Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.661
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 21(8): 857-867, 2020 08.
Article in English | MEDLINE | ID: mdl-32601469

ABSTRACT

Familial Mediterranean fever (FMF) is an autoinflammatory disease caused by homozygous or compound heterozygous gain-of-function mutations in MEFV, which encodes pyrin, an inflammasome protein. Heterozygous carrier frequencies for multiple MEFV mutations are high in several Mediterranean populations, suggesting that they confer selective advantage. Among 2,313 Turkish people, we found extended haplotype homozygosity flanking FMF-associated mutations, indicating evolutionarily recent positive selection of FMF-associated mutations. Two pathogenic pyrin variants independently arose >1,800 years ago. Mutant pyrin interacts less avidly with Yersinia pestis virulence factor YopM than with wild-type human pyrin, thereby attenuating YopM-induced interleukin (IL)-1ß suppression. Relative to healthy controls, leukocytes from patients with FMF harboring homozygous or compound heterozygous mutations and from asymptomatic heterozygous carriers released heightened IL-1ß specifically in response to Y. pestis. Y. pestis-infected MefvM680I/M680I FMF knock-in mice exhibited IL-1-dependent increased survival relative to wild-type knock-in mice. Thus, FMF mutations that were positively selected in Mediterranean populations confer heightened resistance to Y. pestis.


Subject(s)
Disease Resistance/genetics , Familial Mediterranean Fever/genetics , Plague , Pyrin/genetics , Selection, Genetic/genetics , Animals , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Disease Resistance/immunology , Haplotypes , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Mutation , Plague/immunology , Plague/metabolism , Pyrin/immunology , Pyrin/metabolism , Turkey , Virulence Factors/immunology , Virulence Factors/metabolism , Yersinia pestis
2.
Mol Cell Proteomics ; 23(5): 100753, 2024 May.
Article in English | MEDLINE | ID: mdl-38527648

ABSTRACT

Bacterial or viral antigens can contain subdominant protein regions that elicit weak antibody responses upon vaccination or infection although there is accumulating evidence that antibody responses against subdominant regions can enhance the protective immune response. One proposed mechanism for subdominant protein regions is the binding of host proteins that prevent antibody production against epitopes hidden within the protein binding interfaces. Here, we used affinity purification combined with quantitative mass spectrometry (AP-MS) to examine the level of competition between antigen-specific antibodies and host-pathogen protein interaction networks using the M1 protein from Streptococcus pyogenes as a model system. As most humans have circulating antibodies against the M1 protein, we first used AP-MS to show that the M1 protein interspecies protein network formed with human plasma proteins is largely conserved in naïve mice. Immunizing mice with the M1 protein generated a time-dependent increase of anti-M1 antibodies. AP-MS analysis comparing the composition of the M1-plasma protein network from naïve and immunized mice showed significant enrichment of 292 IgG peptides associated with 56 IgG chains in the immune mice. Despite the significant increase of bound IgGs, the levels of interacting plasma proteins were not significantly reduced in the immune mice. The results indicate that the antigen-specific polyclonal IgG against the M1 protein primarily targets epitopes outside the other plasma protein binding interfaces. In conclusion, this study demonstrates that AP-MS is a promising strategy to determine the relationship between antigen-specific antibodies and host-pathogen interaction networks that could be used to define subdominant protein regions of relevance for vaccine development.


Subject(s)
Antigens, Bacterial , Immunoglobulin G , Protein Binding , Streptococcus pyogenes , Animals , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Mice , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Adaptive Immunity , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Antibodies, Bacterial/immunology , Protein Interaction Maps , Mass Spectrometry , Carrier Proteins/metabolism , Carrier Proteins/immunology , Female , Host-Pathogen Interactions/immunology
3.
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
4.
J Biol Chem ; 299(8): 104980, 2023 08.
Article in English | MEDLINE | ID: mdl-37390991

