Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
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
2.
J Pharm Sci ; 113(7): 1794-1803, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38522753

ABSTRACT

Research on innovative mucosal adjuvants is essential to develop new vaccines for safe mucosal application. In this work, we propose the development of a Lactococcus lactis that expresses a variant of flagellin on its surface (FliC131*), to increase the adjuvanticity of the living cell and cell wall-derived particles (CWDP). We optimized the expression of FliC131*, and confirmed its identity and localization by Western blot and flow cytometry. We also generated CWDP containing FliC131* (CDWP-FliC131*) and evaluated their storage stability. Lastly, we measured the human TLR5 stimulating activity in vitro and assessed the adjuvanticity in vivo using ovalbumin (OVA) as a model antigen. As a result, we generated L. lactis/pCWA-FliC131*, that expresses and displays FliC131* on its surface, obtained the corresponding CWDP-FliC131*, and showed that both activated hTLR5 in vitro in a dose-dependent manner. Furthermore, CWDP-FliC131* retained this biological activity after being lyophilized and stored for a year. Finally, intranasal immunization of mice with OVA plus live L. lactis/pCWA-FliC131* or CWDP-FliC131* induced OVA-specific IgG and IgA in serum, intestinal lavages, and bronchoalveolar lavages. Our work demonstrates the potential of this recombinant L. lactis with an enhanced adjuvant effect, prompting its further evaluation for the design of novel mucosal vaccines.


Subject(s)
Adjuvants, Immunologic , Flagellin , Lactococcus lactis , Mice, Inbred BALB C , Ovalbumin , Toll-Like Receptor 5 , Lactococcus lactis/immunology , Animals , Flagellin/immunology , Flagellin/administration & dosage , Mice , Humans , Ovalbumin/immunology , Ovalbumin/administration & dosage , Toll-Like Receptor 5/immunology , Adjuvants, Immunologic/administration & dosage , Female , Immunity, Mucosal/drug effects , Immunity, Mucosal/immunology , Immunization/methods , Administration, Intranasal
3.
ACS Appl Bio Mater ; 4(6): 4953-4961, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34179728

ABSTRACT

We generated self-adjuvanted protein nanoparticles of conserved influenza antigens and immunized mice via skin vaccination with dissolvable microneedle patches (MNPs) to increase the strength and breadth of immune responses. We produced M2e nanoparticles via ethanol desolvation, and double-layered NA1/M2e (shell/core), NA1-FliC/M2e, NA2/M2e, and NA2-FliC/M2e protein nanoparticles by chemically crosslinking influenza NA and flagellin (FliC) onto the surfaces of the M2e nanoparticles. The resulting nanoparticles retained FliC TLR5 innate signaling activity and significantly increased antigen-uptake and dendritic cell maturation in vitro. We incorporated the nanoparticles into MNPs for skin vaccination in mice. The nanoparticle MNPs significantly increased M2e and NA-specific antibody levels, the numbers of germinal center B cells, and IL-4 positive splenocytes. Double-layered nanoparticle MNP skin vaccination protected mice against homologous and heterosubtypic influenza viruses. Our results demonstrated that MNP skin vaccination of NA-FliC/M2e nanoparticles could be developed into a standalone or synergistic component of a universal influenza vaccine strategy.


Subject(s)
Drug Delivery Systems , Flagellin/administration & dosage , Influenza Vaccines/administration & dosage , Nanoparticles/administration & dosage , Neuraminidase/administration & dosage , Orthomyxoviridae Infections/prevention & control , Vaccination/methods , Viral Matrix Proteins/administration & dosage , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cytokines/immunology , Dendritic Cells/immunology , Flagellin/chemistry , Immunoglobulin G/blood , Influenza Vaccines/chemistry , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Microinjections , Nanoparticles/chemistry , Needles , Neuraminidase/chemistry , Neuraminidase/immunology , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/immunology
4.
Toxicol Lett ; 342: 58-72, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33571619

