Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 11(1): 1187, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32132528

ABSTRACT

Induction of antigen-specific immune activation by the maturation of dendritic cells (DCs) is a strategy used for cancer immunotherapy. In this study, we find that FimH, which is an Escherichia coli adhesion portion, induces toll-like receptor 4-dependent and myeloid differentiation protein 2-independent DC maturation in mice in vivo. A combined treatment regimen with FimH and antigen promotes antigen-specific immune activation, including proliferation of T cells, production of IFN-γ and TNF-α, and infiltration of effector T cells into tumors, which consequently inhibits tumor growth in mice in vivo against melanoma and carcinoma. In addition, combined therapeutic treatment of anti-PD-L1 antibodies and FimH treatment efficiently inhibits CT26 tumor growth in BALB/c mice. Finally, FimH promotes human peripheral blood DC activation and syngeneic T-cell proliferation and activation. Taken together, these findings demonstrate that FimH can be a useful adjuvant for cancer immunotherapy.


Subject(s)
Adhesins, Escherichia coli/administration & dosage , Adjuvants, Immunologic/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Dendritic Cells/immunology , Fimbriae Proteins/administration & dosage , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Adhesins, Escherichia coli/immunology , Animals , Cell Line, Tumor/transplantation , Cell Proliferation , Dendritic Cells/metabolism , Disease Models, Animal , Fimbriae Proteins/immunology , Humans , Lymphocyte Activation , Mice , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Chimeric Antigen/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 4/metabolism
2.
Mol Cell Probes ; 45: 19-25, 2019 06.
Article in English | MEDLINE | ID: mdl-30940544

ABSTRACT

FimH is the adhesin of type I fimbriae expressed on Escherichia coli that can mediate specific adherence to host cells. High binding mutations in FimH are related to the adaptive evolution of bacteria. However, additional roles that these allelic variations may play remain elusive. To investigate novel biological functions of the mutations in FimH, we introduced four different variants of FimH by incorporating single amino acid substitutions at specific sites, namely A25P, G73R, A106, and T158P, respectively. In this study, adjuvant potential of FimH variants was evaluated by investigating their ability to trigger innate immune response to DC2.4 and adaptive immunity to improve immunological characteristics. The data revealed that purified A106 and T158P up-regulated the expression of co-stimulatory molecules critically involved in DC2.4 activation by interaction with TLR4, whereas A25P and G73R did not induce the phenotypic maturation of DC2.4. Besides, the culture of DC2.4 with A106 and T158P enhanced the release of cytokines and protein phagocytosis. When formulated with PAc, T158P elicited more robust PAc-specific IgG and IgA antibody responses compared to PBS, PAc and PAc+K12 groups and inhibited bacteria colonization. Collectively, the results confirmed that the T158P mutation located around the inter-domain interface of the protein induced a specific enhancement effect on adjuvant characteristics.


Subject(s)
Adhesins, Escherichia coli/administration & dosage , Antigens, Surface/administration & dosage , Fimbriae Proteins/administration & dosage , Point Mutation , Streptococcal Vaccines/administration & dosage , Streptococcus mutans/immunology , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/immunology , Adjuvants, Immunologic/administration & dosage , Amino Acid Substitution , Animals , Antigens, Surface/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Cell Line , Cytokines/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Mice , Phagocytosis , Streptococcal Vaccines/genetics , Streptococcal Vaccines/immunology
3.
APMIS ; 124(6): 444-52, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26918627

ABSTRACT

Urinary tract infections (UTIs) are among the most common infections in human. Innate immunity recognizes pathogen-associated molecular patterns (PAMPs) by Toll-like receptors (TLRs) to activate responses against pathogens. Recently, we demonstrated that MrpH.FimH fusion protein consisting of MrpH from Proteus mirabilis and FimH from Uropathogenic Escherichia coli (UPEC) results in the higher immunogenicity and protection, as compared with FimH and MrpH alone. In this study, we evaluated the innate immunity and adjuvant properties induced by fusion MrpH.FimH through in vitro and in vivo methods. FimH and MrpH.FimH were able to induce significantly higher IL-8 and IL-6 responses than untreated or MrpH alone in cell lines tested. The neutrophil count was significantly higher in the fusion group than other groups. After 6 h, IL-8 and IL-6 production reached a peak, with a significant decline at 24 h post-instillation in both bladder and kidney tissues. Mice instilled with the fusion and challenged with UPEC or P. mirabilis showed a significant decrease in the number of bacteria in bladder and kidney compared to control mice. The results of these studies demonstrate that the use of recombinant fusion protein encoding TLR-4 ligand represents an effective vaccination strategy that does not require the use of a commercial adjuvant. Furthermore, MrpH.FimH was presented as a promising vaccine candidate against UTIs caused by UPEC and P. mirabilis.


