Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Fish Shellfish Immunol ; 153: 109836, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147177

ABSTRACT

Pseudomonas plecoglossicida is one of most important pathogenic bacterial species in large yellow croaker and several other commercially valuable fish species. In our previous study, a GacS deficient mutant (ΔgacS) was constructed and its virulence showed substantially attenuated. In present study, the safety, immunogenicity and protective effect of the ΔgacS were evaluated in large yellow croaker as a live-attenuated vaccine candidate. It was shown that the ΔgacS strain exhibited good safety to large yellow croaker and there was no mortality or clinical symptoms observed in all fish that infected by ΔgacS strain with the doses range from 2 × 105~107 CFU per fish via intraperitoneal injection (IP) or immersion (IM), and almost all bacteria were cleaned up in the spleen of the fish at 14-day post infection. Specific antibodies could be detected at 7-day and 14-day post infection by direct agglutination method, and the valences of antibodies and bactericidal activities of the serum were significant increased with vaccination doses and vaccination time. Moreover, the expressions of some molecules and cytokines involved in specific immune responses were detected in the ΔgacS strain immunization group and control group. After challenged by the wild-type (WT) strain XSDHY-P, the relative percentage survival (RPS) showed highly correlated with the immunized dosage regardless of vaccination methods. It showed that the RPS of the IP groups were 39.47 %, 57.89 %, 71.05 % with the immune dosage in a descending order, respectively, and the RPS of the IM groups were 26.31 %, 36.84 %, 76.31 % with the immune dosage in a descending order, respectively. In summary, the ΔgacS strain exhibited safety and good protective effect to large yellow croaker and was a potential live vaccine candidate.


Subject(s)
Fish Diseases , Perciformes , Pseudomonas Infections , Pseudomonas , Vaccines, Attenuated , Animals , Fish Diseases/immunology , Fish Diseases/prevention & control , Perciformes/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Pseudomonas Infections/veterinary , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/genetics , Immunogenicity, Vaccine
3.
Fish Shellfish Immunol ; 151: 109688, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38857817

ABSTRACT

This study marks the first utilization of reverse vaccinology to develop recombinant subunit vaccines against Pseudomonas koreensis infection in Empurau (Tor tambroides). The proteome (5538 proteins) was screened against various filters to prioritize proteins based on features that are associated with virulence, subcellular localization, transmembrane helical structure, antigenicity, essentiality, non-homology with the host proteome, molecular weight, and stability, which led to the identification of eight potential vaccine candidates. These potential vaccine candidates were cloned and expressed, with six achieving successful expression and purification. The antigens were formulated into two distinct vaccine mixtures, Vac A and Vac B, and their protective efficacy was assessed through in vivo challenge experiments. Vac A and Vac B demonstrated high protective efficacies of 100 % and 81.2 %, respectively. Histological analyses revealed reduced tissue damage in vaccinated fish after experimental infection, with Vac A showing no adverse effects, whereas Vac B exhibited mild degenerative changes. Quantitative real-time PCR results showed a significant upregulation of TNF-α and downregulation of IL-1ß in the kidneys, spleen, gills, and intestine in both Vac A- and Vac B-immunized fish after challenged with P. koreensis. Additionally, IL-8 exhibits tissue-specific differential expression, with significant upregulation in the kidney, gills, and intestine, and downregulation in the spleen, particularly notable in Vac A-immunized fish. The research underscores the effectiveness of the reverse vaccinology approach in fish and demonstrates the promising potential of Vac A and Vac B as recombinant subunit vaccines.


Subject(s)
Fish Diseases , Pseudomonas Infections , Pseudomonas , Animals , Fish Diseases/immunology , Fish Diseases/prevention & control , Pseudomonas/immunology , Pseudomonas Infections/veterinary , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccinology , Vaccines, Synthetic/immunology , Cyprinidae/immunology , Pseudomonas Vaccines/immunology , Proteome/immunology
4.
Infect Immun ; 89(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33199354

