Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.801
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 28: 107-30, 2010.
Article in English | MEDLINE | ID: mdl-19968562

ABSTRACT

The capsular polysaccharides (CPSs) of most pathogenic bacteria are T cell-independent antigens whose conjugation to carrier proteins evokes a carbohydrate-specific response eliciting T cell help. However, certain bacterial CPSs, known as zwitterionic polysaccharides (ZPSs), activate the adaptive immune system through processing by antigen-presenting cells and presentation by the major histocompatibility complex class II pathway to CD4(+) T cells. This discovery was the first mechanistic insight into how carbohydrates-a class of biological molecules previously thought to be T cell independent-can in fact activate T cells. Through their ability to activate CD4(+) T cells, ZPSs direct the cellular and physical maturation of the developing immune system. In this review, we explore the still-enigmatic relations between CPSs and the adaptive immune machinery at the cellular and molecular levels, and we discuss how new insights into the biological impact of ZPSs expand our concepts of the role of carbohydrates in microbial interactions with the adaptive immune system.


Subject(s)
Adaptive Immunity , Antigens, Bacterial/immunology , Bacteria/immunology , Polysaccharides, Bacterial/immunology , Animals , Antigens, Bacterial/chemistry , Bacteria/chemistry , Bacterial Vaccines/immunology , Humans , Polysaccharides, Bacterial/chemistry , T-Lymphocytes/immunology
2.
Nature ; 609(7927): 582-589, 2022 09.
Article in English | MEDLINE | ID: mdl-36071157

ABSTRACT

Increased levels of proteases, such as trypsin, in the distal intestine have been implicated in intestinal pathological conditions1-3. However, the players and mechanisms that underlie protease regulation in the intestinal lumen have remained unclear. Here we show that Paraprevotella strains isolated from the faecal microbiome of healthy human donors are potent trypsin-degrading commensals. Mechanistically, Paraprevotella recruit trypsin to the bacterial surface through type IX secretion system-dependent polysaccharide-anchoring proteins to promote trypsin autolysis. Paraprevotella colonization protects IgA from trypsin degradation and enhances the effectiveness of oral vaccines against Citrobacter rodentium. Moreover, Paraprevotella colonization inhibits lethal infection with murine hepatitis virus-2, a mouse coronavirus that is dependent on trypsin and trypsin-like proteases for entry into host cells4,5. Consistently, carriage of putative genes involved in trypsin degradation in the gut microbiome was associated with reduced severity of diarrhoea in patients with SARS-CoV-2 infection. Thus, trypsin-degrading commensal colonization may contribute to the maintenance of intestinal homeostasis and protection from pathogen infection.


Subject(s)
Gastrointestinal Microbiome , Intestine, Large , Symbiosis , Trypsin , Administration, Oral , Animals , Bacterial Secretion Systems , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Bacteroidetes/isolation & purification , Bacteroidetes/metabolism , COVID-19/complications , Citrobacter rodentium/immunology , Diarrhea/complications , Feces/microbiology , Gastrointestinal Microbiome/genetics , Humans , Immunoglobulin A/metabolism , Intestine, Large/metabolism , Intestine, Large/microbiology , Mice , Murine hepatitis virus/metabolism , Murine hepatitis virus/pathogenicity , Proteolysis , SARS-CoV-2/pathogenicity , Trypsin/metabolism , Virus Internalization
3.
Immunity ; 48(3): 584-598.e5, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29548673