ABSTRACT

Acetaminophen (APAP) poisoning is the most common cause of drug-induced acute liver injury (ALI). Our results showed that toll-like receptor 5 (TLR5) was abundantly expressed in hepatocytes and dramatically downregulated in the toxic mouse livers. Hence, we herein investigated the role of TLR5 signaling after APAP overdose. Mice were intraperitoneally (i.p.) injected with APAP to induce ALI, and then injected with flagellin at one hour after APAP administration. Flagellin attenuated APAP-induced ALI based on decreased histopathologic lesions, serum biochemical, oxidative stress, and inflammation. Furthermore, the protective effects of flagellin were abolished by TH1020 (a TLR5 antagonist) treatment. These results suggest that flagellin exerted protective effects on ALI via TLR5 activation. Mechanistically, flagellin injection promoted the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) to the nucleus in hepatocytes. Consistent with the in vivo results, flagellin increased the activation of Nrf2 in hepatocytes, resulting in decreased APAP toxicity. ML385, a selective inhibitor of Nrf2, abolished the flagellin-mediated hepatoprotective effects in damaged livers and hepatocytes. Additionally, the flagellin-induced Nrf2 translocation was dependent upon the activation of TLR5-JNK/p38 pathways. These findings suggest that TLR5 signaling-induced Nrf2 activation, at least partially, contributed to the protection against APAP-induced ALI by flagellin treatment.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Flagellin/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 5/metabolism , Animals , Chemical and Drug Induced Liver Injury/metabolism , Down-Regulation , Flagellin/administration & dosage , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL
5.
Int J Biol Macromol ; 174: 457-465, 2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33493561

ABSTRACT

Vibrio anguillarum is a globally distributed aquatic pathogen, and its flagellin B (FlaB) protein can evoke innate immune responses in hosts. In order to explore the role of FlaB in V. anguillarum infection, we constructed a FlaB-deficient mutant using overlapping PCR and two-step homologous recombination, and gene sequencing confirmed successful knockout of the FlaB gene. Scanning electron microscopy showed no significant differences in the morphological structure of the flagellum between wild-type and FlaB-deficient strains. The mutant was subsequently injected into the freshwater prawn (Macrobrachium rosenbergii) to explore its pathogenicity in the host, and expression of myeloid differentiation factor 88, prophenoloxidase, catalase, superoxide dismutase and glutathione peroxidase was investigated by real-time PCR. The results showed that deletion of FlaB had little effect on V. anguillarum-induced expression of these immune-related genes (p > 0.05). In general, the FlaB mutant displayed similar flagella morphology and immune characteristics to the wild-type strain, hence we speculated that knockout of FlaB might promote the expression and function of other flagellin proteins. Furthermore, this study provides a rapid and simple method for obtaining stable mutants of V. anguillarum free from foreign plasmid DNA.


Subject(s)
Arthropod Proteins/genetics , Flagellin/administration & dosage , Mutation , Palaemonidae/immunology , Vibrio/metabolism , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Catalase/genetics , Catechol Oxidase/genetics , Cloning, Molecular , Enzyme Precursors/genetics , Flagellin/genetics , Flagellin/immunology , Gene Expression Regulation , Gene Knockout Techniques , Glutathione Peroxidase/genetics , Immunity, Innate , Microscopy, Electron, Scanning , Myeloid Differentiation Factor 88/genetics , Palaemonidae/genetics , Superoxide Dismutase/genetics , Vibrio/immunology
6.
ACS Appl Mater Interfaces ; 13(4): 4844-4852, 2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33486952