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative pathogen that causes severe pulmonary infections associated with high morbidity and mortality in immunocompromised patients. The development of a vaccine against P. aeruginosa could help prevent infections caused by this highly antibiotic-resistant microorganism. We propose that identifying the vaccine-induced correlates of protection against P. aeruginosa will facilitate the development of a vaccine against this pathogen. In this study, we investigated the mechanistic correlates of protection of a curdlan-adjuvanted P. aeruginosa whole-cell vaccine (WCV) delivered intranasally. The WCV significantly decreased bacterial loads in the respiratory tract after intranasal P. aeruginosa challenge and raised antigen-specific antibody titers. To study the role of B and T cells during vaccination, anti-CD4, -CD8, and -CD20 depletions were performed prior to WCV vaccination and boosting. The depletion of CD4+, CD8+, or CD20+ cells had no impact on the bacterial burden in mock-vaccinated animals. However, depletion of CD20+ B cells, but not CD8+ or CD4+ T cells, led to the loss of vaccine-mediated bacterial clearance. Also, passive immunization with serum from WCV group mice alone protected naive mice against P. aeruginosa, supporting the role of antibodies in clearing P. aeruginosa We observed that in the absence of T cell-dependent antibody production, mice vaccinated with the WCV were still able to reduce bacterial loads. Our results collectively highlight the importance of the humoral immune response for protection against P. aeruginosa and suggest that the production of T cell-independent antibodies may be sufficient for bacterial clearance induced by whole-cell P. aeruginosa vaccination.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Bacterial/immunology , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/administration & dosage , Pseudomonas Vaccines/immunology , Animals , Humans , Immunization , Mice , Models, Animal , Pneumonia, Bacterial/physiopathology , Pseudomonas Infections/physiopathology , Vaccination
5.
Biol Pharm Bull ; 43(10): 1469-1475, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32779581

ABSTRACT

Hospital-acquired infections with Pseudomonas aeruginosa have become a great challenge in caring for critically ill and immunocompromised patients. The cause of high mortality is the presence of multi-drug resistant (MDR) strains, which confers a pressing need for vaccines. Although vaccines against P. aeruginosa have been in development for more than several decades, there is no vaccine for patients at present. In this study, we purified genomic DNA of P. aeruginosa from sera of patients affected, constructed genome-wide library with random recombinants, and screened candidate protein antigens by evaluating their protective effects in vivo. After 13-round of screening, 115 reactive recombinants were obtained, among which 13 antigens showed strong immunoreactivity (more than 10% reaction to PcrV, a well-characterized V-antigen of P. aeruginosa). These 13 antigens were: PpiA, PtsP, OprP, CAZ10_34235, HmuU_2, PcaK, CarAd, RecG, YjiR_5, LigD, KinB, RtcA, and PscF. In vivo studies showed that vaccination with PscF protected against lethal P. aeruginosa challenge, and decreased lung inflammation and injury. A genomic library of P. aeruginosa could be constructed in this way for the first time, which could not only screen candidate antigens but also in a high-throughput way. PscF was considered as an ideal promising vaccine candidate for combating P. aeruginosa infection and was supported for further evaluation of its safety and efficacy.


Subject(s)
Antigens, Bacterial/genetics , Cross Infection/prevention & control , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Cross Infection/blood , Cross Infection/immunology , Cross Infection/microbiology , DNA, Bacterial/genetics , Disease Models, Animal , Female , Genomic Library , High-Throughput Screening Assays/methods , Humans , Immunogenicity, Vaccine , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Pseudomonas Infections/blood , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Vaccines/administration & dosage , Pseudomonas Vaccines/genetics , Pseudomonas aeruginosa/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
Proc Natl Acad Sci U S A ; 113(41): E6153-E6161, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27671632

ABSTRACT

Infection is the single greatest threat to survival during cancer chemotherapy because of depletion of bone marrow-derived immune cells. Phagocytes, especially neutrophils, are key effectors in immunity to extracellular pathogens, which has limited the development of new approaches to protect patients with cancer and chemotherapy-induced neutropenia. Using a model of vaccine-induced protection against lethal Pseudomonas aeruginosa pneumonia in the setting of chemotherapy-induced neutropenia, we found a population of resident lung macrophages in the immunized lung that mediated protection in the absence of neutrophils, bone marrow-derived monocytes, or antibodies. These vaccine-induced macrophages (ViMs) expanded after immunization, locally proliferated, and were closely related to alveolar macrophages (AMs) by surface phenotype and gene expression profiles. By contrast to AMs, numbers of ViMs were stable through chemotherapy, showed enhanced phagocytic activity, and prolonged survival of neutropenic mice from lethal P. aeruginosa pneumonia upon intratracheal adoptive transfer. Thus, induction of ViMs by tissue macrophage remodeling may become a framework for new strategies to activate immune-mediated reserves against infection in immunocompromised hosts.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Marrow/drug effects , Bone Marrow/immunology , Disease Resistance/immunology , Host-Pathogen Interactions/immunology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Animals , Biomarkers , Bone Marrow/pathology , Cell Cycle/genetics , Disease Models, Animal , Gene Expression Profiling , Host-Pathogen Interactions/genetics , Immunophenotyping , Macrophages, Alveolar/metabolism , Mice , Mice, Knockout , Neutropenia/etiology , Phagocytosis/genetics , Phagocytosis/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Survival Analysis
7.
Microbiol Immunol ; 62(12): 774-785, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30378708