ABSTRACT

Live vaccines historically afford superior protection, yet the cellular and molecular mechanisms mediating protective immunity remain unclear. Here we found that vaccination of mice with live, but not dead, Gram-negative bacteria heightened follicular T helper cell (Tfh) differentiation, germinal center formation, and protective antibody production through the signaling adaptor TRIF. Complementing the dead vaccine with an innate signature of bacterial viability, bacterial RNA, recapitulated these responses. The interferon (IFN) and inflammasome pathways downstream of TRIF orchestrated Tfh responses extrinsically to B cells and classical dendritic cells. Instead, CX3CR1+CCR2- monocytes instructed Tfh differentiation through interleukin-1ß (IL-1ß), a tightly regulated cytokine secreted upon TRIF-dependent IFN licensing of the inflammasome. Hierarchical production of IFN-ß and IL-1ß dictated Tfh differentiation and elicited the augmented humoral responses characteristic of live vaccines. These findings identify bacterial RNA, an innate signature of microbial viability, as a trigger for Tfh differentiation and suggest new approaches toward vaccine formulations for coordinating augmented Tfh and B cell responses.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/immunology , Lymphocyte Activation/immunology , Microbial Viability/immunology , RNA, Bacterial/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antibodies, Neutralizing/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B-Lymphocytes/metabolism , Bacterial Vaccines/immunology , Biomarkers , Cell Differentiation/immunology , Cytokines/metabolism , Germinal Center , Host-Pathogen Interactions/immunology , Immunity, Cellular , Immunity, Innate , Inflammasomes/metabolism , Mice , Monocytes/immunology , Monocytes/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/metabolism
4.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39133098

ABSTRACT

Pseudomonas aeruginosa is a complex nosocomial infectious agent responsible for numerous illnesses, with its growing resistance variations complicating treatment development. Studies have emphasized the importance of virulence factors OprE and OprF in pathogenesis, highlighting their potential as vaccine candidates. In this study, B-cell, MHC-I, and MHC-II epitopes were identified, and molecular linkers were active to join these epitopes with an appropriate adjuvant to construct a vaccine. Computational tools were employed to forecast the tertiary framework, characteristics, and also to confirm the vaccine's composition. The potency was weighed through population coverage analysis and immune simulation. This project aims to create a multi-epitope vaccine to reduce P. aeruginosa-related illness and mortality using immunoinformatics resources. The ultimate complex has been determined to be stable, soluble, antigenic, and non-allergenic upon inspection of its physicochemical and immunological properties. Additionally, the protein exhibited acidic and hydrophilic characteristics. The Ramachandran plot, ProSA-web, ERRAT, and Verify3D were employed to ensure the final model's authenticity once the protein's three-dimensional structure had been established and refined. The vaccine model showed a significant binding score and stability when interacting with MHC receptors. Population coverage analysis indicated a global coverage rate of 83.40%, with the USA having the highest coverage rate, exceeding 90%. Moreover, the vaccine sequence underwent codon optimization before being cloned into the Escherichia coli plasmid vector pET-28a (+) at the EcoRI and EcoRV restriction sites. Our research has developed a vaccine against P. aeruginosa that has strong binding affinity and worldwide coverage, offering an acceptable way to mitigate nosocomial infections.


Subject(s)
Computational Biology , Pseudomonas Infections , Pseudomonas aeruginosa , Sepsis , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Humans , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Sepsis/prevention & control , Sepsis/immunology , Sepsis/microbiology , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Pneumonia/prevention & control , Pneumonia/immunology , Pneumonia/microbiology , Pseudomonas Vaccines/immunology , Bacterial Vaccines/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics
5.
Clin Microbiol Rev ; 37(3): e0016022, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39120140

ABSTRACT

SUMMARYHealthcare-associated infections (HAIs) represent a burden for public health with a high prevalence and high death rates associated with them. Pathogens with a high potential for antimicrobial resistance, such as ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) and Clostridioides difficile, are responsible for most HAIs. Despite the implementation of infection prevention and control intervention, globally, HAIs prevalence is stable and they are mainly due to endogenous pathogens. It is undeniable that complementary to infection prevention and control measures, prophylactic approaches by active or passive immunization are needed. Specific groups at-risk (elderly people, chronic condition as immunocompromised) and also healthcare workers are key targets. Medical procedures and specific interventions are known to be at risk of HAIs, in addition to hospital environmental exposure. Vaccines or monoclonal antibodies can be seen as attractive preventive approaches for HAIs. In this review, we present an overview of the vaccines and monoclonal antibodies in clinical development for prevention of the major bacterial HAIs pathogens. Based on the current state of knowledge, we look at the challenges and future perspectives to improve prevention by these means.