Subject(s)
Adhesins, Bacterial/administration & dosage , Adhesins, Escherichia coli/administration & dosage , Bacterial Vaccines/administration & dosage , Escherichia coli Infections/prevention & control , Fimbriae Proteins/administration & dosage , Proteus Infections/prevention & control , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/immunology , Adhesins, Bacterial/immunology , Adhesins, Escherichia coli/immunology , Animals , Bacterial Load , Cytokines/metabolism , Disease Models, Animal , Female , Fimbriae Proteins/immunology , Mice, Inbred BALB C , Neutrophils/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Urinary Tract/microbiology , Vaccines, Synthetic/administration & dosage
4.
Int Immunopharmacol ; 28(1): 70-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26033493

ABSTRACT

Urinary tract infections (UTIs) caused by Escherichia coli and Proteus mirabilis are an important cause of morbidity and with the high rate of relapse and spread of multi-drug resistant pathogens, pose a significant public health challenge worldwide. Lack of an efficacious commercial vaccine targeting both uropathogens makes development of a combined vaccine highly desirable. In this study the immunogenicity and protective efficacy of different formulations of FimH of UPEC, MrpH of P. mirabilis and their fusion protein (MrpH.FimH) subcutaneously administered with and without Monophosphoryl lipid A (MPL) adjuvant were evaluated. Our data showed that the subcutaneously administered proteins induced both serum and mucosal IgG, which MPL significantly improved developing a mixed Th1 and Th2 immune response. However, the preparations induced a higher systemic and mucosal IgG and IL-2 levels by this route compared to the intranasal. Immunization of mice with MrpH.FimH fusion with MPL or a mixture of FimH, MrpH and MPL conferred the highest protection of the bladder and kidneys when challenged with UPEC and P. mirabilis in a UTI mouse model. Therefore considering these results MrpH.FimH fusion with MPL administered subcutaneously or intranasally could be a promising vaccine candidate for elimination of UTIs caused by UPEC and P. mirabilis.


Subject(s)
Adhesins, Bacterial/immunology , Adhesins, Escherichia coli/immunology , Adjuvants, Immunologic/pharmacology , Fimbriae Proteins/immunology , Lipid A/analogs & derivatives , Proteus mirabilis/immunology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Adhesins, Bacterial/administration & dosage , Adhesins, Escherichia coli/administration & dosage , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Female , Fimbriae Proteins/administration & dosage , Immunoglobulin G/biosynthesis , Injections, Subcutaneous , Interleukin-2/biosynthesis , Kidney/immunology , Lipid A/administration & dosage , Lipid A/pharmacology , Mice , Mice, Inbred BALB C , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Urinary Bladder/immunology , Urinary Tract Infections/microbiology , Urinary Tract Infections/prevention & control
5.
Mol Immunol ; 64(2): 285-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25562574

ABSTRACT

Urinary tract infections (UTIs) caused by Uropathogenic Escherichia coli (UPEC) and Proteus mirabilis are among the most common infections in the world. Currently there are no vaccines available to confer protection against UTI in humans. In this study, the immune responses and protection of FimH of UPEC with MrpH antigen of P. mirabilis in different vaccine formulations with and without MPL adjuvant were assessed. Mice intranasally immunized with the novel fusion protein MrpH·FimH induced a significant increase in IgG and IgA in serum, nasal wash, vaginal wash, and urine samples. Mice immunized with fusion MrpH·FimH also showed a significant boost in cellular immunity. Addition of MPL as the adjuvant enhanced FimH and MrpH specific humoral and cellular responses in both systemic and mucosal samples. Vaccination with MrpH·FimH alone or in combination with MPL showed the highest efficiency in clearing bladder and kidney infections in mice challenged with UPEC and P. mirabilis. These findings may indicate that the protection observed correlates with the systemic, mucosal and cellular immune responses induced by vaccination with these preparations. Our data suggest MrpH·FimH fusion protein with or without MPL as adjuvant could be potential vaccine candidates for elimination of UPEC and P. mirabilis. These data altogether are promising and these formulations are good candidates for elimination of UPEC and P. mirabilis.