ABSTRACT

An effective vaccine against Pseudomonas aeruginosa would be hugely beneficial to people who are susceptible to the serious infections it can cause. Vaccination against PcrV of the P. aeruginosa type III secretion system is a potential prophylactic strategy for improving the incidence and prognosis of P. aeruginosa pneumonia. Here, the effect of nasal PcrV adjuvanted with CpG oligodeoxynucleotide (CpG) was compared with a nasal PcrV/aluminum hydroxide gel (alum) vaccine. Seven groups of mice were vaccinated intranasally with one of the following: 1, PcrV-CpG; 2, PcrV-alum; 3, PcrV alone; 4, CpG alone; 5, alum alone; 6 and 7, saline control. Fifty days after the first immunization, anti-PcrV IgG, IgA and IgG isotype titers were measured; significant increases in these titers were detected only in the PcrV-CpG vaccinated mice. The vaccinated mice were then intratracheally infected with a lethal dose of P. aeruginosa and their body temperatures and survival monitored for 24 hr, edema, bacteria, myeloperoxidase activity and lung histology also being evaluated at 24 hr post-infection. It was found that 73% of the PcrV-CpG-vaccinated mice survived, whereas fewer than 30% of the mice vaccinated with PcrV-alum or adjuvant alone survived. Lung edema and other inflammation-related variables were less severe in the PcrV-CpG group. The significant increase in PcrV-specific IgA titers detected following PcrV-CpG vaccination is probably a component of the disease protection mechanism. Overall, our data show that intranasal PcrV-CpG vaccination has potential efficacy for clinical application against P. aeruginosa pneumonia.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Oligodeoxyribonucleotides/immunology , Pneumonia/prevention & control , Pore Forming Cytotoxic Proteins/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/drug effects , Vaccination , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Toxins/genetics , Body Temperature , Disease Models, Animal , Edema , Lung/immunology , Lung/pathology , Male , Mice , Oligodeoxyribonucleotides/genetics , Peroxidase/analysis , Pore Forming Cytotoxic Proteins/genetics , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Vaccines/administration & dosage , Pseudomonas aeruginosa/pathogenicity , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Rate , Type III Secretion Systems/immunology
8.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 49(1): 13-17, 2018 Jan.
Article in Zh | MEDLINE | ID: mdl-29737082

ABSTRACT

OBJECTIVE: To construct the recombinant Bb-pGEX-OprI vaccine of Pseudomonas aeruginosa (Pa) outer membrane protein I (OprI) and study its protection effect in mice against Pa. METHODS: The OprI gene was amplified by PCR,and cloned into pGEX-1λT to generate pGEX-OprI. The pGEX-OprI was transformed into Bifidobacterium bifidum(Bb) to construct recombinant Bb-pGEX-OprI vaccine by electroporation. After identification with double enzyme digestion,PCR and sequencing,the vaccine was then induced with IPTG,and its expression was analyzed and identified by SDS-PAGE and Western blot respectively. Twenty-one mice were randomly divided into 3 groups and vaccinated by intragastric administration with Bb-pGEX-OprI,Bb-pGEX-1λT and Bb respectively. All mice were challenged with PA01 strain at 4 weeks after the first vaccination. At 2 weeks after the challenge,mice were sacrificed to separate their lungs,and the numbers of bacterial colonies in lungs were counted. Venous blood was collected before vaccination,at 4 weeks after the first vaccination and 2 weeks after the challenge of PA01 strain. The serum IgG,IgG subclasses and IgE were detected by routine ELISA. RESULTS: The OprI gene of 194 bp was successfully amplified by PCR. Double enzyme digestion,PCR and sequencing confirmed that the OprI gene was successfully cloned into pGEX-1λT and pGEX-OprI was successfully transformed into Bb,constructing the Bb-pGEX-OprI vaccine. SDS-PAGE indicated that Bb-pGEX-OprI vaccine expressed an OprI-GST fusion protein with the relative molecular mass of approximately 32×103. Western blot verified that the fusion protein could be specifically identified by the sera of mice infected with Pa. The number of bacterial colonies in lung of Bb-pGEX-OprI vaccine group was lower than that of Bb-pGEX-1λT or Bb control ( P<0.01). The levels of serum IgG,IgG2b,IgG3 and IgE in Bb-pGEX-OprI vaccine group rose at 4 weeks after the first vaccination and 2 weeks after the challenge successively. The levels of serum antibodies in Bb-pGEX-OprI vaccine group were higher than those in Bb-pGEX-1λT or Bb control at the same time point ( P<0.01 or P<0.05). CONCLUSION: The recombinant Bb-pGEX-OprI vaccine was successfully constructed and produced an effective humoral immune response against the Pa infection.