Subject(s)
Antibodies, Monoclonal , Bacterial Infections , Bacterial Vaccines , Cross Infection , Humans , Cross Infection/prevention & control , Bacterial Infections/prevention & control , Bacterial Infections/immunology , Bacterial Infections/epidemiology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/immunology , Bacterial Vaccines/immunology
6.
J Virol ; 98(3): e0192323, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38358289

ABSTRACT

Helicobacter pylori is a human pathogen that infects almost half of the population. Antibiotic resistance in H. pylori threatens health and increases the demand for prophylactic and therapeutic vaccines. Traditional oral vaccine research faces considerable challenges because of the epithelial barrier, potential enterotoxicity of adjuvants, and the challenging conditions of the gastric environment. We developed an intranasal influenza A virus (IAV) vector vaccine based on two live attenuated influenza viruses with modified acidic polymerase protein (PA) genes encoding the A subunit of H. pylori neutrophil-activating protein (NapA), named IAV-NapA, including influenza virus A/WSN/33 (WSN)-NapA and A/Puerto Rico/8/34 (PR8)-NapA. These recombinant influenza viruses were highly attenuated and exhibited strong immunogenicity in mice. Vaccination with IAV-NapA induced antigen-specific humoral and mucosal immune responses while stimulating robust Th1 and Th17 cell immune responses in mice. Our findings suggest that prophylactic and therapeutic vaccination with influenza virus vector vaccines significantly reduces colonization of H. pylori and inflammation in the stomach of mice.IMPORTANCEHelicobacter pylori is the most common cause of chronic gastritis and leads to severe gastroduodenal pathology in some patients. Many studies have shown that Th1 and Th17 cellular and gastric mucosal immune responses are critical in reducing H. pylori load. IAV vector vaccines can stimulate these immune responses while overcoming potential adjuvant toxicity and antigen dosing issues. To date, no studies have demonstrated the role of live attenuated IAV vector vaccines in preventing and treating H. pylori infection. Our work indicates that vaccination with IAV-NapA induces antigen-specific humoral, cellular, and mucosal immunity, producing a protective and therapeutic effect against H. pylori infection in BALB/c mice. This undescribed H. pylori vaccination approach may provide valuable information for developing vaccines against H. pylori infection.


Subject(s)
Helicobacter pylori , Influenza Vaccines , Animals , Humans , Mice , Adjuvants, Immunologic , Bacterial Vaccines/immunology , Helicobacter pylori/physiology , Influenza A virus/physiology , Influenza Vaccines/administration & dosage , Mice, Inbred BALB C , Helicobacter Infections/prevention & control , Administration, Intranasal
7.
Proc Natl Acad Sci U S A ; 119(25): e2202059119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35714289

ABSTRACT

The bacterial genus Bartonella comprises numerous emerging pathogens that cause a broad spectrum of disease manifestations in humans. The targets and mechanisms of the anti-Bartonella immune defense are ill-defined and bacterial immune evasion strategies remain elusive. We found that experimentally infected mice resolved Bartonella infection by mounting antibody responses that neutralized the bacteria, preventing their attachment to erythrocytes and suppressing bacteremia independent of complement or Fc receptors. Bartonella-neutralizing antibody responses were rapidly induced and depended on CD40 signaling but not on affinity maturation. We cloned neutralizing monoclonal antibodies (mAbs) and by mass spectrometry identified the bacterial autotransporter CFA (CAMP-like factor autotransporter) as a neutralizing antibody target. Vaccination against CFA suppressed Bartonella bacteremia, validating CFA as a protective antigen. We mapped Bartonella-neutralizing mAb binding to a domain in CFA that we found is hypervariable in both human and mouse pathogenic strains, indicating mutational antibody evasion at the Bartonella subspecies level. These insights into Bartonella immunity and immune evasion provide a conceptual framework for vaccine development, identifying important challenges in this endeavor.


Subject(s)
Antibodies, Neutralizing , Antigens, Bacterial , Bacteremia , Bartonella Infections , Bartonella , Type V Secretion Systems , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacteremia/immunology , Bacteremia/microbiology , Bacteremia/prevention & control , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , Bartonella/genetics , Bartonella/immunology , Bartonella Infections/immunology , Bartonella Infections/microbiology , Bartonella Infections/prevention & control , Cloning, Molecular , Immune Evasion , Mice , Type V Secretion Systems/immunology , Vaccination
8.
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
9.
J Infect Dis ; 230(3): 569-577, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-38394609

