help us understand the efficacy of the two recently introduced immunisations against serotype B meningococci, which both target the Factor H-binding protein (fHbp) that inhibits complement deposition on the bacterial surface. Therefore, I3 vaccine designs may help overcome the current challenges of developing protein-based vaccines to prevent bacterial infections.


Subject(s)
Meningococcal Vaccines , Neisseria meningitidis , Animals , Humans , Bacterial Vaccines/genetics , Bacterial Proteins/genetics , Antigens, Bacterial/genetics , Antibodies, Bacterial , Neisseria meningitidis/genetics
15.
Microb Pathog ; 189: 106596, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38395317

ABSTRACT

Botulism is a severe disease caused by potent botulinum neurotoxins (BoNTs) produced by Clostridium botulinum. This disease is associated with high-lethality outbreaks in cattle, which have been linked to the ingestion of preformed BoNT serotypes C and D, emphasizing the need for effective vaccines. The potency of current commercial toxoids (formaldehyde-inactivated BoNTs) is assured through tests in guinea pigs according to government regulatory guidelines, but their short-term immunity raises concerns. Recombinant vaccines containing the receptor-binding domain have demonstrated potential for eliciting robust protective immunity. Previous studies have demonstrated the safety and effectiveness of recombinant E. coli bacterin, eliciting high titers of neutralizing antibodies against C. botulinum and C. perfringens in target animal species. In this study, neutralizing antibody titers in cattle and the long-term immune response against BoNT/C and D were used to assess the efficacy of the oil-based adjuvant compared with that of the aluminum hydroxide adjuvant in cattle. The vaccine formulation containing Montanide™ ISA 50 yielded significantly higher titers of neutralizing antibody against BoNT/C and D (8.64 IU/mL and 9.6 IU/mL, respectively) and induced an immune response that lasted longer than the response induced by aluminum, extending between 30 and 60 days. This approach represents a straightforward, cost-effective strategy for recombinant E. coli bacterin, enhancing both the magnitude and duration of the immune response to botulism.


Subject(s)
Botulinum Toxins , Botulism , Clostridium botulinum , Cattle , Animals , Guinea Pigs , Botulism/prevention & control , Botulism/veterinary , Aluminum Hydroxide , Escherichia coli/genetics , Bacterial Vaccines/genetics , Botulinum Toxins/genetics , Clostridium botulinum/genetics , Adjuvants, Immunologic , Antibodies, Neutralizing , Immunity , Antibodies, Bacterial
16.
Vaccine ; 42(8): 1868-1872, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38365481

ABSTRACT

Vaccination is the most cost-effective tool to control contagious bovine pleuropneumonia. The vaccines currently used in Africa are derived from a live strain called T1, which was attenuated by passage in embryonated eggs and broth culture. The number of passages is directly correlated to the degree of attenuation of the vaccinal strains and inversely correlated to their immunogenicity in cattle. Current quality control protocols applied to vaccine batches allow the assessment of identity, purity, and titers, but cannot assess the level of genetic drift form the parental vaccine strains. Deep sequencing was used to assess the genetic drift generated over controlled in vitro passages of the parental strain, as well as on commercial vaccine batches. Signatures of cloning procedures were detected in some batches, which imply a deviation from the standard production protocol. Deep sequencing is proposed as a new tool for the identity and stability control of T1 vaccines.


Subject(s)
Cattle Diseases , Mycoplasma mycoides , Pleuropneumonia, Contagious , Pleuropneumonia , Animals , Cattle , Bacterial Vaccines/genetics , Africa , Vaccines, Attenuated/genetics , Quality Control , High-Throughput Nucleotide Sequencing , Pleuropneumonia, Contagious/prevention & control , Mycoplasma mycoides/genetics
17.
Vet Microbiol ; 291: 110008, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38364465

ABSTRACT

Mycoplasma gallisepticum infection in poultry leads to disease and pathology that can reduce producer profits. Live attenuated vaccines are available that can limit or completely prevent the effects of infection. Field isolates that are genetically related to the attenuated vaccine strains have been isolated, raising the question of whether the attenuation of the vaccine strains is limited and can lead the strains to revert to more virulent forms. The 6/85 live attenuated vaccine is derived from a field isolate collected in the United States. Analysis of the genome of sequenced M. gallisepticum strains revealed a cluster of 10 6/85-like strains that group with the 6/85 vaccine strain. Four genomic regions were identified that allowed for strain differentiation. The genetic differences between strains points toward nine of the ten strains most likely being sister strains to the 6/85 vaccine strain. Insufficient differences are present in the tenth strain to make a definitive conclusion. These results suggest that most if not all strains similar to the live attenuated vaccine strain are field isolates of the parent strain used to derive the live attenuated vaccine.


Subject(s)
Mycoplasma Infections , Mycoplasma gallisepticum , Poultry Diseases , Animals , Vaccines, Attenuated , Bacterial Vaccines/genetics , Chickens , Poultry Diseases/prevention & control , Mycoplasma Infections/prevention & control , Mycoplasma Infections/veterinary
18.
Braz J Microbiol ; 55(1): 943-953, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38217795

ABSTRACT

Mycoplasma hyopneumoniae (M. hyopneumoniae) is a primary agent of porcine enzootic pneumonia, a disease that causes significant economic losses to pig farming worldwide. Commercial vaccines induce partial protection, evidencing the need for a new vaccine against M. hyopneumoniae. In our work, three chimeric proteins were constructed, composed of potentially immunogenic domains from M. hyopneumoniae proteins. We designed three chimeric proteins (Q1, Q2, and Q3) based on bioinformatics analysis that identified five potential proteins with immunogenic potential (MHP418, MHP372, MHP199, P97, and MHP0461). The chimeric proteins were inoculated in the murine model to evaluate the immune response. The mice vaccinated with the chimeras presented IgG and IgG1 against proteins of M. hyopneumoniae. There was induction of IgG in mice immunized with Q3 starting from 30 days post-vaccination, and groups Q1 and Q2 showed induction at 45 days. Mice of the group immunized with Q3 showed the production of IgA. In addition, the mice inoculated with chimeric proteins showed a proinflammatory cytokine response; Q1 demonstrated higher levels of TNF, IL-6, IL2, and IL-17. In contrast, animals immunized with Q2 showed an increase in the concentrations of TNF, IL-6, and IL-4, whereas those immunized with Q3 exhibited an increase in the concentrations of TNF, IL-6, IL-10, and IL-4. The results of the present study indicate that these three chimeric proteins can be used in future vaccine trials with swine because of the promising antigenicity.


Subject(s)
Mycoplasma hyopneumoniae , Animals , Swine , Mice , Mycoplasma hyopneumoniae/genetics , Interleukin-4 , Interleukin-6 , Bacterial Vaccines/genetics , Immunoglobulin G , Recombinant Fusion Proteins/genetics
19.
J Biotechnol ; 380: 51-63, 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38151110

ABSTRACT

Vibriosis is caused by Vibrio anguillarum in various species of aquaculture. A novel, secure, and stable vaccine is needed to eradicate vibriosis. Here, for reverse vaccinology and plant-based expression, the outer membrane protein K (OmpK) of V. anguillarum was chosen due to its conserved nature in all Vibrio species. OmpK, an ideal vaccine candidate against vibriosis, demonstrated immunogenic, non-allergic, and non-toxic behavior by using various bioinformatics tools. Docking showed the interaction of the OmpK model with TLR-5. In comparison to costly platforms, plants can be used as alternative and economic bio-factories to produce vaccine antigens. We expressed OmpK antigen in Nicotiana tabacum using Agrobacterium-mediated transformation. The expression vector was constructed using Gateway® cloning. Transgene integration was verified by polymerase chain reaction (PCR), and the copy number via qRT-PCR, which showed two copies of transgenes. Western blotting detected monomeric form of OmpK protein. The total soluble protein (TSP) fraction of OmpK was equivalent to 0.38% as detected by ELISA. Mice and fish were immunized with plant-derived OmpK antigen, which showed a significantly high level of anti-OmpK antibodies. The present study is the first report of OmpK antigen expression in higher plants for the potential use as vaccine in aquaculture against vibriosis, which could provide protection against multiple Vibrio species due to the conserved nature OmpK antigen.


Subject(s)
Fish Diseases , Vibrio Infections , Vibrio , Animals , Mice , Nicotiana/genetics , Bacterial Vaccines/genetics , Vibrio/genetics , Vibrio Infections/prevention & control , Vibrio Infections/veterinary , Fish Diseases/prevention & control
20.
Braz. j. microbiol ; 49(1): 18-19, Jan.-Mar. 2018.
Article in English | LILACS | ID: biblio-889201

ABSTRACT

ABSTRACT Bacillus anthracis strain SPV842_15 was isolated from bovine fetus, while B. anthracis strain Brazilian vaccinal was recovered from a commercial vaccine. We report here the genome sequences of both strains. The SPV842_15 genome is composed of a single circular chromosome with a length of 5,228,664 base pairs, and comprises 5911 coding sequences. In turn, the Brazilian vaccinal genome remains in 201 contigs with 5733 coding sequences. Both genomes have an overall C + G content of 35.4%, and 11 genes encoding the ribosomal RNAs (rRNAs) 5S, 16S and 23S. Only the plasmid pX01 sequence, which carries genes for toxins synthesis, was detected and completely assembled for both strains. These plasmids have a length of 181,684 base pairs and a C + G content of 32.5%. These genomic data generate insights about vaccinal B. anthracis virulence.


Subject(s)
Animals , Cattle , Bacillus anthracis/isolation & purification , Bacillus anthracis/genetics , Bacterial Vaccines/genetics , Cattle Diseases/microbiology , Genome, Bacterial , Phylogeny , Plasmids/genetics , Bacillus anthracis/classification , Base Composition , DNA, Bacterial/genetics , Molecular Sequence Data , Bacterial Vaccines/isolation & purification , Base Sequence
SELECTION OF CITATIONS
SEARCH DETAIL