Subject(s)
Bacterial Proteins/immunology , Lipoproteins/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Animals , Antibodies, Bacterial/blood , Immunity, Humoral , Immunoglobulin E/blood , Immunoglobulin G/blood , Mice , Pseudomonas aeruginosa , Random Allocation , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/immunology
9.
Microbiol Immunol ; 61(2): 64-74, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28370521

ABSTRACT

Vaccination against the type III secretion system of P. aeruginosa is a potential prophylactic strategy for reducing the incidence and improving the poor prognosis of P. aeruginosa pneumonia. In this study, the efficacies of three different adjuvants, Freund's adjuvant (FA), aluminum hydroxide (alum) and CpG oligodeoxynucleotide (ODN), were examined from the viewpoint of inducing PcrV-specific immunity against virulent P. aeruginosa. Mice that had been immunized intraperitoneally with recombinant PcrV formulated with one of the above adjuvants were challenged intratracheally with a lethal dose of P. aeruginosa. The PcrV-FA immunized group attained a survival rate of 91%, whereas the survival rates of the PcrV-alum and PcrV-CpG groups were 73% and 64%, respectively. In terms of hypothermia recovery after bacterial instillation, PcrV-alum was the most protective, followed by PcrV-FA and PcrV-CpG. The lung edema index was lower in the PcrV-CpG vaccination group than in the other groups. PcrV-alum immunization was associated with the greatest decrease in myeloperoxidase in infected lungs, and also decreased the number of lung bacteria to a similar number as in the PcrV-FA group. There was less neutrophil recruitment in the lungs of mice vaccinated with PcrV-alum or PcrV-CpG than in those of mice vaccinated with PcrV-FA or PcrV alone. Overall, in terms of mouse survival the PcrV-CpG vaccine, which could be a relatively safe next-generation vaccine, showed a comparable effect to the PcrV-alum vaccine.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Pneumonia, Bacterial/prevention & control , Pore Forming Cytotoxic Proteins/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/immunology , Aluminum Hydroxide/administration & dosage , Animals , Bacterial Load , Freund's Adjuvant/administration & dosage , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred ICR , Oligodeoxyribonucleotides/administration & dosage , Pseudomonas Vaccines/administration & dosage , Survival Analysis , Treatment Outcome
10.
Biologicals ; 47: 11-17, 2017 May.
Article in English | MEDLINE | ID: mdl-28404459

ABSTRACT

Mucoid strains of Pseudomonas aeruginosa are closely associated with chronic pulmonary infections. In this report we describe a straightforward approach to conjugate high molecular weight alginate to type b-flagellin (FLB) and investigation of its bioactivity. The conjugation process was performed by using ADH and EDAC. The endotoxin was eliminated from the candidate vaccine by LPS removal resin followed by LAL test. The bioconjugate molecules were verified by simultaneously determination of polysaccharide/protein content followed by gel filtration chromatography and FTIR spectroscopy. Groups of eight BALB/c mice were injected intranasally with 5 µg (per each nostril) of purified alginate, FLB and conjugated alginate-FLB with two week intervals. The functional activity of the vaccine was evaluated by ELISA and opsonophagocytosis tests. Vaccination with the alginate-FLB conjugate induced a significant (P = 0.0033) rise in alginate specific IgG in mice. At all dilution ranges, the opsonic activity of the conjugate vaccine antisera was significantly higher than alginate alone (61.9% vs. 17.3% at 1:4 dilution; P = 0.0067). The alginate-FLB conjugate could elicit high specific antibodies titer against alginate by improving its immunogenicity. In addition, the antisera raised against conjugate vaccine act as a suitable opsonin for phagocytosis of the mucoid strains of P. aeruginosa.


Subject(s)
Flagellin , Immunoconjugates , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines , Pseudomonas aeruginosa , Animals , Female , Flagellin/chemistry , Flagellin/immunology , Flagellin/pharmacology , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/pharmacology , Mice , Mice, Inbred BALB C , Pseudomonas Infections/immunology , Pseudomonas Infections/pathology , Pseudomonas Vaccines/chemistry , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/pharmacology , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/immunology
11.
Microbiol Immunol ; 59(9): 533-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26249788