ABSTRACT

BACKGROUND: Chlamydia trachomatis testing and treatment strategies have not decreased infection rates, justifying need for a chlamydia vaccine. A murine study showed that a vaccine consisting of major outer membrane protein (MOMP) and polymorphic membrane proteins (Pmps) E, F, G, and H elicited protective immunity; studies on human cellular immune responses to Pmps are sparse. METHODS: Interferon gamma (IFN-γ) responses to these 5 proteins were measured by ELISPOT in peripheral blood mononuclear cells from women returning for treatment of a positive chlamydia test. Responses were compared in those with spontaneous chlamydia clearance versus persisting infection at baseline and no reinfection versus reinfection at a 3-month follow-up visit. RESULTS: IFN-γ response to 1 or more proteins was detected in 39% at baseline and 51.5% at follow-up, most often to PmpE and MOMP. IFN-γ responses to MOMP were detected less often at follow-up versus baseline in women with reinfection, but were maintained in those without reinfection. Women with spontaneous clearance had a higher magnitude of IFN-γ response to PmpE and MOMP. CONCLUSIONS: IFN-γ responses to these 5 C. trachomatis vaccine candidate proteins were heterogenous and primarily directed against MOMP and PmpE. Spontaneous chlamydia clearance and absence of reinfection may be clinical correlates of protection.


Subject(s)
Bacterial Vaccines , Chlamydia Infections , Chlamydia trachomatis , Interferon-gamma , Leukocytes, Mononuclear , Humans , Female , Chlamydia trachomatis/immunology , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Interferon-gamma/metabolism , Bacterial Vaccines/immunology , Young Adult , Adult , Leukocytes, Mononuclear/immunology , Adolescent , Bacterial Proteins/immunology , Bacterial Outer Membrane Proteins/immunology , Reinfection/immunology , Reinfection/prevention & control , Enzyme-Linked Immunospot Assay
10.
J Infect Dis ; 230(3): 578-589, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-38401891

ABSTRACT

Klebsiella pneumoniae is the leading cause of neonatal sepsis and is increasingly difficult to treat owing to antibiotic resistance. Vaccination represents a tractable approach to combat this resistant bacterium; however, there is currently not a licensed vaccine. Surface polysaccharides, including O-antigens of lipopolysaccharide, have long been attractive candidates for vaccine inclusion. Herein we describe the generation of a bioconjugate vaccine targeting 7 predominant O-antigen subtypes in K. pneumoniae. Each bioconjugate was immunogenic in isolation, with limited cross-reactivity among subtypes. Vaccine-induced antibodies demonstrated varying degrees of binding to a wide variety of K. pneumoniae strains. Furthermore, serum from vaccinated mice induced complement-mediated killing of many of these strains. Finally, increased capsule interfered with the ability of O-antigen antibodies to bind and mediate killing of some K. pneumoniae strains. Taken together, these data indicate that this novel heptavalent O-antigen bioconjugate vaccine formulation exhibits limited efficacy against some, but not all, K. pneumoniae isolates.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Klebsiella Infections , Klebsiella pneumoniae , O Antigens , Klebsiella pneumoniae/immunology , O Antigens/immunology , O Antigens/chemistry , Animals , Antibodies, Bacterial/immunology , Klebsiella Infections/immunology , Klebsiella Infections/prevention & control , Klebsiella Infections/microbiology , Bacterial Vaccines/immunology , Mice , Female , Vaccines, Conjugate/immunology , Mice, Inbred BALB C , Humans
11.
J Proteome Res ; 23(7): 2576-2586, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38860290

ABSTRACT

The relationship between antibiotic resistance and bacterial virulence has not yet been fully explored. Here, we use Edwardsiella tarda as the research model to investigate the proteomic change upon oxytetracycline resistance (LTB4-ROTC). Compared to oxytetracycline-sensitive E. tarda (LTB4-S), LTB4-ROTC has 234 differentially expressed proteins, of which the abundance of 84 proteins is downregulated and 15 proteins are enriched to the Type III secretion system, Type VI secretion system, and flagellum pathways. Functional analysis confirms virulent phenotypes, including autoaggregation, biofilm formation, hemolysis, swimming, and swarming, are impaired in LTB4-ROTC. Furthermore, the in vivo bacterial challenge in both tilapia and zebrafish infection models suggests that the virulence of LTB4-ROTC is attenuated. Analysis of immune gene expression shows that LTB4-ROTC induces a stronger immune response in the spleen but a weaker response in the head kidney than that induced by LTB4-S, suggesting it's a potential vaccine candidate. Zebrafish and tilapia were challenged with a sublethal dose of LTB4-ROTC as a live vaccine followed by LTB4-S challenge. The relative percentage of survival of zebrafish is 60% and that of tilapia is 75% after vaccination. Thus, our study suggests that bacteria that acquire antibiotic resistance may attenuate virulence, which can be explored as a potential live vaccine to tackle bacterial infection in aquaculture.