ABSTRACT

Coiled coil-forming M proteins of the widespread and potentially deadly bacterial pathogen Streptococcus pyogenes (strep A) are immunodominant targets of opsonizing antibodies. However, antigenic sequence variability of M proteins into >220 M types, as defined by their hypervariable regions (HVRs), is considered to limit M proteins as vaccine immunogens because of type specificity in the antibody response. Surprisingly, a multi-HVR immunogen in clinical vaccine trials was shown to elicit M-type crossreactivity. The basis for this crossreactivity is unknown but may be due in part to antibody recognition of a 3D pattern conserved in many M protein HVRs that confers binding to human complement C4b-binding protein (C4BP). To test this hypothesis, we investigated whether a single M protein immunogen carrying the 3D pattern would elicit crossreactivity against other M types carrying the 3D pattern. We found that a 34-amino acid sequence of S. pyogenes M2 protein bearing the 3D pattern retained full C4BP-binding capacity when fused to a coiled coil-stabilizing sequence from the protein GCN4. We show that this immunogen, called M2G, elicited cross-reactive antibodies against a number of M types that carry the 3D pattern but not against those that lack the 3D pattern. We further show that the M2G antiserum-recognized M proteins displayed natively on the strep A surface and promoted the opsonophagocytic killing of strep A strains expressing these M proteins. As C4BP binding is a conserved virulence trait of strep A, we propose that targeting the 3D pattern may prove advantageous in vaccine design.


Subject(s)
Antigens, Bacterial , Bacterial Outer Membrane Proteins , Carrier Proteins , Streptococcus pyogenes , Humans , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/chemistry , Carrier Proteins/immunology , Protein Binding , Streptococcus pyogenes/immunology , Cross Reactions
5.
PLoS Pathog ; 18(5): e1010511, 2022 05.
Article in English | MEDLINE | ID: mdl-35605029

ABSTRACT

Hematogenous dissemination is a critical step in the evolution of local infection to systemic disease. The Lyme disease (LD) spirochete, which efficiently disseminates to multiple tissues, has provided a model for this process, in particular for the key early event of pathogen adhesion to the host vasculature. This occurs under shear force mediated by interactions between bacterial adhesins and mammalian cell-surface proteins or extracellular matrix (ECM). Using real-time intravital imaging of the Lyme spirochete in living mice, we previously identified BBK32 as the first LD spirochetal adhesin demonstrated to mediate early vascular adhesion in a living mouse; however, deletion of bbk32 resulted in loss of only about half of the early interactions, suggesting the existence of at least one other adhesin (adhesin-X) that promotes early vascular interactions. VlsE, a surface lipoprotein, was identified long ago by its capacity to undergo rapid antigenic variation, is upregulated in the mammalian host and required for persistent infection in immunocompetent mice. In immunodeficient mice, VlsE shares functional overlap with OspC, a multi-functional protein that displays dermatan sulfate-binding activity and is required for joint invasion and colonization. In this research, using biochemical and genetic approaches as well as intravital imaging, we have identified VlsE as adhesin-X; it is a dermatan sulfate (DS) adhesin that efficiently promotes transient adhesion to the microvasculature under shear force via its DS binding pocket. Intravenous inoculation of mice with a low-passage infectious B. burgdorferi strain lacking both bbk32 and vlsE almost completely eliminated transient microvascular interactions. Comparative analysis of binding parameters of VlsE, BBK32 and OspC provides a possible explanation why these three DS adhesins display different functionality in terms of their ability to promote early microvascular interactions.


Subject(s)
Adhesins, Bacterial , Antigenic Variation , Antigens, Bacterial , Bacterial Proteins , Borrelia burgdorferi , Lipoproteins , Lyme Disease , Microvessels , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Animals , Antigenic Variation/genetics , Antigenic Variation/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Dermatan Sulfate/immunology , Lipoproteins/genetics , Lipoproteins/immunology , Lyme Disease/genetics , Lyme Disease/immunology , Lyme Disease/microbiology , Mammals , Mice , Microvessels/immunology , Microvessels/microbiology , Shear Strength
6.
Scand J Immunol ; 99(4): e13353, 2024 Apr.
Article in English | MEDLINE | ID: mdl-39007994