ABSTRACT

Pseudomonas aeruginosa poses a major threat to human health and to the mink industry. Thus, development of vaccines that elicit robust humoral and cellular immunity against P. aeruginosa is greatly needed. In this study, a recombinant attenuated Salmonella vaccine (RASV) that expresses the outer membrane proteins fusion OprF190-342 -OprI21-83 (F1I2) from P. aeruginosa was constructed and the potency of this vaccine candidate assessed by measuring F1I2-specific humoral immune responses upon vaccination through s.c. or oral routes. S.C. administration achieved higher serum IgG titers and IgA titers in the intestine and induced stronger F1I2-specific IgG and IgA titers in lung homogenate than did oral administration, which resulted in low IgG titers and no local IgA production. High titers of IFN-γ, IL-4, and T-lymphocyte subsets induced a mixed Th1/Th2 response in mice immunized s.c., indicating elicitation of cellular immunity. Importantly, when immunized mice were challenged with P. aeruginosa by the intranasal route 30 days after the initial immunization, s.c. vaccination achieved 77.78% protection, in contrast to 41.18% via oral administration and 66.67% via Escherichia coli-expressed F1I2 (His-F1I2) vaccination. These results indicate that s.c. vaccination provides a better protective response against P. aeruginosa infection than do oral administration and the His-F1I2 vaccine.


Subject(s)
Bacterial Proteins/immunology , Drug Carriers , Lipoproteins/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/immunology , Salmonella typhimurium/genetics , Administration, Oral , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Bacterial Proteins/genetics , Disease Models, Animal , Escherichia coli , Female , Immunoglobulin A/analysis , Immunoglobulin G/blood , Injections, Subcutaneous , Interferon-gamma/metabolism , Interleukin-4/metabolism , Intestinal Mucosa/immunology , Lipoproteins/genetics , Lung/immunology , Mice, Inbred BALB C , Mink , Pseudomonas Vaccines/administration & dosage , Pseudomonas Vaccines/genetics , Pseudomonas aeruginosa/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/genetics , Salmonella typhimurium/growth & development , Serum/immunology , Survival Analysis , T-Lymphocyte Subsets/immunology , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
12.
Appl Microbiol Biotechnol ; 99(2): 667-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25381907

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that localizes to and colonizes mucosal tissue. Thus, vaccines that elicit a strong mucosal response against P. aeruginosa should be superior to other vaccination strategies. In this study, to stimulate rapid and enhanced mucosal immune responses, mannose-modified chitosan microspheres loaded with the recombinant outer membrane protein OprF190-342-OprI21-83 (FI) (FI-MCS-MPs) of P. aeruginosa were developed as a potent subunit vaccine for mucosal delivery. FI-MCS-MPs were successfully obtained via the tripolyphosphate ionic crosslinking method. Confocal and immunohistochemical analyses indicated that FI-MCS-MPs exhibited the ability to bind the macrophage mannose receptor (MMR, CD206) in vitro and in vivo. After intranasal immunization of mice with FI-MCS-MPs, FI-specific humoral immune responses were detected, measured as local IgM antibody titers in lung tissue slurry; IgA antibody titers in nasal washes, bronchoalveolar lavage (BAL), and intestinal lavage; and systemic IgA and IgG antibody titers in serum. FI-MCS-MPs induced early and high mucosal and systemic humoral antibody responses comparable to those in the group vaccinated with unmodified mannose. High levels of IFN-γ and IL-4 in addition to T lymphocyte subsets induced a mixed Th1/Th2 response in mice immunized with FI-MCS-MPs, resulting in the establishment of cellular immunity. Additionally, when immunized mice were challenged with P. aeruginosa via the nasal cavity, FI-MCS-MPs demonstrated 75 % protective efficacy. Together, these data indicate that mannose-modified chitosan microspheres are a promising subunit delivery system for vaccines against P. aeruginosa infection.


Subject(s)
Chitosan/pharmacology , Immunity, Mucosal , Mannose/pharmacology , Pseudomonas Infections/immunology , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antibody Formation , Bacterial Proteins/immunology , Base Sequence , Cell Line , Chitosan/chemistry , Female , Immunoglobulin A/blood , Immunoglobulin A/chemistry , Immunoglobulin G/blood , Interferon-gamma/immunology , Interleukin-4/immunology , Lipoproteins/immunology , Macrophages/chemistry , Macrophages/immunology , Mannose/chemistry , Mice , Mice, Inbred BALB C , Microspheres , Molecular Sequence Data , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/chemistry , Recombinant Proteins/immunology , T-Lymphocytes/immunology
13.
Front Immunol ; 15: 1372349, 2024.
Article in English | MEDLINE | ID: mdl-38698863