Subject(s)
Drug Resistance, Bacterial , Edwardsiella tarda , Enterobacteriaceae Infections , Oxytetracycline , Tilapia , Zebrafish , Edwardsiella tarda/pathogenicity , Edwardsiella tarda/drug effects , Edwardsiella tarda/genetics , Animals , Oxytetracycline/pharmacology , Virulence/drug effects , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/drug therapy , Tilapia/microbiology , Fish Diseases/microbiology , Fish Diseases/immunology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Proteomics/methods , Bacterial Vaccines/immunology
12.
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
13.
BMC Immunol ; 25(1): 27, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38706005

ABSTRACT

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


Subject(s)
Bacterial Vaccines , Klebsiella Infections , Klebsiella pneumoniae , Klebsiella pneumoniae/immunology , Bacterial Vaccines/immunology , Klebsiella Infections/immunology , Klebsiella Infections/prevention & control , Animals , Epitopes, T-Lymphocyte/immunology , Mice , Humans , Molecular Dynamics Simulation , Antigens, Bacterial/immunology , Oligodeoxyribonucleotides/immunology , Epitopes/immunology , Molecular Docking Simulation
14.
BMC Immunol ; 25(1): 46, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39034396

ABSTRACT

OBJECTIVES: The pathogenic microorganisms that cause intestinal diseases can significantly jeopardize people's health. Currently, there are no authorized treatments or vaccinations available to combat the germs responsible for intestinal disease. METHODS: Using immunoinformatics, we developed a potent multi-epitope Combination (combo) vaccine versus Salmonella and enterohemorrhagic E. coli. The B and T cell epitopes were identified by performing a conservancy assessment, population coverage analysis, physicochemical attributes assessment, and secondary and tertiary structure assessment of the chosen antigenic polypeptide. The selection process for vaccine development included using several bioinformatics tools and approaches to finally choose two linear B-cell epitopes, five CTL epitopes, and two HTL epitopes. RESULTS: The vaccine had strong immunogenicity, cytokine production, immunological properties, non-toxicity, non-allergenicity, stability, and potential efficacy against infections. Disulfide bonding, codon modification, and computational cloning were also used to enhance the stability and efficacy of expression in the host E. coli. The vaccine's structure has a strong affinity for the TLR4 ligand and is very durable, as shown by molecular docking and molecular modeling. The results of the immunological simulation demonstrated that both B and T cells had a heightened response to the vaccination component. CONCLUSIONS: The comprehensive in silico analysis reveals that the proposed vaccine will likely elicit a robust immune response against pathogenic bacteria that cause intestinal diseases. Therefore, it is a promising option for further experimental testing.


Subject(s)
Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Vaccinology , Humans , Epitopes, T-Lymphocyte/immunology , Vaccinology/methods , Epitopes, B-Lymphocyte/immunology , Vaccines, Combined/immunology , Genomics/methods , Enterohemorrhagic Escherichia coli/immunology , Salmonella/immunology , Animals , Computational Biology/methods , Molecular Docking Simulation , Escherichia coli Vaccines/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/immunology , Salmonella Infections/immunology , Salmonella Infections/prevention & control , Antigens, Bacterial/immunology , Vaccine Development/methods , Bacterial Vaccines/immunology
15.
BMC Microbiol ; 24(1): 249, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977999