ABSTRACT

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


Subject(s)
Antibodies, Bacterial , Antigens, Bacterial , Bacterial Outer Membrane Proteins , Borrelia burgdorferi , Lyme Neuroborreliosis , Humans , Lyme Neuroborreliosis/immunology , Lyme Neuroborreliosis/blood , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Middle Aged , Female , Male , Adult , Aged , Borrelia burgdorferi/immunology , Antigens, Bacterial/immunology , Virulence Factors/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Chemokine CXCL13/blood , Chemokine CXCL13/immunology , Bacterial Proteins/immunology , Antibody Formation/immunology
7.
Immunity ; 42(2): 344-355, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680274

ABSTRACT

Bacteria, such as Fusobacterium nucleatum, are present in the tumor microenvironment. However, the immunological consequences of intra-tumoral bacteria remain unclear. Here, we have shown that natural killer (NK) cell killing of various tumors is inhibited in the presence of various F. nucleatum strains. Our data support that this F. nucleatum-mediated inhibition is mediated by human, but not by mouse TIGIT, an inhibitory receptor present on all human NK cells and on various T cells. Using a library of F. nucleatum mutants, we found that the Fap2 protein of F. nucleatum directly interacted with TIGIT, leading to the inhibition of NK cell cytotoxicity. We have further demonstrated that tumor-infiltrating lymphocytes expressed TIGIT and that T cell activities were also inhibited by F. nucleatum via Fap2. Our results identify a bacterium-dependent, tumor-immune evasion mechanism in which tumors exploit the Fap2 protein of F. nucleatum to inhibit immune cell activity via TIGIT.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/microbiology , Colonic Neoplasms/immunology , Colonic Neoplasms/microbiology , Fusobacterium nucleatum/immunology , Receptors, Immunologic/immunology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , Bacterial Outer Membrane Proteins/immunology , Cell Line , Cell Proliferation , Humans , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Protein Binding
8.
Genome ; 67(6): 204-209, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38330385

ABSTRACT

Mycoplasmopsis bovis is a worldwide economically important pathogen of cattle that can cause or indirectly contribute to bovine respiratory disease. M. bovis is also a primary etiological agent of respiratory disease in bison with high mortality rates. A major challenge in the development of an efficacious M. bovis vaccine is the design of antigens that contain both MHC-1 and MHC-2 T-cell epitopes, and that account for population level diversity within the species. Publicly available genomes and sequence read archive libraries of 381 M. bovis strains isolated from cattle (n = 202) and bison (n = 179) in North America were used to identify a core genome of 575 genes, including 38 that encode either known or predicted secreted or outer membrane proteins. The antigenic potentials of the proteins were characterized by the presence and strength of their T-cell epitopes, and their protein variant diversity at the population-level. The proteins had surprisingly low diversity and varying predictive levels of T-cell antigenicity. These results provide a reference for the selection or design of antigens for vaccine testing against strains infecting North American cattle and bison.


Subject(s)
Bison , Animals , Bison/microbiology , Cattle , North America , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Genetic Variation , Mycoplasma bovis/genetics , Mycoplasma bovis/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Genome, Bacterial , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Cattle Diseases/microbiology , Cattle Diseases/immunology
9.
Fish Shellfish Immunol ; 151: 109665, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38830521

ABSTRACT

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


Subject(s)
Aeromonas hydrophila , Bacterial Vaccines , Cyprinidae , Fish Diseases , Gram-Negative Bacterial Infections , Vaccines, Subunit , Aeromonas hydrophila/immunology , Animals , Fish Diseases/prevention & control , Fish Diseases/immunology , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/prevention & control , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Cyprinidae/immunology , Bacterial Outer Membrane Proteins/immunology , Immunity, Humoral
10.
Fish Shellfish Immunol ; 151: 109701, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38878911

ABSTRACT

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


Subject(s)
Aeromonas hydrophila , Bacterial Vaccines , Carps , Fish Diseases , Gram-Negative Bacterial Infections , Animals , Fish Diseases/immunology , Fish Diseases/prevention & control , Carps/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/immunology , Aeromonas hydrophila/immunology , Administration, Oral , Vaccination/veterinary , Bacillus , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics
11.
Nanotechnology ; 35(39)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38917779

ABSTRACT

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


Subject(s)
Brucellosis , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Recombinant Proteins , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Brucellosis/prevention & control , Brucellosis/immunology , Mice , Nanoparticles/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/chemistry , Mice, Inbred BALB C , Female , Brucella Vaccine/immunology , Brucella Vaccine/genetics , Brucella Vaccine/administration & dosage , Brucella abortus/immunology , Brucella abortus/genetics , Drug Carriers/chemistry , Nanovaccines
12.
Appl Microbiol Biotechnol ; 108(1): 424, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037584

ABSTRACT

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


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Leptospirosis , Lipoproteins , Animals , Leptospirosis/prevention & control , Leptospirosis/immunology , Lipoproteins/immunology , Lipoproteins/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cricetinae , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Adjuvants, Immunologic/administration & dosage , Immunoglobulin G/blood , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Leptospira interrogans/immunology , Leptospira interrogans/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Vaccination , Immunity, Humoral , Leptospira/immunology , Leptospira/genetics , Immunogenicity, Vaccine
13.
J Nanobiotechnology ; 22(1): 273, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773507

ABSTRACT

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


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Bacteriophages , Animals , Acinetobacter Infections/prevention & control , Mice , Female , Mice, Inbred BALB C , Bacterial Vaccines/immunology , Immunization , Extracellular Vesicles , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane Proteins/immunology , Disease Models, Animal , Humans , Administration, Intranasal , Viral Proteins
14.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000557

ABSTRACT

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


Subject(s)
Antibodies, Bacterial , Bacterial Outer Membrane Proteins , Escherichia coli , Humans , Escherichia coli/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/immunology , Adult , Female , Staphylococcus aureus/immunology , Male , Antibody Formation/immunology , Middle Aged , Escherichia coli Proteins/immunology , Young Adult , Aged , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Adolescent , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology
15.
World J Microbiol Biotechnol ; 40(8): 250, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38910219

ABSTRACT

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


Subject(s)
Aeromonas hydrophila , Bacterial Outer Membrane Proteins , Bacterial Vaccines , Fish Diseases , Gram-Negative Bacterial Infections , Nanoparticles , Recombinant Proteins , Zebrafish , Animals , Zebrafish/immunology , Aeromonas hydrophila/immunology , Aeromonas hydrophila/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Fish Diseases/prevention & control , Fish Diseases/immunology , Fish Diseases/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Administration, Oral , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Vaccination , Nanovaccines
16.
J Biol Chem ; 298(3): 101715, 2022 03.
Article in English | MEDLINE | ID: mdl-35151691

ABSTRACT

Infection by the bacterium Bordetella pertussis continues to cause considerable morbidity and mortality worldwide. Many current acellular pertussis vaccines include the antigen pertactin, which has presumptive adhesive and immunomodulatory activities, but is rapidly lost from clinical isolates after the introduction of these vaccines. To better understand the contributions of pertactin antibodies to protection and pertactin's role in pathogenesis, we isolated and characterized recombinant antibodies binding four distinct epitopes on pertactin. We demonstrate that four of these antibodies bind epitopes that are conserved across all three classical Bordetella strains, and competition assays further showed that antibodies binding these epitopes are also elicited by B. pertussis infection of baboons. Surprisingly, we found that representative antibodies binding each epitope protected mice against experimental B. pertussis infection. A cocktail of antibodies from each epitope group protected mice against a subsequent lethal dose of B. pertussis and greatly reduced lung colonization levels after sublethal challenge. Each antibody reduced B. pertussis lung colonization levels up to 100-fold when administered individually, which was significantly reduced when antibody effector functions were impaired, with no antibody mediating antibody-dependent complement-induced lysis. These data suggest that antibodies binding multiple pertactin epitopes protect primarily by the same bactericidal mechanism, which overshadows contributions from blockade of other pertactin functions. These antibodies expand the available tools to further dissect pertactin's role in infection and understand the impact of antipertactin antibodies on bacterial fitness.


Subject(s)
Antibodies , Bacterial Outer Membrane Proteins , Bordetella pertussis , Virulence Factors, Bordetella , Whooping Cough , Animals , Antibodies/immunology , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Epitopes , Mice , Pertussis Vaccine/immunology , Virulence Factors, Bordetella/chemistry , Virulence Factors, Bordetella/immunology , Virulence Factors, Bordetella/metabolism , Whooping Cough/prevention & control
17.
Microb Pathog ; 175: 105959, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36581307

ABSTRACT

The growing emergence of resistant bacteria is the current global concern for the humans and animals. Vaccination could be the desirable method to preventing such infectious diseases. Safe and effective vaccines are urgently needed to manage and prevent Salmonella contamination. Subunit vaccines are safe approaches for the protection against Salmonella spp. The bioinformatics methods were performed to determine the gene structure. Gene cassette (rLPSI) was ordered in pET28a (+), and cloned into E.coli BL21 (DE3), and the recombinant protein was expressed using IPTG (1 mM). Mice were immunized by subcutaneous administration of recombinant protein. Then, the mice were challenged by oral administration of 100LD50 of live S. Typhimurium. The Codon adaptation index of the chimeric gene was multiplied by 0.92. Validation results showed that >90% of residues lie in the desired or extra allowed area of the Ramachandran plot. The recombinant protein (65.9 kDa) was expressed in E.coli. Antibody titers in vaccinated mice were significantly different from those in the control groups. Recombinant protein immunization of the mice provided 90% and 70% protection against 10LD50 and 100LD50 of S. Typhimurium, respectively. In general, the results showed the high efficiency of rLPSI chimeric protein as a protective antigen against S. Typhimurium infection. The rLPSI chimeric protein could be an effective recombinant vaccine candidate against S. Typhimurium infection, but more research is needed.


Subject(s)
Escherichia coli Proteins , Salmonella Vaccines , Salmonella typhimurium , Animals , Mice , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Escherichia coli/genetics , Immunization , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Proteins/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Vaccines, Attenuated , Vaccines, Synthetic
18.
J Immunol ; 207(4): 1138-1149, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34341168

ABSTRACT

Group A streptococcal infections are a significant cause of global morbidity and mortality. A leading vaccine candidate is the surface M protein, a major virulence determinant and protective Ag. One obstacle to the development of M protein-based vaccines is the >200 different M types defined by the N-terminal sequences that contain protective epitopes. Despite sequence variability, M proteins share coiled-coil structural motifs that bind host proteins required for virulence. In this study, we exploit this potential Achilles heel of conserved structure to predict cross-reactive M peptides that could serve as broadly protective vaccine Ags. Combining sequences with structural predictions, six heterologous M peptides in a sequence-related cluster were predicted to elicit cross-reactive Abs with the remaining five nonvaccine M types in the cluster. The six-valent vaccine elicited Abs in rabbits that reacted with all 11 M peptides in the cluster and functional opsonic Abs against vaccine and nonvaccine M types in the cluster. We next immunized mice with four sequence-unrelated M peptides predicted to contain different coiled-coil propensities and tested the antisera for cross-reactivity against 41 heterologous M peptides. Based on these results, we developed an improved algorithm to select cross-reactive peptide pairs using additional parameters of coiled-coil length and propensity. The revised algorithm accurately predicted cross-reactive Ab binding, improving the Matthews correlation coefficient from 0.42 to 0.74. These results form the basis for selecting the minimum number of N-terminal M peptides to include in potentially broadly efficacious multivalent vaccines that could impact the overall global burden of group A streptococcal diseases.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Carrier Proteins/immunology , Cross Reactions/immunology , Streptococcal Vaccines/immunology , Animals , Antibodies, Bacterial/immunology , Epitopes/immunology , Female , Humans , Male , Mice , Peptides/immunology , Vaccines, Synthetic/immunology
19.
PLoS Pathog ; 16(12): e1008602, 2020 12.
Article in English | MEDLINE | ID: mdl-33290434

ABSTRACT

There is a pressing need for a gonorrhea vaccine due to the high disease burden associated with gonococcal infections globally and the rapid evolution of antibiotic resistance in Neisseria gonorrhoeae (Ng). Current gonorrhea vaccine research is in the stages of antigen discovery and the identification of protective immune responses, and no vaccine has been tested in clinical trials in over 30 years. Recently, however, it was reported in a retrospective case-control study that vaccination of humans with a serogroup B Neisseria meningitidis (Nm) outer membrane vesicle (OMV) vaccine (MeNZB) was associated with reduced rates of gonorrhea. Here we directly tested the hypothesis that Nm OMVs induce cross-protection against gonorrhea in a well-characterized female mouse model of Ng genital tract infection. We found that immunization with the licensed Nm OMV-based vaccine 4CMenB (Bexsero) significantly accelerated clearance and reduced the Ng bacterial burden compared to administration of alum or PBS. Serum IgG and vaginal IgA and IgG that cross-reacted with Ng OMVs were induced by 4CMenB vaccination by either the subcutaneous or intraperitoneal routes. Antibodies from vaccinated mice recognized several Ng surface proteins, including PilQ, BamA, MtrE, NHBA (known to be recognized by humans), PorB, and Opa. Immune sera from both mice and humans recognized Ng PilQ and several proteins of similar apparent molecular weight, but MtrE was only recognized by mouse serum. Pooled sera from 4CMenB-immunized mice showed a 4-fold increase in serum bactericidal50 titers against the challenge strain; in contrast, no significant difference in bactericidal activity was detected when sera from 4CMenB-immunized and unimmunized subjects were compared. Our findings directly support epidemiological evidence that Nm OMVs confer cross-species protection against gonorrhea, and implicate several Ng surface antigens as potentially protective targets. Additionally, this study further defines the usefulness of murine infection model as a relevant experimental system for gonorrhea vaccine development.


Subject(s)
Cross Protection/immunology , Meningococcal Vaccines/pharmacology , Neisseria gonorrhoeae/immunology , Animals , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Case-Control Studies , Cross Reactions/immunology , Female , Gonorrhea/immunology , Humans , Immune Sera/immunology , Immunization/methods , Male , Meningococcal Infections/microbiology , Meningococcal Vaccines/immunology , Meningococcal Vaccines/metabolism , Mice , Mice, Inbred BALB C , Neisseria meningitidis/immunology , Neisseria meningitidis, Serogroup B/immunology , Retrospective Studies , Serogroup , Vaccination/methods
20.
PLoS Pathog ; 16(5): e1008576, 2020 05.
Article in English | MEDLINE | ID: mdl-32392230

ABSTRACT

Yersinia suppress neutrophil responses by using a type 3 secretion system (T3SS) to inject 6-7 Yersinia effector proteins (Yops) effectors into their cytoplasm. YopH is a tyrosine phosphatase that causes dephosphorylation of the adaptor protein SKAP2, among other targets in neutrophils. SKAP2 functions in reactive oxygen species (ROS) production, phagocytosis, and integrin-mediated migration by neutrophils. Here we identify essential neutrophil functions targeted by YopH, and investigate how the interaction between YopH and SKAP2 influence Yersinia pseudotuberculosis (Yptb) survival in tissues. The growth defect of a ΔyopH mutant was restored in mice defective in the NADPH oxidase complex, demonstrating that YopH is critical for protecting Yptb from ROS during infection. The growth of a ΔyopH mutant was partially restored in Skap2-deficient (Skap2KO) mice compared to wild-type (WT) mice, while induction of neutropenia further enhanced the growth of the ΔyopH mutant in both WT and Skap2KO mice. YopH inhibited both ROS production and degranulation triggered via integrin receptor, G-protein coupled receptor (GPCR), and Fcγ receptor (FcγR) stimulation. SKAP2 was required for integrin receptor and GPCR-mediated ROS production, but dispensable for degranulation under all conditions tested. YopH blocked SKAP2-independent FcγR-stimulated phosphorylation of the proximal signaling proteins Syk, SLP-76, and PLCγ2, and the more distal signaling protein ERK1/2, while only ERK1/2 phosphorylation was dependent on SKAP2 following integrin receptor activation. These findings reveal that YopH prevents activation of both SKAP2-dependent and -independent neutrophilic defenses, uncouple integrin- and GPCR-dependent ROS production from FcγR responses based on their SKAP2 dependency, and show that SKAP2 is not required for degranulation.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Intracellular Signaling Peptides and Proteins/immunology , Neutrophils/immunology , Protein Tyrosine Phosphatases/immunology , Signal Transduction/immunology , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis/immunology , Animals , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/pathology , Reactive Oxygen Species/immunology , Signal Transduction/genetics , Yersinia pseudotuberculosis/pathogenicity , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/pathology
SELECTION OF CITATIONS
SEARCH DETAIL