ABSTRACT

Pseudomonas aeruginosa (Pa) is an opportunistic bacterial pathogen responsible for severe hospital acquired infections in immunocompromised and elderly individuals. Emergence of increasingly drug resistant strains and the absence of a broad-spectrum prophylactic vaccine against both T3SA+ (type III secretion apparatus) and ExlA+/T3SA- Pa strains worsen the situation in a post-pandemic world. Thus, we formulated a candidate subunit vaccine (called ExlA/L-PaF/BECC/ME) against both Pa types. This bivalent vaccine was generated by combining the C-terminal active moiety of exolysin A (ExlA) produced by non-T3SA Pa strains with our T3SA-based vaccine platform, L-PaF, in an oil-in-water emulsion. The ExlA/L-PaF in ME (MedImmune emulsion) was then mixed with BECC438b, an engineered lipid A analogue and a TLR4 agonist. This formulation was administered intranasally (IN) to young and elderly mice to determine its potency across a diverse age-range. The elderly mice were used to mimic the infection seen in elderly humans, who are more susceptible to serious Pa disease compared to their young adult counterparts. After Pa infection, mice immunized with ExlA/L-PaF/BECC/ME displayed a T cell-mediated adaptive response while PBS-vaccinated mice experienced a rapid onset inflammatory response. Important genes and pathways were observed, which give rise to an anti-Pa immune response. Thus, this vaccine has the potential to protect aged individuals in our population from serious Pa infection.


Subject(s)
Emulsions , Pseudomonas Infections , Pseudomonas Vaccines , Pseudomonas aeruginosa , Vaccines, Subunit , Animals , Pseudomonas aeruginosa/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/administration & dosage , Female , Vaccine Development , Humans , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Disease Models, Animal , Bacterial Proteins/immunology , Bacterial Proteins/genetics
14.
Nat Commun ; 15(1): 6766, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117651

ABSTRACT

Live vaccines are ideal for inducing immunity but suffer from the need to attenuate their pathogenicity or replication to preclude the possibility of escape. Unnatural amino acids (UAAs) provide a strategy to engineer stringent auxotrophies, yielding conditionally replication incompetent live bacteria with excellent safety profiles. Here, we engineer Pseudomonas aeruginosa to maintain auxotrophy for the UAA p-benzoyl-L-phenylalanine (BzF) through its incorporation into the essential protein DnaN. In vivo evolution using an Escherichia coli-based two-hybrid selection system enabled engineering of a mutant DnaN homodimeric interface completely dependent on a BzF-specific interaction. This engineered strain, Pa Vaccine, exhibits undetectable escape frequency (<10-11) and shows excellent safety in naïve mice. Animals vaccinated via intranasal or intraperitoneal routes are protected from lethal challenge with pathogenic P. aeruginosa PA14. These results establish UAA-auxotrophic bacteria as promising candidates for bacterial vaccine therapy and outline a platform for expanding this technology to diverse bacterial pathogens.


Subject(s)
Pseudomonas Infections , Pseudomonas Vaccines , Pseudomonas aeruginosa , Animals , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Mice , Female , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/genetics , Amino Acids , Phenylalanine/analogs & derivatives , Escherichia coli/immunology , Escherichia coli/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Mice, Inbred BALB C
15.
Toxins (Basel) ; 16(6)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38922165

ABSTRACT

Extensively drug-resistant Pseudomonas aeruginosa infections are emerging as a significant threat associated with adverse patient outcomes. Due to this organism's inherent properties of developing antibiotic resistance, we sought to investigate alternative strategies such as identifying "high value" antigens for immunotherapy-based purposes. Through extensive database mining, we discovered that numerous Gram-negative bacterial (GNB) genomes, many of which are known multidrug-resistant (MDR) pathogens, including P. aeruginosa, horizontally acquired the evolutionarily conserved gene encoding Zonula occludens toxin (Zot) with a substantial degree of homology. The toxin's genomic footprint among so many different GNB stresses its evolutionary importance. By employing in silico techniques such as proteomic-based phylogenetic tracing, in conjunction with comparative structural modeling, we discovered a highly conserved intermembrane associated stretch of 70 amino acids shared among all the GNB strains analyzed. The characterization of our newly identified antigen reveals it to be a "high value" vaccine candidate specific for P. aeruginosa. This newly identified antigen harbors multiple non-overlapping B- and T-cell epitopes exhibiting very high binding affinities and can adopt identical tertiary structures among the least genetically homologous P. aeruginosa strains. Taken together, using proteomic-driven reverse vaccinology techniques, we identified multiple "high value" vaccine candidates capable of eliciting a polarized immune response against all the P. aeruginosa genetic variants tested.


Subject(s)
Phylogeny , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Humans , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/chemistry , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics
16.
Infect Immun ; 81(12): 4509-18, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24082079

ABSTRACT

Bacteria can naturally secrete outer membrane vesicles (OMVs) as pathogenic factors, while these vesicles may also serve as immunologic regulators if appropriately prepared. However, it is largely unknown whether Pseudomonas aeruginosa OMVs can activate inflammatory responses and whether immunization with OMVs can provide immune protection against subsequent infection. We purified and identified OMVs, which were then used to infect lung epithelial cells in vitro as well as C57BL/6J mice to investigate the immune response and the underlying signaling pathway. The results showed that OMVs generated from P. aeruginosa wild-type strain PAO1 were more cytotoxic to alveolar epithelial cells than those from quorum-sensing (QS)-deficient strain PAO1-ΔlasR. The levels of Toll-like receptor 4 (TLR4) and proinflammatory cytokines, including interleukin-1ß (IL-1ß) and IL-6, increased following OMV infection. Compared with lipopolysaccharide (LPS), lysed OMVs in which the membrane structures were broken induced a weak immune response. Furthermore, expression levels of TLR4-mediated responders (i.e., cytokines) were markedly downregulated by the TLR4 inhibitor E5564. Active immunization with OMVs or passive transfer of sera with a high cytokine quantity acquired from OMV-immunized mice could protect healthy mice against subsequent lethal PAO1 challenges (1.5 × 10(11) CFU). Collectively, these findings indicate that naturally secreted P. aeruginosa OMVs may trigger significant inflammatory responses via the TLR4 signaling pathway and protect mice against pseudomonal lung infection.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Cytoplasmic Vesicles/immunology , Pseudomonas Infections/immunology , Pseudomonas Vaccines/immunology , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Epithelial Cells , Humans , Immunization , Inflammation/immunology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipid A/analogs & derivatives , Lipid A/pharmacology , Lipopolysaccharides , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/immunology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/microbiology , Respiratory Mucosa/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/antagonists & inhibitors , Vaccination
17.
J Immunol ; 185(2): 1142-9, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20566829

ABSTRACT

TLRs are required for generation of protective lung mucosal immune responses against microbial pathogens. In this study, we evaluated the effect of the TLR5 ligand flagellin on stimulation of antibacterial mucosal immunity in a lethal murine Pseudomonas aeruginosa pneumonia model. The intranasal pretreatment of mice with purified P. aeruginosa flagellin induced strong protection against intratracheal P. aeruginosa-induced lethality, which was attributable to markedly improved bacterial clearance, reduced dissemination, and decreased alveolar permeability. The protective effects of flagellin on survival required TLR5 and were observed even in the absence of neutrophils. Flagellin induced strong induction of innate genes, most notably the antimicrobial peptide cathelicidin-related antimicrobial peptide. Finally, flagellin-induced protection was partially abrogated in cathelicidin-related antimicrobial peptide-deficient mice. Our findings illustrate the profound stimulatory effect of flagellin on lung mucosal innate immunity, a response that might be exploited therapeutically to prevent the development of gram-negative bacterial infection of the respiratory tract.


Subject(s)
Cathelicidins/immunology , Flagellin/immunology , Immunity, Mucosal/immunology , Lung/immunology , Toll-Like Receptor 5/immunology , Administration, Intranasal , Animals , Antimicrobial Cationic Peptides , Blotting, Western , Cathelicidins/genetics , Cathelicidins/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Flagellin/administration & dosage , Gene Expression/drug effects , Gene Expression/immunology , Immunity, Mucosal/drug effects , Immunity, Mucosal/genetics , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Lung/metabolism , Lung/microbiology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/administration & dosage , Pseudomonas Vaccines/immunology , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism
18.
Article in Russian | MEDLINE | ID: mdl-22830272

ABSTRACT

AIM: Production of preparation consisting of amino acid sequences of 2 proteins of outer membrane--OprF and OprI--of P. aeruginosa and study of its protective properties from experimental P. aeruginosa infection. MATERIALS AND METHODS: Nucleotide sequences coding OprF protein (1 kb) as well as its C-terminal region (0.6 kb) and OprI protein (0.25 kb) were integrated into pQE-30 plasmid (QIAGEN). And oprF gene (C-terminal region of oprF in variant 2) and oprI gene were combined and cloned sequentially into a single vector. E. coli M15 strain cells (QIAGEN) were used for the production of producent strains of recombinant proteins. Protein products were analyzed by electrophoresis in polyacrylamide gel by Lammle. Purification of recombinant proteins was performed by affinity chromatography in Ni-sepharose columns. Live virulent culture P. aeruginosa PA-170015 strain was used for the analysis of protective properties of recombinant proteins. RESULTS: 2 hybrid recombinant proteins were produced including amino acid sequences of F and I proteins of outer membrane (OprF and OprI) of P. aeruginosa. Recombinant protein 1 included whole size sequences of OprF and OprI and protein 2--C-terminal region (including amino acid residues 192-342) of OprF and whole size sequence of OprI. These recombinant proteins after 2 immunizations protected mice from the experimental intraperitoneal infection with P. aeruginosa. Hybrid protein consisting of whole size sequences had the best protective effect. CONCLUSION: The results obtained open a perspective for further immunobiological testing of hybrid recombinant protein OprF-OprI with the aim of creating immunopreparations for prophylaxis of P. aeruginosa infection.