ABSTRACT

Rhodococcus equi (R. equi) is a zoonotic opportunistic pathogen that mainly causes fatal lung and extrapulmonary abscesses in foals and immunocompromised individuals. To date, no commercial vaccine against R. equi exists. We previously screened all potential vaccine candidates from the complete genome of R. equi using a reverse vaccinology approach. Five of these candidates, namely ABC transporter substrate-binding protein (ABC transporter), penicillin-binding protein 2 (PBD2), NlpC/P60 family protein (NlpC/P60), esterase family protein (Esterase), and M23 family metallopeptidase (M23) were selected for the evaluation of immunogenicity and immunoprotective effects in BALB/c mice model challenged with R. equi. The results showed that all five vaccine candidate-immunized mice experienced a significant increase in spleen antigen-specific IFN-γ- and TNF-α-positive CD4 + and CD8 + T lymphocytes and generated robust Th1- and Th2-type immune responses and antibody responses. Two weeks after the R. equi challenge, immunization with the five vaccine candidates reduced the bacterial load in the lungs and improved the pathological damage to the lungs and livers compared with those in the control group. NlpC/P60, Esterase, and M23 were more effective than the ABC transporter and PBD2 in inducing protective immunity against R. equi challenge in mice. In addition, these vaccine candidates have the potential to induce T lymphocyte memory immune responses in mice. In summary, these antigens are effective candidates for the development of protective vaccines against R. equi. The R. equi antigen library has been expanded and provides new ideas for the development of multivalent vaccines.


Subject(s)
Actinomycetales Infections , Bacterial Vaccines , Disease Models, Animal , Immunity, Humoral , Mice, Inbred BALB C , Rhodococcus equi , Animals , Rhodococcus equi/immunology , Rhodococcus equi/genetics , Mice , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Actinomycetales Infections/prevention & control , Actinomycetales Infections/immunology , Actinomycetales Infections/microbiology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Immunity, Cellular , Female , Lung/microbiology , Lung/immunology , Lung/pathology , Bacterial Load , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism
16.
BMC Microbiol ; 24(1): 280, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068414

ABSTRACT

BACKGROUND: Enterococcus faecium and Staphylococcus aureus are the Gram-positive pathogens of the ESKAPE group, known to represent a great threat to human health due to their high virulence and multiple resistances to antibiotics. Combined, enterococci and S. aureus account for 26% of healthcare-associated infections and are the most common organisms responsible for blood stream infections. We previously showed that the peptidyl-prolyl cis/trans isomerase (PPIase) PpiC of E. faecium elicits the production of specific, opsonic, and protective antibodies that are effective against several strains of E. faecium and E. faecalis. Due to the ubiquitous characteristics of PPIases and their essential function within Gram-positive cells, we hypothesized a potential cross-reactive effect of anti-PpiC antibodies. RESULTS: Opsonophagocytic assays combined with bioinformatics led to the identification of the foldase protein PrsA as a new potential vaccine antigen in S. aureus. We show that PrsA is a stable dimeric protein able to elicit opsonic antibodies against the S. aureus strain MW2, as well as cross-binding and cross-opsonic in several S. aureus, E. faecium and E. faecalis strains. CONCLUSIONS: Given the multiple antibiotic resistances S. aureus and enterococci present, finding preventive strategies is essential to fight those two nosocomial pathogens. The study shows the potential of PrsA as an antigen to use in vaccine formulation against the two dangerous Gram-positive ESKAPE bacteria. Our findings support the idea that PPIases should be further investigated as vaccine targets in the frame of pan-vaccinomics strategy.


Subject(s)
Bacterial Proteins , Enterococcus faecalis , Enterococcus faecium , Peptidylprolyl Isomerase , Staphylococcus aureus , Staphylococcus aureus/immunology , Staphylococcus aureus/genetics , Enterococcus faecium/immunology , Enterococcus faecium/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Peptidylprolyl Isomerase/immunology , Peptidylprolyl Isomerase/genetics , Enterococcus faecalis/immunology , Enterococcus faecalis/genetics , Humans , Gram-Positive Bacterial Infections/prevention & control , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/microbiology , Bacterial Vaccines/immunology , Opsonin Proteins/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Animals , Cross Reactions , Mice , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Phagocytosis , Staphylococcal Infections/prevention & control , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology
17.
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
18.
Microb Pathog ; 195: 106913, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39236968