ABSTRACT

The combination of photothermal therapy (PTT) and toll-like receptor (TLR)-mediated immunotherapy can elicit antitumor immunity and modulate the immunosuppressive tumor microenvironment (TME). Unlike other TLRs, TLR-5 is a promising target for immune activation, as its expression is well-maintained even during immunosenescence. Here, we developed a unique tumor microenvironment-regulating immunosenescence-independent nanostimulant consisting of TLR-5 adjuvant Vibrio vulnificus flagellin B (FlaB) conjugated onto the surface to an IR 780-loaded hyaluronic acid-stearylamine (HIF) micelles. These HIF micelles induced immune-mediated cell death via PTT when irradiated with a near-infrared laser. In comparison with PTT alone, the combination of in situ-generated tumor-associated antigens produced during PTT and the immune adjuvant FlaB demonstrated enhanced vaccine-like properties and modulated the TME by suppressing immune-suppressive regulatory cells (Tregs) and increasing the fraction of CD103+ migratory dendritic cells, which are responsible for trafficking tumor antigens to draining lymph nodes (DLNs). This combinatorial strategy (i.e., applying a TLR-5 adjuvant targeted to immunosenescence-independent TLR-5 and the in situ photothermal generation of tumor-associated antigens) is a robust system for next-generation immunotherapy and could even be applied in elderly patients, thus broadening the clinical scope of immunotherapy strategies.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Flagellin/therapeutic use , Immunotherapy , Nanoparticles/therapeutic use , Neoplasms/therapy , Photothermal Therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Cell Line, Tumor , Female , Flagellin/administration & dosage , Flagellin/immunology , HEK293 Cells , Humans , Immunosenescence/drug effects , Immunosenescence/radiation effects , Immunotherapy/methods , Infrared Rays/therapeutic use , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Neoplasms/immunology , Neoplasms/pathology , Photothermal Therapy/methods , Toll-Like Receptor 5/antagonists & inhibitors , Toll-Like Receptor 5/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Vibrio vulnificus/immunology
7.
J Vis Exp ; (178)2021 12 16.
Article in English | MEDLINE | ID: mdl-34978289

ABSTRACT

With the increase of incidence, inflammatory bowel diseases (IBD), which are chronic diseases affecting the gastrointestinal tract, impose a considerable health and financial burden on individuals and society. Therefore, it is critical to investigate the mechanisms underlying the pathogenesis and development of IBD. Here, a gut microbiota antigen-specific T cell transfer colitis model is described. CBir1 flagellin has been recognized as the immunodominant gut bacterial antigen in experimental colitis and patients with Crohn's disease. CBir1 TCR transgenic naϊve CD4+ T cells, specific to CBir1 flagellin, can induce chronic colitis after adoptive transfer into immune-deficient Rag1-/- mice. The disease severity is assessed by histopathology. The CD4+ T cell phenotypes in colonic lamina propria are also determined. This model closely resembles the development of IBD, which provides an ideal murine model for investigating the mechanisms driving the pathogenesis of IBD and testing the potential drugs for treating IBD.


Subject(s)
CD4-Positive T-Lymphocytes , Colitis , Flagellin , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Flagellin/administration & dosage , Flagellin/genetics , Flagellin/immunology , Humans , Inflammation , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
8.
Front Immunol ; 11: 581165, 2020.
Article in English | MEDLINE | ID: mdl-33312172

ABSTRACT

Bacteria-released components can modulate host innate immune response in the absence of direct host cell-bacteria interaction. In particular, bacteria-derived outer membrane vesicles (OMVs) were recently shown to activate host caspase-11-mediated non-canonical inflammasome pathway via deliverance of OMV-bound lipopolysaccharide. However, further precise understanding of innate immune-modulation by bacterial OMVs remains elusive. Here, we present evidence that flagellated bacteria-released OMVs can trigger NLRC4 canonical inflammasome activation via flagellin delivery to the cytoplasm of host cells. Salmonella typhimurium-derived OMVs caused a robust NLRC4-mediated caspase-1 activation and interleukin-1ß secretion in macrophages in an endocytosis-dependent, but guanylate-binding protein-independent manner. Notably, OMV-associated flagellin is crucial for Salmonella OMV-induced inflammasome response. Flagellated Pseudomonas aeruginosa-released OMVs consistently promoted robust NLRC4 inflammasome activation, while non-flagellated Escherichia coli-released OMVs induced NLRC4-independent non-canonical inflammasome activation leading to NLRP3-mediated interleukin-1ß secretion. Flagellin-deficient Salmonella OMVs caused a weak interleukin-1ß production in a NLRP3-dependent manner. These findings indicate that Salmonella OMV triggers NLRC4 inflammasome activation via OMV-associated flagellin in addition to a mild induction of non-canonical inflammasome signaling via OMV-bound lipopolysaccharide. Intriguingly, flagellated Salmonella-derived OMVs induced more rapid inflammasome response than flagellin-deficient Salmonella OMV and non-flagellated Escherichia coli-derived OMVs. Supporting these in vitro results, Nlrc4-deficient mice showed significantly reduced interleukin-1ß production after intraperitoneal challenge with Salmonella-released OMVs. Taken together, our results here propose that NLRC4 inflammasome machinery is a rapid sensor of bacterial OMV-bound flagellin as a host defense mechanism against bacterial pathogen infection.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Bacterial Outer Membrane/immunology , Calcium-Binding Proteins/immunology , Flagellin/immunology , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Bacterial Proteins/immunology , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Caspase 1/metabolism , Cytosol/immunology , Endocytosis , Enzyme Activation , Flagellin/administration & dosage , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Host Microbial Interactions/immunology , Immunity, Innate , Inflammasomes/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology
9.
J Immunol ; 205(10): 2873-2882, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33008952

ABSTRACT

The TLR5 agonist flagellin is a potent adjuvant and is currently being developed for use in vaccines. The mechanisms that drive flagellin's activity are influenced by its administration route. Previous studies showed that lung structural cells (especially epithelial cells lining the conducting airways) are pivotal for the efficacy of intranasally administered flagellin-containing vaccines. In this study, we looked at how the airway epithelial cells (AECs) regulate the flagellin-dependent stimulation of Ag-specific CD4+ T cells and the Ab response in mice. Our results demonstrate that after sensing flagellin, AECs trigger the release of GM-CSF in a TLR5-dependent fashion and the doubling of the number of activated type 2 conventional dendritic cells (cDC2s) in draining lymph nodes. Furthermore, the neutralization of GM-CSF reduced cDC2s activation. This resulted in lower of Ag-specific CD4+ T cell count and Ab titers in mice. Our data indicate that during pulmonary immunization, the GM-CSF released by AECs orchestrates the cross-talk between cDC2s and CD4+ T cells and thus drives flagellin's adjuvant effect.


Subject(s)
Epithelial Cells/metabolism , Flagellin/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Respiratory Mucosa/immunology , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells/immunology , Female , Flagellin/administration & dosage , Immunity, Mucosal , Immunogenicity, Vaccine , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Knockout , Models, Animal , Primary Cell Culture , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Toll-Like Receptor 5/agonists , Toll-Like Receptor 5/genetics , Vaccines/administration & dosage
10.
Mol Med Rep ; 22(4): 2753-2766, 2020 10.
Article in English | MEDLINE | ID: mdl-32945411

ABSTRACT

Acute lung injury (ALI) is a complex condition frequently encountered in the clinical setting. The aim of the present study was to investigate the effect of conditioned media (CM) from human adipose­derived mesenchymal stromal cells (MSCs) activated by flagellin (F­CM), a Toll­like receptor 5 ligand, on inflammation­induced lung injury. In the in vitro study, RAW264.7 macrophages treated with F­CM had a higher proportion of cells with the M2 phenotype, lower expression of pro­inflammatory factors and stronger expression of anti­inflammatory genes compared with the CM from normal adipose­derived MSCs. Furthermore, in vivo experiments were performed in mice with ALI induced by intraperitoneal injection of lipopolysaccharide. F­CM significantly alleviated the lung exudation, inhibited inflammatory cell recruitment in lung tissues and decreased the concentration of inflammatory factors in the bronchoalveolar lavage fluid. These findings indicated that F­CM has superior anti­inflammation ability compared with CM, and that it may represent a promising therapeutic approach to the treatment of inflammation­induced ALI.


Subject(s)
Acute Lung Injury/therapy , Anti-Inflammatory Agents/administration & dosage , Culture Media, Conditioned/chemistry , Flagellin/administration & dosage , Lipopolysaccharides/adverse effects , Mesenchymal Stem Cells/cytology , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Cell Culture Techniques , Cells, Cultured , Disease Models, Animal , Flagellin/pharmacology , Gene Expression Regulation , Humans , Injections, Intraperitoneal , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , RAW 264.7 Cells , THP-1 Cells
11.
Respir Res ; 21(1): 222, 2020 Aug 26.
Article in English | MEDLINE | ID: mdl-32847538

ABSTRACT

Cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) are associated with acute and chronic bacterial infections of the lung. Excessive differentiation of basal cells to mucus-producing goblet cells can result in mucus hyperproduction and loss of mucociliary clearance in the airways of CF and COPD patients. Here, we aimed to investigate the effect of pathogen-associated molecular patterns (PAMPs) on the differentiation of human 3D bronchospheres. Primary human bronchial epithelial cells (HBECs) were differentiated to bronchospheres in the presence of bacterial flagellin and LPS and the synthetic Toll-like receptor (TLR) ligands Pam3CSK4 (TLR-2) and polyinosinic:polycytidylic acid (pIC, TLR-3). Electron and fluorescence microscopy showed that the differentiation of bronchospheres associated with the formation of lumina and appearance of cilia within 30 days after seeding. Incubation with flagellin resulted in a decreased formation of lumina and loss of cilia formation. Incubation with Pam3CSK, pIC, and LPS did not significantly affect formation of lumina and ciliation. Mucus production was strongly increased in response to flagellin and, to a lesser degree, in response to Pam3CSK4. Our results indicate that bacterial factors, such as flagellin, drive the differentiation of the respiratory epithelium towards mucus hyperproduction.


Subject(s)
Bronchi/metabolism , Flagellin/metabolism , Mucociliary Clearance/physiology , Mucus/metabolism , Organoids/metabolism , Respiratory Mucosa/metabolism , Bronchi/microbiology , Cells, Cultured , Flagellin/administration & dosage , Humans , Mucus/microbiology , Organoids/microbiology , Organoids/ultrastructure , Respiratory Mucosa/microbiology , Respiratory Mucosa/ultrastructure
12.
Arch Virol ; 165(6): 1299-1309, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32253616

ABSTRACT

Since late 2010, outbreaks of porcine epidemic diarrhea (PED) have been reported in the swine industry in China. A variant PEDV strain that differs from strain CV777 causes prevalent PEDV infections which commercial vaccines based on CV777 cannot provide complete protection. In this study, we designed a new vaccine based on the epidemic PEDV strain AH2012/12, adjuvanted with flagellin, a mucosal adjuvant that induces mucosal and systemic production of IgA. Three groups of pregnant sows were immunized twice, with a 14-day interval, with PEDV adjuvanted with flagellin, PEDV alone, or PBS before farrowing, and newborn piglets from each group were selected and challenged with PEDV. Immunization with this vaccine elicited high levels of IgG, IgA, and neutralizing antibodies in the serum and colostrum of sows, and newborn piglets were protected against PEDV while suckling. This study should guide the prevention and control strategies for PEDV infection, thereby reducing the losses associated with this virus.


Subject(s)
Coronavirus Infections/veterinary , Flagellin/administration & dosage , Porcine epidemic diarrhea virus/immunology , Swine Diseases/prevention & control , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cell Line , Colostrum/chemistry , Coronavirus Infections/pathology , Coronavirus Infections/prevention & control , Female , Flagellin/immunology , Immunization , Pregnancy , Swine , Swine Diseases/pathology , Vaccines, Inactivated/immunology , Viral Vaccines/administration & dosage
13.
Physiol Genomics ; 52(5): 217-221, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32275178
14.
Front Immunol ; 11: 171, 2020.
Article in English | MEDLINE | ID: mdl-32132997

ABSTRACT

Intranasal mucosal vaccines are an attractive approach to induce protective mucosal immune responses. Activation of lung antigen presenting cells (APCs), a phenotypically and functionally heterogeneous cell population located at distinct mucosal sites, may be key to the immunogenicity of such vaccines. Understanding responsiveness of newborn lung APCs to adjuvants may the inform design of efficacious intranasal vaccines for early life, when most infections occur. Here, we characterized and phenotyped APCs from neonatal (7 days of life) and adult (6-8 weeks of age) mice. Neonatal mice demonstrated a relatively high abundance of alveolar macrophages (AMs), with lower percentages of plasmacytoid dendritic cells (pDCs), CD103+ (cDC1), and CD11b+ (cDC2) DCs. Furthermore, neonatal CD103+ and CD11b+ DC subsets demonstrated a significantly lower expression of maturation markers (CD40, CD80, and CD86) as compared to adult mice. Upon stimulation of lung APC subsets with a panel of pattern recognition receptor (PRR) agonists, including those engaging TLRs or STING, CD11c+ enriched cells from neonatal and adult mice lungs demonstrated distinct maturation profiles. Of the agonists tested, the TLR5 ligand, flagellin, was most effective at activating neonatal lung APCs, inducing significantly higher expression of maturation markers on CD103+ (cDC1) and CD11b+ (cDC2) subsets. Intranasal administration of flagellin induced a distinct migration of CD103+ and CD11b+ DC subsets to the mediastinal lymph nodes (mLNs) of neonatal mice. Overall, these findings highlight age-specific differences in the maturation and responsiveness of lung APC subsets to different PRR agonists. The unique efficacy of flagellin in enhancing lung APC activity suggests that it may serve as an effective adjuvant for early life mucosal vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Flagellin/administration & dosage , Lung/immunology , Phenotype , Respiratory Mucosa/immunology , Toll-Like Receptor 5/agonists , Administration, Intranasal , Animals , Animals, Newborn , Cells, Cultured , Dendritic Cells/immunology , Female , Immunity, Mucosal , Lung/drug effects , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Respiratory Mucosa/drug effects , Vaccination/methods , Vaccines/immunology
15.
Biosci Rep ; 40(4)2020 04 30.
Article in English | MEDLINE | ID: mdl-32202301

ABSTRACT

Toll-like receptor (TLR) ligands have emerged as the attractive adjuvant for subunit vaccines. However, selection of TLR ligands needs to be rationally chosen on the basis of antigen and adjuvant properties. In the present study, we expressed the Ag473 lipoprotein from Neisseria meningitides, flagellin FlaB from Vibrio vulnificus and heat shock protein 70 from Mycobacterium tuberculosis (mHsp70) in Escherichia coli as single proteins and fusion proteins with VP2 protein of infectious bursal disease virus (IBDV). Both cellular and humoral adjuvanticities of the three TLR ligands were compared by immunization of mice in two different ways. Among the three co-administered TLR ligands, recombinant Ag473 lipoprotein exhibited the highest cellular and humoral adjuvanticities, including promotion of IL-4, IL-12, IFN-γ and IBDV VP2-specific antibody production. Among the three genetically fused TLR ligands, fusion with Ag473 D1 domain exhibited the highest cellular and humoral adjuvanticities. Overall, the adjuvanticities of genetically fused TRL ligands were significantly higher than that of co-administered TLR ligands. Fusion with Ag473 D1 domain exhibited superior adjuvanticity among the three TLR ligands delivered in two different ways.


Subject(s)
Bacterial Proteins/immunology , Immunogenicity, Vaccine , Recombinant Fusion Proteins/immunology , Vaccines, Subunit/immunology , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Female , Flagellin/administration & dosage , Flagellin/genetics , Flagellin/immunology , HSP70 Heat-Shock Proteins/administration & dosage , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/immunology , Ligands , Mice , Models, Animal , Protein Domains/genetics , Protein Domains/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vibrio vulnificus/genetics , Vibrio vulnificus/immunology , Viral Structural Proteins/administration & dosage , Viral Structural Proteins/genetics , Viral Structural Proteins/immunology
16.
Am J Pathol ; 190(3): 614-629, 2020 03.
Article in English | MEDLINE | ID: mdl-31972159

ABSTRACT

Bacterial flagellin, recognized by cell surface of Toll-like receptor (TLR) 5, is a potent activator of many types of cells, leading to the activation of innate or adaptive immunity, which are pivotal in regulating fibrotic process. However, the exact role of TLR5 signaling in hepatic fibrogenesis remains unclear, and this study aims to elucidate its underlying mechanisms. Flagellin was injected to hepatotoxin- and cholestasis-induced liver fibrosis murine models. Flagellin-induced TLR5 activation significantly decreased the severity of liver fibrosis. Interestingly, the expression levels of IL-1 receptor antagonist (IL1RN) and interferon (IFN)ß markedly increased in fibrotic livers on flagellin treatment. Consistently, in vivo activation of TLR5 signaling markedly increased IFNß and IL1RN expression in the livers. Notably, flagellin injection significantly exacerbated the severity of liver fibrosis in IFN-α/ß receptor 1 (IFNAR1) knockout mice. Furthermore, hepatic expression of IL1RN in the fibrotic livers of IFNAR1 knockout mice was significantly lower than those of wild-type mice. In support of these findings, flagellin-mediated IL1RN production is not sufficient to alleviate the severity of hepatic fibroinflammatory responses in IFNAR1-deficient milieu. Finally, hepatic stellate cells treated with IL1RN had significantly decreased cellular activation and its associated fibrogenic responses. Collectively, manipulation of TLR5 signaling may be a promising therapeutic strategy for the treatment of liver fibrosis.


Subject(s)
Cholestasis/complications , Interferon-beta/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Liver Cirrhosis/physiopathology , Signal Transduction , Toll-Like Receptor 5/metabolism , Adaptive Immunity , Animals , Disease Models, Animal , Disease Progression , Flagellin/administration & dosage , Immunity, Innate , Interferon-beta/genetics , Interleukin 1 Receptor Antagonist Protein/genetics , Liver Cirrhosis/chemically induced , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Toll-Like Receptor 5/genetics
17.
Vaccine ; 38(3): 416-422, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31735501

ABSTRACT

The immunization of poultry where H5 and H7 influenza viruses (IVs) are endemic is one of the strategies to prevent unexpected zoonoses. Our group has been focused on conserved HA-epitopes as potential vaccine candidates to obtain multivalent immune responses against distinct IV subtypes. In this study, two conserved epitopes (NG-34 and CS-17) fused to flagellin were produced in a Baculovirus platform based on Trichoplusia ni larvae as living biofactories. Soluble extracts obtained from larvae expressing "flagellin-NG34/CS17 antigen" were used to immunize chickens and the efficacy of the vaccine was evaluated against a heterologous H7N1 HPAIV challenge in chickens. The flagellin-NG34/CS17 vaccine protected the vaccinated chickens and blocked viral shedding orally and cloacally. Furthermore, no apparent clinical signs were monitored in 10/12 vaccinated individuals. The mechanism of protection conferred is under investigation.


Subject(s)
Flagellin/administration & dosage , Granulovirus , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Influenza A Virus, H7N1 Subtype , Influenza in Birds/prevention & control , Administration, Intranasal , Amino Acid Sequence , Animals , Chickens , Dogs , Flagellin/immunology , Granulovirus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization/methods , Influenza A Virus, H7N1 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza in Birds/immunology , Larva/immunology , Madin Darby Canine Kidney Cells , Zoonoses/immunology , Zoonoses/prevention & control
18.
Nat Commun ; 10(1): 5650, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827095

ABSTRACT

Alterations in gut microbiota composition are associated with metabolic syndrome and chronic inflammatory diseases such as inflammatory bowel disease. One feature of inflammation-associated gut microbiotas is enrichment of motile bacteria, which can facilitate microbiota encroachment into the mucosa and activate pro-inflammatory gene expression. Here, we set out to investigate whether elicitation of mucosal anti-flagellin antibodies by direct administration of purified flagellin might serve as a general vaccine against subsequent development of chronic gut inflammation. We show, in mice, that repeated injection of flagellin elicits increases in fecal anti-flagellin IgA and alterations in microbiota composition, reduces fecal flagellin concentration, prevents microbiota encroachment, protects against IL-10 deficiency-induced colitis, and ameliorates diet-induced obesity. Flagellin's impact on the microbiota is B-lymphocyte dependent and, in humans, obese subjects exhibit increased levels of fecal flagellin and reduced levels of fecal flagellin-specific IgA, relative to normal weight subjects. Thus, administration of flagellin, and perhaps other pathobiont antigens, may confer some protection against chronic inflammatory diseases.


Subject(s)
Adaptive Immunity , Bacterial Vaccines/immunology , Colitis/prevention & control , Flagellin/immunology , Gastrointestinal Microbiome , Animals , Antibodies, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Colitis/genetics , Colitis/immunology , Colitis/microbiology , Feces/microbiology , Flagellin/administration & dosage , Flagellin/genetics , Humans , Immunoglobulin A/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice, Inbred C57BL , Obesity/genetics , Obesity/immunology , Obesity/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology
19.
Theranostics ; 9(16): 4811-4826, 2019.
Article in English | MEDLINE | ID: mdl-31367259

ABSTRACT

The viral E proteins of dengue virus (DENV) and Zika virus (ZIKV) are the major viral proteins involved in receptor binding and fusion, and for the induction of protective antibodies against viral infections. DIII of the E proteins is an independent domain and stretches out on the virion surface that can elicit type-specific neutralizing antibodies. For recombinant DIII vaccine development, prime-boost immunizations can provide an advantage of eliciting more type-specific neutralizing antibodies by recalling DIII antigens after DIII booster to improve protection. Methods: The DIII of the E genes of DENV and ZIKV were fused with bacterial fliC gene for the expression of flagellin-DIII (FliC-DIII) fusion proteins. Prime-boost immunization strategies by the second-dose booster of four DENV serotype or ZIKV FliC-DIII fusion proteins were used to investigate the induction of neutralizing antibodies and protection against viral infections. Cross-reactive non-neutralizing antibodies in each group of antisera were also examined using in vitro antibody-dependent enhancement (ADE) assay. A series of glycan-masking E antigens were finally constructed for prime-boost immunizations to abolish the elicitation of cross-reactive non-neutralizing antibodies for ADE activity. Results: We showed that inclusion of a bivalent live-attenuated vaccine with a FliC-DIII booster is superior in eliciting neutralization titers and protection in vivo against all four-serotype DENVs. We also demonstrated that recombinant adenovirus vectors encoding four-serotype DENV prMEs with a FliC-DIII prime-boost scheme is capable of eliciting good antibody responses. In contract, recombinant adenovirus vector of ZIKV prME gene priming, followed by ZIKV FliC-DIII booster did not improve vaccine efficacy. The glycan-masking mutation on the ZIKV E protein ij loop (E-248NHT), but not on DENV2 E protein ij loop (E-242NHT), resulted in abolishing the elicitation of cross-reactive antibodies for DENV and ZIKV infection enhancements. Conclusions: Our findings can provide useful information for designing novel immunogens and vaccination strategies in an attempt to develop a safe and efficacious DENV or ZIKV vaccine.


Subject(s)
Dengue Virus/immunology , Dengue/immunology , Flagellin/immunology , Viral Envelope Proteins/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Antibodies, Viral/immunology , Cross Reactions , Dengue/prevention & control , Dengue/virology , Dengue Virus/chemistry , Dengue Virus/genetics , Flagellin/administration & dosage , Flagellin/genetics , Humans , Immunization , Immunization, Secondary , Mice , Mice, Inbred BALB C , Polysaccharides/administration & dosage , Polysaccharides/immunology , Protein Domains , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
20.
Exp Mol Med ; 51(7): 1-13, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31292433

ABSTRACT

We hypothesized that NADPH oxidase 4 (Nox4) is involved in the formation of neointimal atherosclerotic plaques through the migration of smooth muscle cells (SMCs) in response to flagellin. Here, we demonstrate that TLR5-mediated Nox4 activation regulates the migration of SMCs, leading to neointimal plaque formation in atherosclerosis. To investigate the molecular mechanism by which the TLR5-Nox4 cascade mediates SMC migration, we analyzed the signaling cascade in primary vascular SMCs (VSMCs) from wild-type (WT) or Nox4 KO mice. Stimulation of VSMCs from Nox4 KO mice with flagellin failed to induce H2O2 production and Rac activation compared with stimulation of VSMCs from WT mice. Moreover, the migration of Nox4-deficient VSMCs was attenuated in response to flagellin in transwell migration and wound healing assays. Finally, we performed partial carotid artery ligation in ApoE KO and Nox4ApoE DKO mice fed a high-fat diet (HFD) with or without recombinant FliC (rFliC) injection. Injection of rFliC into ApoE KO mice fed a HFD resulted in significantly increased SMC migration into the intimal layer, whereas SMC accumulation was not detected in Nox4ApoE DKO mice. We conclude that activation of the TLR5-Nox4 cascade plays an important role in the formation of neointimal atherosclerotic plaques.


Subject(s)
Atherosclerosis/enzymology , Flagellin/administration & dosage , NADPH Oxidase 4/metabolism , Plaque, Atherosclerotic/enzymology , Signal Transduction , Toll-Like Receptor 5/metabolism , Animals , Atherosclerosis/pathology , Carotid Arteries/pathology , Cell Movement , Chemokines/analysis , Diet, High-Fat/adverse effects , Flagellin/genetics , Male , Mice, Knockout, ApoE , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 4/genetics , Neointima/enzymology , Neointima/pathology , Phenotype , Plaque, Atherosclerotic/pathology , Salmonella enteritidis/genetics , Toll-Like Receptor 5/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...