Subject(s)
Bacterial Proteins/genetics , Lipoproteins/genetics , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/immunology , Recombinant Fusion Proteins/isolation & purification , Animals , Bacterial Proteins/immunology , Chromatography, Affinity , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Immunization , Lethal Dose 50 , Lipoproteins/immunology , Mice , Open Reading Frames , Plasmids/genetics , Polymerase Chain Reaction , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Vaccines/administration & dosage , Pseudomonas aeruginosa/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
19.
Sci Rep ; 12(1): 22324, 2022 12 24.
Article in English | MEDLINE | ID: mdl-36566282

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen considered a common cause of nosocomial infection with high morbidity and mortality in burn patients. Immunoprophylaxis techniques may lower the mortality rate of patients with burn wounds infected by P. aeruginosa; consequently, this may be an efficient strategy to manage infections caused by this bacterium. Several pathogenic Gram-negative bacteria like P. aeruginosa release outer membrane vesicles (OMVs), and structurally OMV consists of several antigenic components capable of generating a wide range of immune responses. Here, we evaluated the immunogenicity and efficacy of P. aeruginosa PA-OMVs (PA-OMVs) conjugated with the diphtheria toxoid (DT) formulated with alum adjuvant (PA-OMVs-DT + adj) in a mice model of burn wound infection. ELISA results showed that in the group of mice immunized with PA-OMVs-DT + adj conjugated, there was a significant increase in specific antibodies titer compared to non-conjugated PA-OMVs or control groups. In addition, the vaccination of mice with PA-OMVs-DT + adj conjugated generated greater protective effectiveness, as seen by lower bacterial loads, and eightfold decreased inflammatory cell infiltration with less tissue damage in the mice burn model compared to the control group. The opsonophagocytic killing results confirmed that humoral immune response might be critical for PA-OMVs mediated protection. These findings suggest that PA-OMV-DT conjugated might be used as a new vaccine against P. aeruginosa in burn wound infection.


Subject(s)
Burns , Diphtheria Toxoid , Pseudomonas Vaccines , Pseudomonas aeruginosa , Wound Infection , Animals , Mice , Bacterial Outer Membrane Proteins/immunology , Burns/microbiology , Diphtheria Toxoid/immunology , Pseudomonas aeruginosa/immunology , Wound Infection/microbiology , Wound Infection/prevention & control , Pseudomonas Vaccines/immunology
20.
Infect Immun ; 79(3): 1289-99, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21149583

ABSTRACT

Many animal studies investigating adaptive immune effectors important for protection against Pseudomonas aeruginosa have implicated opsonic antibody to the antigenically variable lipopolysaccharide (LPS) O antigens as a primary effector. However, active and passive vaccination of humans against these antigens has not shown clinical efficacy. We hypothesized that optimal immunity would require inducing multiple immune effectors targeting multiple bacterial antigens. Therefore, we evaluated a multivalent live-attenuated mucosal vaccination strategy in a murine model of acute P. aeruginosa pneumonia to assess the contributions to protective efficacy of various bacterial antigens and host immune effectors. Vaccines combining 3 or 4 attenuated strains having different LPS serogroups were associated with the highest protective efficacy compared to vaccines with fewer components. Levels of opsonophagocytic antibodies, which were directed not only to the LPS O antigens but also to the LPS core and surface proteins, correlated with protective immunity. The multivalent live-attenuated vaccines overcame prior problems involving immunologic interference in the development of O-antigen-specific antibody responses when closely related O antigens were combined in multivalent vaccines. Antibodies to the LPS core were associated with in vitro killing and in vivo protection against strains with O antigens not expressed by the vaccine strains, whereas antibodies to the LPS core and surface proteins augmented the contribution of O-antigen-specific antibodies elicited by vaccine strains containing a homologous O antigen. Local CD4 T cells in the lung also contributed to vaccine-based protection when opsonophagocytic antibodies to the challenge strain were absent. Thus, multivalent live-attenuated vaccines elicit multifactorial protective immunity to P. aeruginosa lung infections.


Subject(s)
Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Disease Models, Animal , Female , Immunity, Mucosal/immunology , Mice , Mice, Inbred C3H , O Antigens/immunology , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/microbiology , Pseudomonas Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL