Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.060
Filter
1.
Nat Commun ; 15(1): 3762, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704378

ABSTRACT

Plants initiate specific defense responses by recognizing conserved epitope peptides within the flagellin proteins derived from bacteria. Proteolytic cleavage of epitope peptides from flagellin by plant apoplastic proteases is thought to be crucial for the perception of the epitope by the plant receptor. However, the identity of the plant proteases involved in this process remains unknown. Here, we establish an efficient identification system for the target proteases in Arabidopsis apoplastic fluid; the method employs native two-dimensional electrophoresis followed by an in-gel proteolytic assay using a fluorescence-quenching peptide substrate. We designed a substrate to specifically detect proteolytic activity at the C-terminus of the flg22 epitope in flagellin and identified two plant subtilases, SBT5.2 and SBT1.7, as specific proteases responsible for the C-terminal cleavage of flg22. In the apoplastic fluid of Arabidopsis mutant plants deficient in these two proteases, we observe a decrease in the C-terminal cleavage of the flg22 domain from flagellin, leading to a decrease in the efficiency of flg22 epitope liberation. Consequently, defensive reactive oxygen species (ROS) production is delayed in sbt5.2 sbt1.7 double-mutant leaf disks compared to wild type following flagellin exposure.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Epitopes , Flagellin , Reactive Oxygen Species , Subtilisins , Arabidopsis/immunology , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/immunology , Epitopes/immunology , Epitopes/metabolism , Flagellin/metabolism , Flagellin/immunology , Mutation , Proteolysis , Reactive Oxygen Species/metabolism , Subtilisins/metabolism , Subtilisins/genetics
2.
Signal Transduct Target Ther ; 9(1): 114, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678055

ABSTRACT

Developing a mucosal vaccine against SARS-CoV-2 is critical for combatting the epidemic. Here, we investigated long-term immune responses and protection against SARS-CoV-2 for the intranasal vaccination of a triple receptor-binding domain (RBD) scaffold protein (3R-NC) adjuvanted with a flagellin protein (KFD) (3R-NC + KFDi.n). In mice, the vaccination elicited RBD-specific broad-neutralizing antibody responses in both serum and mucosal sites sustained at high level over a year. This long-lasting humoral immunity was correlated with the presence of long-lived RBD-specific IgG- and IgA-producing plasma cells, alongside the Th17 and Tfh17-biased T-cell responses driven by the KFD adjuvant. Based upon these preclinical findings, an open labeled clinical trial was conducted in individuals who had been primed with the inactivated SARS-CoV-2 (IAV) vaccine. With a favorable safety profile, the 3R-NC + KFDi.n boost elicited enduring broad-neutralizing IgG in plasma and IgA in salivary secretions. To meet the challenge of frequently emerged variants, we further designed an updated triple-RBD scaffold protein with mutated RBD combinations, which can induce adaptable antibody responses to neutralize the newly emerging variants, including JN.1. Our findings highlight the potential of the KFD-adjuvanted triple-RBD scaffold protein is a promising prototype for the development of a mucosal vaccine against SARS-CoV-2 infection.


Subject(s)
Administration, Intranasal , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Flagellin , SARS-CoV-2 , SARS-CoV-2/immunology , Humans , Flagellin/immunology , Flagellin/genetics , Flagellin/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , Animals , Mice , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Neutralizing/immunology , Female , Antibodies, Viral/immunology , Vaccination , Male , Adult , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Immunoglobulin G/immunology , Immunoglobulin G/blood , Immunoglobulin A/immunology , Middle Aged
3.
Biochemistry (Mosc) ; 89(3): 574-582, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38648774

ABSTRACT

Rabies is a zoonotic disease with high lethality. Most human deaths are associated with the bites received from dogs and cats. Vaccination is the most effective method of preventing rabies disease in both animals and humans. In this study, the ability of an adjuvant based on recombinant Salmonella typhimurium flagellin to increase protective activity of the inactivated rabies vaccine in mice was evaluated. A series of inactivated dry culture vaccine for dogs and cats "Rabikan" (strain Shchelkovo-51) with addition of an adjuvant at various dilutions were used. The control preparation was a similar series of inactivated dry culture vaccine without an adjuvant. Protective activity of the vaccine preparations was evaluated by the NIH potency test, which is the most widely used and internationally recommended method for testing effectiveness of the inactivated rabies vaccines. The value of specific activity of the tested rabies vaccine when co-administered with the adjuvant was significantly higher (48.69 IU/ml) than that of the vaccine without the adjuvant (3.75 IU/ml). Thus, recombinant flagellin could be considered as an effective adjuvant in the composition of future vaccine preparations against rabies virus.


Subject(s)
Adjuvants, Immunologic , Flagellin , Rabies Vaccines , Rabies , Vaccines, Inactivated , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Animals , Flagellin/immunology , Mice , Rabies/prevention & control , Rabies/immunology , Vaccines, Inactivated/immunology , Dogs , Rabies virus/immunology , Salmonella typhimurium/immunology , Female , Cats
4.
Infect Immun ; 92(5): e0006024, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38619302

ABSTRACT

Melioidosis is an emerging tropical infection caused by inhalation, inoculation, or ingestion of the flagellated, facultatively intracellular pathogen Burkholderia pseudomallei. The melioidosis case fatality rate is often high, and pneumonia, the most common presentation, doubles the risk of death. The alveolar macrophage is a sentinel pulmonary host defense cell, but the human alveolar macrophage in B. pseudomallei infection has never been studied. The objective of this study was to investigate the host-pathogen interaction of B. pseudomallei infection with the human alveolar macrophage and to determine the role of flagellin in modulating inflammasome-mediated pathways. We found that B. pseudomallei infects primary human alveolar macrophages but is gradually restricted in the setting of concurrent cell death. Electron microscopy revealed cytosolic bacteria undergoing division, indicating that B. pseudomallei likely escapes the alveolar macrophage phagosome and may replicate in the cytosol, where it triggers immune responses. In paired human blood monocytes, uptake and intracellular restriction of B. pseudomallei are similar to those observed in alveolar macrophages, but cell death is reduced. The alveolar macrophage cytokine response to B. pseudomallei is characterized by marked interleukin (IL)-18 secretion compared to monocytes. Both cytotoxicity and IL-18 secretion in alveolar macrophages are partially flagellin dependent. However, the proportion of IL-18 release that is driven by flagellin is greater in alveolar macrophages than in monocytes. These findings suggest differential flagellin-mediated inflammasome pathway activation in the human alveolar macrophage response to B. pseudomallei infection and expand our understanding of intracellular pathogen recognition by this unique innate immune lung cell.


Subject(s)
Burkholderia pseudomallei , Flagellin , Host-Pathogen Interactions , Inflammasomes , Macrophages, Alveolar , Humans , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Inflammasomes/immunology , Inflammasomes/metabolism , Burkholderia pseudomallei/immunology , Flagellin/immunology , Flagellin/metabolism , Host-Pathogen Interactions/immunology , Melioidosis/immunology , Melioidosis/microbiology , Cells, Cultured
5.
Nature ; 625(7996): 750-759, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200311

ABSTRACT

Iron is critical during host-microorganism interactions1-4. Restriction of available iron by the host during infection is an important defence strategy, described as nutritional immunity5. However, this poses a conundrum for externally facing, absorptive tissues such as the gut epithelium or the plant root epidermis that generate environments that favour iron bioavailability. For example, plant roots acquire iron mostly from the soil and, when iron deficient, increase iron availability through mechanisms that include rhizosphere acidification and secretion of iron chelators6-9. Yet, the elevated iron bioavailability would also be beneficial for the growth of bacteria that threaten plant health. Here we report that microorganism-associated molecular patterns such as flagellin lead to suppression of root iron acquisition through a localized degradation of the systemic iron-deficiency signalling peptide Iron Man 1 (IMA1) in Arabidopsis thaliana. This response is also elicited when bacteria enter root tissues, but not when they dwell on the outer root surface. IMA1 itself has a role in modulating immunity in root and shoot, affecting the levels of root colonization and the resistance to a bacterial foliar pathogen. Our findings reveal an adaptive molecular mechanism of nutritional immunity that affects iron bioavailability and uptake, as well as immune responses.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Bacteria , Intracellular Signaling Peptides and Proteins , Iron , Pathogen-Associated Molecular Pattern Molecules , Plant Roots , Arabidopsis/immunology , Arabidopsis/metabolism , Arabidopsis/microbiology , Arabidopsis Proteins/metabolism , Bacteria/immunology , Bacteria/metabolism , Flagellin/immunology , Gene Expression Regulation, Plant , Intracellular Signaling Peptides and Proteins/metabolism , Iron/metabolism , Plant Immunity , Plant Roots/immunology , Plant Roots/metabolism , Plant Roots/microbiology , Plant Shoots/immunology , Plant Shoots/metabolism , Plant Shoots/microbiology , Rhizosphere , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism
6.
Science ; 381(6653): 37-38, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37410824
7.
Int J Mol Sci ; 23(24)2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36555214

ABSTRACT

The nasal-associated lymphoid tissues (NALT) are generally accepted as an immune induction site, but the activation of naïve T-cells in that compartment has not been well-characterized. I wanted to determine if early events in naïve CD4+ T cell activation and the extent of antigen specific cell division are similar in NALT to that observed in other secondary lymphoid compartments. I performed antigen tracking experiments and analyzed the activation of naïve antigen-specific CD4+ T cells in the nasal-associated lymphoid tissues (NALT). I directly observed transepithelial transport of fluorescently labeled antigen from the lumen of the airway to the interior of the NALT two hours following immunization. One day following intranasal (i.n.) immunization with antigen and adjuvant, antigen-specific CD4+ T cells in the NALT associated as clusters, while antigen-specific CD4+ T cells in control mice immunized with adjuvant only remained dispersed. The antigen-specific CD4+ populations in the NALT and cranial deep cervical lymph nodes of immunized mice expanded significantly by day three following immunization. These findings are consistent with initial activation of naïve CD4+ T cells in the NALT and offer insight into adjuvant mechanism of flagellin in the upper respiratory compartment.


Subject(s)
Flagellin , Lymphoid Tissue , Vaccines, Subunit , Animals , Mice , Adjuvants, Immunologic , Administration, Intranasal , CD4-Positive T-Lymphocytes , Flagellin/immunology , Immunization , Mice, Inbred BALB C , Nasal Mucosa , T-Lymphocytes , Vaccines, Subunit/immunology
8.
Sci Adv ; 8(38): eabq2422, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36149952

ABSTRACT

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating disease with an unclear etiology and pathogenesis. Both an involvement of the immune system and gut microbiota dysbiosis have been implicated in its pathophysiology. However, potential interactions between adaptive immune responses and the microbiota in ME/CFS have been incompletely characterized. Here, we profiled antibody responses of patients with severe ME/CFS and healthy controls against microbiota and viral antigens represented as a phage-displayed 244,000 variant library. Patients with severe ME/CFS exhibited distinct serum antibody epitope repertoires against flagellins of Lachnospiraceae bacteria. Training machine learning algorithms on this antibody-binding data demonstrated that immune responses against gut microbiota represent a unique layer of information beyond standard blood tests, providing improved molecular diagnostics for ME/CFS. Together, our results point toward an involvement of the microbiota-immune axis in ME/CFS and lay the foundation for comparative studies with inflammatory bowel diseases and illnesses characterized by long-term fatigue symptoms, including post-COVID-19 syndrome.


Subject(s)
Antibody Formation , Fatigue Syndrome, Chronic , Flagellin , Gastrointestinal Microbiome , Epitopes , Fatigue Syndrome, Chronic/diagnosis , Fatigue Syndrome, Chronic/immunology , Flagellin/immunology , Humans
9.
PLoS Pathog ; 18(1): e1010253, 2022 01.
Article in English | MEDLINE | ID: mdl-35073369

ABSTRACT

Flagellin is a key bacterial virulence factor that can stimulate molecular immune signaling in both animals and plants. The detailed mechanisms of recognizing flagellin and mounting an efficient immune response have been uncovered in vertebrates; however, whether invertebrates can discriminate flagellin remains largely unknown. In the present study, the homolog of human SHOC2 leucine rich repeat scaffold protein in kuruma shrimp (Marsupenaeus japonicus), designated MjShoc2, was found to interact with Vibrio anguillarum flagellin A (FlaA) using yeast two-hybrid and pull-down assays. MjShoc2 plays a role in antibacterial response by mediating the FlaA-induced expression of certain antibacterial effectors, including lectin and antimicrobial peptide. FlaA challenge, via MjShoc2, led to phosphorylation of extracellular regulated kinase (Erk), and the subsequent activation of signal transducer and activator of transcription (Stat), ultimately inducing the expression of effectors. Therefore, by establishing the FlaA/MjShoc2/Erk/Stat signaling axis, this study revealed a new antibacterial strategy in shrimp, and provides insights into the flagellin sensing mechanism in invertebrates.


Subject(s)
Arthropod Proteins/immunology , Flagellin/immunology , Intracellular Signaling Peptides and Proteins/immunology , Penaeidae/immunology , Vibrio Infections/immunology , Animals , MAP Kinase Signaling System/immunology , Penaeidae/microbiology , STAT Transcription Factors/immunology , Vibrio
10.
Sci Rep ; 12(1): 53, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997041

ABSTRACT

Zika virus (ZIKV) is an arbovirus from the Flaviviridae family and Flavivirus genus. Neurological events have been associated with ZIKV-infected individuals, such as Guillain-Barré syndrome, an autoimmune acute neuropathy that causes nerve demyelination and can induce paralysis. With the increase of ZIKV infection incidence in 2015, malformation and microcephaly cases in newborns have grown considerably, which suggested congenital transmission. Therefore, the development of an effective vaccine against ZIKV became an urgent need. Live attenuated vaccines present some theoretical risks for administration in pregnant women. Thus, we developed an in silico multiepitope vaccine against ZIKV. All structural and non-structural proteins were investigated using immunoinformatics tools designed for the prediction of CD4 + and CD8 + T cell epitopes. We selected 13 CD8 + and 12 CD4 + T cell epitopes considering parameters such as binding affinity to HLA class I and II molecules, promiscuity based on the number of different HLA alleles that bind to the epitopes, and immunogenicity. ZIKV Envelope protein domain III (EDIII) was added to the vaccine construct, creating a hybrid protein domain-multiepitope vaccine. Three high scoring continuous and two discontinuous B cell epitopes were found in EDIII. Aiming to increase the candidate vaccine antigenicity even further, we tested secondary and tertiary structures and physicochemical parameters of the vaccine conjugated to four different protein adjuvants: flagellin, 50S ribosomal protein L7/L12, heparin-binding hemagglutinin, or RS09 synthetic peptide. The addition of the flagellin adjuvant increased the vaccine's predicted antigenicity. In silico predictions revealed that the protein is a probable antigen, non-allergenic and predicted to be stable. The vaccine's average population coverage is estimated to be 87.86%, which indicates it can be administered worldwide. Peripheral Blood Mononuclear Cells (PBMC) of individuals with previous ZIKV infection were tested for cytokine production in response to the pool of CD4 and CD8 ZIKV peptide selected. CD4 + and CD8 + T cells showed significant production of IFN-γ upon stimulation and IL-2 production was also detected by CD8 + T cells, which indicated the potential of our peptides to be recognized by specific T cells and induce immune response. In conclusion, we developed an in silico universal vaccine predicted to induce broad and high-coverage cellular and humoral immune responses against ZIKV, which can be a good candidate for posterior in vivo validation.


Subject(s)
Computational Biology/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Viral Proteins/immunology , Viral Vaccines/chemistry , Viral Vaccines/immunology , Zika Virus/immunology , Adjuvants, Immunologic , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Epitopes, B-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/chemistry , Flagellin/immunology , Humans , Immunity, Humoral , Immunogenicity, Vaccine , Lectins/immunology , Leukocytes, Mononuclear/immunology , Peptides/immunology , Phylogeny , Ribosomal Proteins/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Viral Proteins/chemistry , Zika Virus/chemistry , Zika Virus Infection/immunology , Zika Virus Infection/virology
11.
Fish Shellfish Immunol ; 120: 658-673, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34500055

ABSTRACT

The purpose of the current study was to explore the immunomodulatory effects of different adjuvants combined with inactivated vaccines under Aeromonas veronii TH0426 infection in crucian carp. This study explored the best conditions for A. veronii as an inactivated vaccine, and included an animal safety test. Furthermore, we expressed the flagellin FlaA of the A. veronii TH0426 strain for use as an adjuvant supplemented in the diet. Crucian carp were fed 12 different experimental diets for 35 days, including the administration of 10 different adjuvants and inactivated vaccine combinations (50% aluminum hydroxide gel and inactivated vaccine combination, and inactivated vaccine with 20%, 30%, or 50% glucan, astragalus polysaccharide or flagellin), inactivated vaccine alone, and PBS control without adjuvant and inactivated vaccine. After the 42 day feeding trials, the fish were challenged with A. veronii TH0426, and the survival rate over 14 days was recorded. In addition, flagellin FlaA can be expressed normally in large amounts. All experimental groups produced higher levels of IgM serum titres than the control group in the different feeding cycles. Moreover, the activity of serum ACP, AKP, SOD, and LZM, and the expression of inflammatory factors were significantly increased in the experimental groups compared with the control group. The results of qRT-PCR analysis showed that the transcription levels of the IL-10, IL-1ß, IFN-γ and TNF-α genes in heart, liver, spleen and kidney tissues were significantly enhanced by adjuvant treatment, indicating that the addition of adjuvants can significantly promote the body's inflammatory response. In addition, the phagocytic activity of leukocytes in each adjuvant treated group was significantly enhanced compared to that in the groups without adjuvant. After the A. veronii challenge, the survival rate of all adjuvant-treated groups was significantly higher than that of the control group, and the 50% flagellin adjuvant group had the highest rate of 78.37%. Overall, our findings strongly indicate that adjuvants not only significantly improve the body's immunity, but also exhibit a strong anti-infection ability. Importantly, this work provides a new perspective for the prevention and control of aquaculture diseases.


Subject(s)
Adjuvants, Immunologic , Bacterial Vaccines/immunology , Carps/immunology , Fish Diseases , Gram-Negative Bacterial Infections , Adjuvants, Immunologic/pharmacology , Aeromonas veronii/immunology , Animals , Disease Resistance , Fish Diseases/prevention & control , Flagellin/immunology , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/veterinary , Vaccines, Inactivated
12.
Fish Shellfish Immunol ; 120: 102-110, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34737057

ABSTRACT

Vibrio parahaemolyticus is an important marine pathogen that cause inflammation even death in teleost. It has brought huge economic losses to aquaculture and serious threats to the sustainable development of marine fisheries. Here, we isolated the DNA, RNA, and total flagellin from V. parahaemolyticus, and obtained the primary spleen and head kidney cells (including leukocytes) from Tetraodon nigroviridis. V. parahaemolyticus DNA, RNA, and total flagellin were used to treat the T. nigroviridis primary cells described above. The results show that the nitric oxide (NO) production and respiratory burst response were significantly induced after stimulation with V. parahaemolyticus total flagellin in T. nigroviridis head kidney and spleen cells. And total flagellin could promote the gene expression and protein production of IL-1ß in T. nigroviridis leukocytes. T. nigroviridis TLR5M (TnTLR5M) and TLR5S (TnTLR5S) ORF sequences were obtained as the main recognition receptor for flagellin. Real-time fluorescent quantitative PCR (qRT-PCR) was performed to detect the expression of pattern recognition receptor TnTLR5M and TnTLR5S, the important signal molecule of inflammatory system TnMyD88 and TnTRAF6, and inflammatory cytokines TnIL-1ß and TnIFN-γ2. The results show that there were a significant upregulation after challenge with V. parahaemolyticus total flagellin. We further demonstrated that the total flagellin of V. parahaemolyticus could activate the luciferase activity of the NF-κB reporter gene mediated by TnTLR5M. Overall, our results suggest that V. parahaemolyticus total flagellin activated the NO production, respiratory burst response, and inflammatory cytokines expressions, such as TnIL-1ß and TnIFN-γ2, through the TnTLR5M-NF-κB signaling pathway in T. nigroviridis.


Subject(s)
Flagellin , Tetraodontiformes , Vibrio parahaemolyticus , Animals , Cytokines/immunology , Fish Proteins/genetics , Flagellin/immunology , Interferon-gamma/immunology , Interleukin-1beta/immunology , NF-kappa B/genetics , Tetraodontiformes/immunology , Tetraodontiformes/microbiology , Toll-Like Receptor 5/genetics , Vibrio parahaemolyticus/immunology
13.
J Immunol Methods ; 500: 113172, 2022 01.
Article in English | MEDLINE | ID: mdl-34673003

ABSTRACT

A promising strategy for controlling animal brucellosis is vaccination with commercial vaccine strains (Brucella melitensis Rev.1 and Brucella abortus RB51). Owing to safety concerns associated with these vaccines, developing a more effective and safe vaccine is essential. In this study, we examined the capacity of BhuA, 7α-HSDH or FliC antigens in the presence or absence of adjuvant in eliciting immune responses against brucellosis. After cloning, expression and purification, these proteins were used to examine immunologic responses. All immunized mice induced a vigorous IgG, with a predominant IgG2a response. Moreover, splenocytes of immunized mice proliferated and produced IL-2 and IFN-γ, suggesting the induction of cellular immunity. The high IgG2a/IgG1 ratio and IL-2 and IFN-γ indicated a Th1-oriented immune response in test groups. BhuA-, 7α-HSDH- or FliC- poly I:C formulations were the most effective at inducing Th1 immune response compared to groups immunized with naked proteins. Immunization with proteins protected mice against B. melitensis 16M and B. abortus 544. The proteins in adjuvant induced higher levels of protection than proteins only and exhibited similar degree of protection to live attenuated vaccines. Our results, for first time, introduced five potential candidates for subunit vaccine development against B. melitensis and B. abortus infection.


Subject(s)
Bacterial Proteins/immunology , Brucella Vaccine/immunology , Brucella abortus/physiology , Brucella melitensis/physiology , Brucellosis, Bovine/immunology , Flagellin/immunology , Hydroxysteroid Dehydrogenases/immunology , Membrane Transport Proteins/immunology , Th1 Cells/immunology , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/blood , Cattle , Disease Models, Animal , Female , Immunity, Humoral , Immunoglobulin G/blood , Interferon-gamma/metabolism , Mice , Poly I-C/immunology , Vaccines, Subunit
14.
Cell Rep ; 37(11): 110112, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34863353

ABSTRACT

An ideal vaccine against SARS-CoV-2 is expected to elicit broad immunity to prevent viral infection and disease, with efficient viral clearance in the upper respiratory tract (URT). Here, the N protein and prefusion-full S protein (SFLmut) are combined with flagellin (KF) and cyclic GMP-AMP (cGAMP) to generate a candidate vaccine, and this vaccine elicits stronger systemic and mucosal humoral immunity than vaccines containing other forms of the S protein. Furthermore, the candidate vaccine administered via intranasal route can enhance local immune responses in the respiratory tract. Importantly, human ACE2 transgenic mice given the candidate vaccine are protected against lethal SARS-CoV-2 challenge, with superior protection in the URT compared with that in mice immunized with an inactivated vaccine. In summary, the developed vaccine can elicit a multifaceted immune response and induce robust viral clearance in the URT, which makes it a potential vaccine for preventing disease and infection of SARS-CoV-2.


Subject(s)
COVID-19 Vaccines/immunology , Combined Modality Therapy/methods , SARS-CoV-2/immunology , Adjuvants, Vaccine , Administration, Intranasal , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Viral/immunology , Antigens/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/genetics , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/immunology , Female , Flagellin/immunology , HEK293 Cells , Humans , Immunity/immunology , Immunity/physiology , Immunity, Humoral/immunology , Immunization , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleotides, Cyclic/immunology , Phosphoproteins/immunology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Vero Cells
15.
Front Immunol ; 12: 774233, 2021.
Article in English | MEDLINE | ID: mdl-34912344

ABSTRACT

Vibrio anguillarum, an opportunistic pathogen of aquatic animals, moves using a filament comprised of polymerised flagellin proteins. Flagellins are essential virulence factors for V. anguillarum infection. Herein, we investigated the effects of flagellins (flaA, flaB, flaC, flaD and flaE) on cell apoptosis, TLR5 expression, and production of IL-8 and TNF-α. FlaB exhibited the strongest immunostimulation effects. To explore the functions of flaB in infection, we constructed a flaB deletion mutant using a two-step recombination method, and in vitro experiments showed a significant decrease in the expression of TLR5 and inflammatory cytokines compared with wild-type cells. However in the in vivo study, expression of inflammatory cytokines and intestinal mucosal structure showed no significant differences between groups. Additionally, flaB induced a significant increase in TLR5 expression based on microscopy analysis of fluorescently labelled TLR5, indicating interactions between the two proteins, which was confirmed by native PAGE and yeast two-hybrid assay. Molecular simulation of interactions between flaB and TLR5 was performed to identify the residues involved in binding, revealing two binding sites. Then, based on molecular dynamics simulations, we carried out thirteen site-directed mutations occurring at the amino acid sites of Q57, N83, N87, R91, D94, E122, D152, N312, R313, N320, L97, H316, I324 in binding regions of flaB protein by TLR5, respectively. Surface plasmon resonance (SPR) was employed to compare the affinities of flaB mutants for TLR5, and D152, D94, I324, N87, R313, N320 and H316 were found to mediate interactions between flaB and TLR5. Our comprehensive and systematic analysis of V. anguillarum flagellins establishes the groundwork for future design of flagellin-based vaccines.


Subject(s)
Flagellin/chemistry , Flagellin/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Vibrio Infections/veterinary , Vibrio/immunology , Animals , Apoptosis , Disease Susceptibility , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/metabolism , Fish Diseases/microbiology , Flagellin/genetics , Host-Pathogen Interactions/immunology , Immunophenotyping , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Models, Molecular , Mutation , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Structure-Activity Relationship , Vibrio/pathogenicity , Virulence , Virulence Factors
16.
Cells ; 10(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34685593

ABSTRACT

TLR5 ligand flagellin-containing fusion proteins are potential vaccine candidates for many diseases. A recombinant fusion protein of flagellin A and the major birch pollen allergen Bet v 1 (rFlaA:Betv1) modulates immune responses in vitro and in vivo. We studied the effects of rFlaA:Betv1 on bone marrow-derived macrophages (BMDMs). BMDMs differentiated from BALB/c, C57BL/6, TLR5-/-, or MyD88-/- mice were pre-treated with inhibitors, stimulated with rFlaA:Betv1 or respective controls, and analyzed for activation, cytokine secretion, metabolic state, RNA transcriptome, and modulation of allergen-specific Th2 responses. Stimulation of BMDMs with rFlaA:Betv1 resulted in MyD88-dependent production of IL-1ß, IL-6, TNF-α, IL-10, CD69 upregulation, and a pronounced shift towards glycolysis paralleled by activation of MAPK, NFκB, and mTOR signaling. Inhibition of either mTOR (rapamycin) or SAP/JNK-MAPK signaling (SP600125) resulted in dose-dependent metabolic suppression. In BMDM and T cell co-cultures, rFlaA:Betv1 stimulation suppressed rBet v 1-induced IL-5 and IL-13 secretion while inducing IFN-γ production. mRNA-Seq analyses showed HIF-1a, JAK, STAT, phagosome, NLR, NFκB, TNF, TLR, and chemokine signaling to participate in the interplay of cell activation, glycolysis, and immune response. rFlaA:Betv1 strongly activated BMDMs, resulting in MyD88-, MAPK-, and mTOR-dependent enhancement of glucose metabolism. Our results suggest macrophages are important target cells to consider during restauration of allergen tolerance during AIT.


Subject(s)
Allergens/immunology , Antigens, Bacterial/immunology , Antigens, Plant/immunology , Flagellin/immunology , Recombinant Fusion Proteins/immunology , Animals , Bacterial Proteins/immunology , Cells, Cultured , Glucose/metabolism , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Plant Proteins/immunology , Pollen/immunology
17.
Mol Immunol ; 136: 118-127, 2021 08.
Article in English | MEDLINE | ID: mdl-34130152

ABSTRACT

Pseudomonas aeruginosa (PA) is one of the most dominant causes of nosocomial infections in burn patients. Increasing emergence of antibiotic-resistant strains highlights the need for novel antimicrobial agents. Flagellin, the main component protein of flagellum, is determined as the major antigen interacting with anti-P. aeruginosa IgY antibodies. The current study was aimed to evaluate the antibacterial potency of IgY antibodies raised against recombinant type A, and B flagellins. The immunogenicity and specificity of IgY antibodies were confirmed through indirect ELISA and western blot analysis, respectively. Anti-flagellin IgYs reduced the motility, biofilm formation and invasion potency of both strains. The cell surface hydrophobicity (CSH) of bacteria was increased upon IgY treatment, and in vitro opsonophagocytosis assay confirmed the high protective potency of specific antibodies via polymorphonuclear leukocyte (PMN)-augmented bacterial cell killing. The protective efficacy of IgYs was also studied in both acute pneumonia and burn wound murine models. Anti-flagellin B-IgY induced 100 % and 40 % protection against laboratory, and hospital strains in burn wound model, respectively. Protection in acute pneumonia against all strains was 100 %. Anti-flagellin A-IgY failed to protect mice in burn wound model, but provided 100 % protection against all strains in acute pneumonia challenge. In vitro, ex vivo and in vivo experiments confirmed the dose-dependent and non-type specific essence of anti-flagellin IgY antibodies, providing the benefit of covering all strain types in a dose dependent manner. Our findings provide evidence that anti-flagellin IgY antibodies qualify as novel economical therapeutic option against PA infection.


Subject(s)
Burns/microbiology , Flagellin/immunology , Immunoglobulins/immunology , Pneumonia/microbiology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Animals , Biofilms/growth & development , Burns/immunology , Chickens , Cross Infection/microbiology , Disease Models, Animal , Immunization, Passive , Male , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Pneumonia/immunology , Pseudomonas Infections/immunology
18.
Am J Respir Cell Mol Biol ; 65(4): 378-389, 2021 10.
Article in English | MEDLINE | ID: mdl-34102087

ABSTRACT

Excessive lung inflammation and airway epithelial damage are hallmarks of human inflammatory lung diseases, such as cystic fibrosis (CF). Enhancement of innate immunity provides protection against pathogens while reducing lung-damaging inflammation. However, the mechanisms underlying innate immunity-mediated protection in the lung remain mysterious, in part because of the lack of appropriate animal models for these human diseases. TLR5 (Toll-like receptor 5) stimulation by its specific ligand, the bacterial protein flagellin, has been proposed to enhance protection against several respiratory infectious diseases, although other cellular events, such as calcium signaling, may also control the intensity of the innate immune response. Here, we investigated the molecular events prompted by stimulation with flagellin and its role in regulating innate immunity in the lung of the pig, which is anatomically and genetically more similar to humans than rodent models. We found that flagellin treatment modulated NF-κB signaling and intracellular calcium homeostasis in airway epithelial cells. Flagellin pretreatment reduced the NF-κB nuclear translocation and the expression of proinflammatory cytokines to a second flagellin stimulus as well as to Pseudomonas aeruginosa infection. Moreover, in vivo administration of flagellin decreased the severity of P. aeruginosa-induced pneumonia. Then we confirmed these beneficial effects of flagellin in a pathological model of CF by using ex vivo precision-cut lung slices from a CF pigz model. These results provide evidence that flagellin treatment contributes to a better regulation of the inflammatory response in inflammatory lung diseases such as CF.


Subject(s)
Flagellin/pharmacology , Inflammation/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity , Animals , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Flagellin/immunology , Flagellin/metabolism , Immunity, Innate/drug effects , Lung/immunology , Lung/microbiology , Lung/pathology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Signal Transduction/drug effects , Swine
19.
Sci Rep ; 11(1): 9940, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976336

ABSTRACT

Clostridioides difficile (C. difficile) is an opportunistic anaerobic bacterium that causes severe diseases of the digestive tract of humans and animals. One of the possible methods of preventing C. difficile infection is to develop a vaccine. The most promising candidates for vaccine antigens are the proteins involved in the adhesion phenomena. Among them, the FliC and FliD are considered to be suitable candidates. In this paper, the FliC and FliD protein polypeptide epitopes were mapped in silico and by using PEPSCAN procedure. We identified four promising epitopes: 117QRMRTLS123, 205MSKAG209 of FliC and 226NKVAS230, 306TTKKPKD312 of FliD protein. We showed that 117QRMRTLS123 sequence is not only located in TLR5-binding and activating region, as previously shown, but forms an epitope recognized by C. difficile-infected patients' antibodies. 205MSKAG209 is a C. difficile-unique, immunogenic sequence that forms an exposed epitope on the polymerized flagella structure which makes it a suitable vaccine antigen. 226NKVAS230 and 306TTKKPKD312 are well exposed and possess potential protective properties according to VaxiJen analysis. Our results open the possibility to use these epitopes as suitable anti-C. difficile vaccine antigens.


Subject(s)
Clostridioides difficile/immunology , Clostridium Infections/diagnosis , Flagella/immunology , Amino Acid Sequence/genetics , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Clostridioides/genetics , Clostridioides/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/immunology , Epitopes/immunology , Flagellin/immunology , Humans , Rabbits , Sequence Alignment/methods
20.
Front Immunol ; 12: 641563, 2021.
Article in English | MEDLINE | ID: mdl-33841424

ABSTRACT

At present, the central role played by arginine in the modulation of the inflammatory cellular responses is well-recognized, and many pro-inflammatory stimuli are known to modulate the expression and activity of its transmembrane transporters. In this regard, we have addressed the effects of bacterial flagellin from Pseudomonas aeruginosa (FLA-PA) on the uptake of the amino acid in human epithelial respiratory cells. Among the arginine transporters, only ATB0,+, y+L, and y+ were operative in bronchial epithelial Calu-3 cells under control conditions; however, only the expression and activity of ATB0,+ were stimulated upon incubation with flagellin, whereas those of systems y+L and y+ were not stimulated. As a result, this induction, in turn, led to an increase in the intracellular content of arginine without making any change to its metabolic pathway. In addition, flagellin upregulated the amount of other amino acids substrates of ATB0,+, in particular, all the essential amino acids, such as valine, isoleucine, and leucine, along with the non-essential glutamine. At the molecular level, these effects were directly referable to the stimulation of a toll-like receptor-5 (TLR5) signaling pathway and to the induction of nuclear factor-κB (NF-κB) transcription factor. An induction of ATB0,+ expression has been observed also in EpiAirway™, a model of primary human normal tracheal-bronchial epithelial cells that mimics the in vitro pseudostratified columnar epithelium of the airways. In this tissue model, the incubation with flagellin is associated with the upregulation of messenger RNAs (mRNAs) for the chemokine IL-8 and for the cytokines IL-6 and interleukin-1ß (IL-1ß); as for the latter, a marked secretion in the extracellular medium was also observed due to the concomitant activation of caspase-1. The overall findings indicate that, in human respiratory epithelium, flagellin promotes cellular responses associating the increase of intracellular amino acids through ATB0,+ with the activation of the inflammasome. Given the role of the ATB0,+ transporter as a delivery system for bronchodilators in human airway epithelial cells, its induction under inflammatory conditions gains particular relevance in the field of respiratory pharmacology.


Subject(s)
Amino Acid Transport System ASC/immunology , Arginine/metabolism , Flagellin/immunology , Minor Histocompatibility Antigens/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Amino Acid Transport System ASC/metabolism , Amino Acids, Neutral/metabolism , Antigens, Bacterial/immunology , Cells, Cultured , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Minor Histocompatibility Antigens/metabolism , Pseudomonas aeruginosa
SELECTION OF CITATIONS
SEARCH DETAIL
...