Subject(s)
Adhesins, Bacterial/immunology , Adhesins, Escherichia coli/immunology , Antibodies, Bacterial/biosynthesis , Fimbriae Proteins/immunology , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/immunology , Adhesins, Bacterial/administration & dosage , Adhesins, Bacterial/genetics , Adhesins, Escherichia coli/administration & dosage , Adhesins, Escherichia coli/genetics , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Female , Fimbriae Proteins/administration & dosage , Fimbriae Proteins/genetics , Gene Expression , Humans , Immunity, Humoral/drug effects , Immunity, Mucosal/drug effects , Immunization , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology
6.
Antiviral Res ; 92(2): 346-55, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21945041

ABSTRACT

Fimbriae H protein (FimH) is a novel TLR4 ligand that has been shown to stimulate the innate immune system and elicits protective responses against bacterial and viral infections. Here, we evaluated the protective role of local delivery of FimH against influenza A infection in a mouse model. We show that intranasal delivery of FimH prior to lethal challenge with influenza A virus, resulted in decreased morbidity and mortality in wild-type, but not TLR4(-/-), mice. Importantly, FimH was able to reduce the early viral burden in the lung leading to minimal cell infiltration into the airway lumen and reduced pulmonary pathology following infection in wild type mice compared to TLR4(-/-) mice. Local delivery of FimH to C57BL/6, not TLR4(-/-), mice in a prophylactic manner increased the IL-12 and RANTES responses as well as neutrophil recruitment into the airway lumen. These effects correlate to the course of influenza infection. The FimH-mediated antiviral response against influenza virus appears to be partially dependent on alveolar macrophages. The antiviral effects are likely mediated by the innate mediators (TNF-α, IL-12 or RANTES) and/or by activation of a feedback inhibition loop to curtail the pulmonary inflammation possibly be the potential mechanisms involved in FimH-mediated protection. FimH thus holds promise to be a possible prophylactic mean of control against influenza viral infection.


Subject(s)
Adhesins, Escherichia coli/administration & dosage , Adhesins, Escherichia coli/immunology , Fimbriae Proteins/administration & dosage , Fimbriae Proteins/immunology , Immunity, Innate/drug effects , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Toll-Like Receptor 4/administration & dosage , Toll-Like Receptor 4/immunology , Administration, Intranasal , Animals , Cell Movement , Chemokine CCL5/metabolism , Influenza A virus/immunology , Influenza A virus/pathogenicity , Interleukin-12/metabolism , Lung/pathology , Lung/virology , Macrophages, Alveolar/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Survival Analysis , Tumor Necrosis Factor-alpha/metabolism
7.
Poult Sci ; 83(12): 1973-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15615009

ABSTRACT

Immunization using a plasmid to deliver an encoded protein for expression in situ as the antigen is a promising technology. A plasmid encoding the enterotoxigenic Escherichia coli K88 fimbrial protein FaeG when injected into chickens stimulates the production of antibodies against the fimbrial protein, similar to what has been observed in mice. The efficacy of a genetic adjuvant on fimbrial antibody production was tested by introducing the gene for chicken interleukin-6 in tandem with the faeG gene. Expression of both the fimbrial FaeG protein and chicken interleukin-6 protein was confirmed in COS-M6 cells. Slightly higher antiFaeG antibody titer in chickens was obtained compared with immunization with the plasmid encoding FaeG alone, especially at 10 (19%, P < 0.05) and 12 (27%, P < 0.05) wk, respectively, after the secondary immunization. Elevated antiFaeG antibody titer induced by chicken interleukin-6 and FaeG proteins expressed jointly persisted longer than when induced by FaeG protein alone. This is the first report of an avian cytokine enhancing an immune response, and confirms that coexpression of the antigen and adjuvant from a plasmid delivered by DNA immunization is an effective protocol.


Subject(s)
Adhesins, Escherichia coli/genetics , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Chickens/immunology , Escherichia coli Proteins/immunology , Fimbriae Proteins/immunology , Interleukin-6/genetics , Plasmids/immunology , Adhesins, Escherichia coli/administration & dosage , Adhesins, Escherichia coli/immunology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Chickens/genetics , DNA, Bacterial/immunology , Egg Yolk/immunology , Escherichia coli/immunology , Escherichia coli Proteins/administration & dosage , Escherichia coli Proteins/genetics , Female , Fimbriae Proteins/administration & dosage , Fimbriae Proteins/genetics , Gene Expression Regulation , Immunization/veterinary , Interleukin-6/administration & dosage , Interleukin-6/immunology , Ovum/immunology , Plasmids/administration & dosage , Plasmids/genetics , Time Factors
8.
Vaccine ; 22(31-32): 4291-9, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15474721

ABSTRACT

The importance of adhesins in the pathogenicity of several bacteria resulted in studies on their usefulness in vaccines. In this study, the gene of the F4(K88)-fimbrial adhesin FaeG of the pathogenic enterotoxigenic Escherichia coli (ETEC) strain GIS26 was cloned in the pET30Ek-LIC vector and expressed with an N-terminal His- and S-tag in the cytoplasm of BL21(DE3). Recombinant FaeG (rFaeG) subunits were isolated from insoluble cytoplasmic aggregates and refolded into a native-like F4 receptor (F4R)-binding conformation. Indeed, the presence of conformational epitopes was shown by ELISA and the ability to bind the F4R was observed by inhibiting the adhesion of F4+ ETEC to F4R+ villi with increasing concentrations of native-like refolded rFaeG subunits. The rFaeG subunits appear as monomers, whereas the purified F4 fimbriae are multimers. Oral immunization of newly weaned piglets with native-like rFaeG induced a mucosal and systemic F4-specific immune response, significantly reducing F4+ E. coli excretion from 2 till 5 days following challenge infection. However, improvement of stability and immunogenicity of rFaeG is necessary since a higher F4-specific response was obtained following immunization with purified F4 fimbriae. Furthermore, the N-terminal fusion of a His- and S-tag was not detrimental for binding the F4R, supporting the use of FaeG as mucosal carrier. In conclusion, oral immunization with a recombinant fimbrial adhesin subunit of Escherichia coli induces a mucosal and systemic fimbriae-specific immune response.


Subject(s)
Adhesins, Bacterial/immunology , Adhesins, Escherichia coli/immunology , Antibodies, Bacterial/biosynthesis , Antibody Formation/immunology , Bacterial Vaccines/immunology , Immunity, Mucosal/immunology , Adhesins, Bacterial/administration & dosage , Adhesins, Bacterial/chemistry , Adhesins, Escherichia coli/administration & dosage , Adhesins, Escherichia coli/chemistry , Administration, Oral , Animals , Antibodies, Bacterial/analysis , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/chemistry , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Escherichia coli/chemistry , Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Feces/microbiology , Immunization , Immunoglobulin A/analysis , Immunoglobulin A/biosynthesis , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Immunoglobulin M/analysis , Immunoglobulin M/biosynthesis , Swine/immunology , Vaccines, Synthetic/analysis , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
9.
Vaccine ; 17(15-16): 2020-9, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10217602

ABSTRACT

An effective way of stimulating the mucosal immune system was examined in piglets, using F4 fimbriae of enterotoxigenic Escherichia coli (ETEC). It was demonstrated that purified F4 fimbriae, as opposed to ovalbumin (OVA), are powerful oral immunogens. Indeed, oral administration of purified F4 induced antigen-specific antibody-secreting cells (ASC) in the Peyer's patches, mesenteric lymph nodes (LN), blood and lamina propria 4, 7, 9 and 11 days postimmunization, respectively, indicating a stimulation of the mucosal immune system, whereas upon oral administration of OVA, no immune response was observed. Moreover, the induced F4-specific IgA and IgG antibody responses were comparable with those obtained upon oral infection with viable E. coli and intramuscular (i.m.) F4 injection, respectively. Furthermore, a priming of the mucosal immune system is better obtained by oral infection (ASC localized in mesenteric LN) than by i.m. F4 injection (ASC localized in spleen and retropharyngeal LN) since an oral boost with purified F4 induced a secondary response in the orally infected animal (mainly IgA and IgG ASC, rapid increase of IgA antibodies) while in the i.m. primed animal a secondary (more circulating antigen-specific ASC than in the unprimed animal) as well as a primary IgM and IgA response (mainly IgM ASC, slow increase of IgA antibodies), suggesting a primary mucosal response, were seen. An oral challenge of the naive control displayed a primary response (mainly IgM ASC, slow increase of IgA and IgG antibodies). The capacity of purified F4 to activate the mucosal immune system on oral administration, is of importance for the development of oral vaccines against ETEC infections.


Subject(s)
Antibodies, Bacterial/analysis , Antigens, Surface/immunology , Escherichia coli Proteins , Escherichia coli/immunology , Fimbriae Proteins , Immunity, Mucosal/immunology , Immunization , Intestines/immunology , Swine/immunology , Adhesins, Escherichia coli/administration & dosage , Adhesins, Escherichia coli/immunology , Adhesins, Escherichia coli/isolation & purification , Administration, Oral , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Antigens, Surface/administration & dosage , Antigens, Surface/isolation & purification , Bone Marrow Cells/immunology , Cells, Cultured , Female , Immunization, Secondary , Immunoglobulin Isotypes/analysis , Immunoglobulin Isotypes/blood , Immunoglobulin Isotypes/immunology , Intestines/cytology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...