ABSTRACT

Bacterial septicemia represents a significant disease affecting cultured grass carp culture, with the primary etiological agent identified as the Gram-negative bacterium Aeromonas veronii. In response to an outbreak of septicemia in Guangzhou, we developed a formaldehyde-inactivated vaccine against an A. veronii strain designated AV-GZ21-2. This strain exhibited high pathogenicity in experimental infections across at all developmental stages of grass carp. Mortality rates for grass carp weighing 15 ± 5 g ranged from 16 % to 92 % at exposure temperatures of 19 °C-34 °C, respectively. The median lethal dose (LD50) for grass carp groups weighing 15 ± 5 g, 60 ± 10 g, 150 ± 30 g and 500 ± 50 g were determined to be 1.43, 2.52, 4.65 and 7.12 × 107(CFU/mL), respectively. We investigated the inactivated vaccine in conbination with aluminum hydroxide gel (AV-AHG), Montanide ISA201VG (AV-201VG), and white oil (AV-WO) adjuvants. This study aimed to optimize inactivation conditions and identify the adjuvant that elicits the most robust immune response. The AV-GZ21-2 inactivated bacterial solution (AV),when combined with various adjuvants, was capable of inducing a strong specific immune response in grass carp. The relative percent survival (RPS) following a lethal challenge with AV-GZ21-2 were 94 % for AV-AHG, 88 % for AV-201VG, 84 % for AV-WO and 78 % for AV alone. The minimum immunization dose of the AV-AHG vaccine was determined to be 6.0 × 107 CFU per fish, providing immunity for a duration of six months with an immune protection level exceeding 75 %. Furthermore, the AV-AHG vaccine demonstrated significant protective efficacy against various epidemic isolates of A. veronii. Consequently, we developed an inactivated vaccine targeting a highly pathogenic strain of A. veronii, incorporating an aluminum hydroxide gel adjuvant, which resulted in high immune protection and a duration of immunity exceeding six months. These findings suggest that the AV-AHG vaccine holds substantial potential for industrial application.


Subject(s)
Adjuvants, Immunologic , Aeromonas veronii , Bacterial Vaccines , Carps , Fish Diseases , Gram-Negative Bacterial Infections , Vaccines, Inactivated , Animals , Carps/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Aeromonas veronii/immunology , Fish Diseases/prevention & control , Fish Diseases/microbiology , Fish Diseases/immunology , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/immunology , Virulence , Adjuvants, Immunologic/administration & dosage , Lethal Dose 50 , Temperature , China/epidemiology , Aluminum Hydroxide/administration & dosage
19.
Microb Pathog ; 194: 106777, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002657

ABSTRACT

Francisella tularensis can cause severe disease in humans via the respiratory or cutaneous routes and a case fatality ratio of up to 10 % is reported due to lack of proper antibiotic treatment, while F. novicida causes disease in severely immunocompromised individuals. Efforts are needed to develop effective vaccine candidates against Francisella species. Thus, in this study, a systematic computational work frame was used to deeply investigate the whole proteome of Francisella novicida containing 1728 proteins to develop vaccine against F. tularensis and related species. Whole-proteome analysis revealed that four proteins including (A0Q492) (A0Q7Y4), (A0Q4N4), and (A0Q5D9) are the suitable vaccine targets after the removal of homologous, paralogous and prediction of subcellular localization. These proteins were used to predict the T cell, B cell, and HTL epitopes which were joined together through suitable linkers to construct a multi-epitopes vaccine (MEVC). The MEVC was found to be highly immunogenic and non-allergenic while the physiochemical properties revealed the feasible expression and purification. Moreover, the molecular interaction of MEVC with TLR2, molecular simulation, and binding free energy analyses further validated the immune potential of the construct. According to Jcat analysis, the refined sequence demonstrates GC contents of 41.48 % and a CAI value of 1. The in-silico cloning and optimization process ensured compatibility with host codon usage, thereby facilitating efficient expression. Computational immune simulation studies underscored the capacity of MEVC to induce both primary and secondary immune responses. The conservation analysis further revealed that the selected epitopes exhibit 100 % conservation across different species and thus provides wider protection against Francisella.


Subject(s)
Adaptive Immunity , Bacterial Vaccines , Francisella tularensis , Proteomics , Tularemia , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Francisella tularensis/immunology , Francisella tularensis/genetics , Tularemia/prevention & control , Tularemia/immunology , Tularemia/microbiology , Humans , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Proteome , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Vaccine Development , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL