Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Virus Res ; 325: 199048, 2023 02.
Article in English | MEDLINE | ID: mdl-36681192

ABSTRACT

As antimicrobial resistance (AMR) continues to increase, the therapeutic use of phages has re-emerged as an attractive alternative. However, knowledge of phage resistance development and bacterium-phage interaction complexity are still not fully interpreted. In this study, two lytic T4-like and T7-like phage infecting model Escherichia coli strain C600 are selected, and host genetic determinants involved in phage susceptibility and resistance are also identified using TraDIS strategy. Isolation and identification of the lytic T7-like show that though it belongs to the phage T7 family, genes encoding replication and transcription protein exhibit high differences. The TraDIS results identify a huge number of previously unidentified genes involved in phage infection, and a subset (six in susceptibility and nine in resistance) are shared under pressure of the two kinds of lytic phage. Susceptible gene wbbL has the highest value and implies the important role in phage susceptibility. Importantly, two susceptible genes QseE (QseE/QseF) and RstB (RstB/RstA), encoding the similar two-component system sensor histidine kinase (HKs), also identified. Conversely and strangely, outer membrane protein gene ompW, unlike the gene ompC encoding receptor protein of T4 phage, was shown to provide phage resistance. Overall, this study exploited a genome-wide fitness assay to uncover susceptibility and resistant genes, even the shared genes, important for the E. coli strain of both most popular high lytic T4-like and T7-like phages. This knowledge of the genetic determinants can be further used to analysis the behind function signatures to screen the potential agents to aid phage killing of MDR pathogens, which will greatly be valuable in improving the phage therapy outcome in fighting with microbial resistance.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Host Microbial Interactions , T-Phages , Bacteriophage T7/genetics , Bacteriophage T7/immunology , DNA-Binding Proteins , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Receptors, Adrenergic , Bacteriophage T4/genetics , Bacteriophage T4/immunology , T-Phages/genetics , T-Phages/immunology , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology
2.
Front Immunol ; 12: 745625, 2021.
Article in English | MEDLINE | ID: mdl-34712234

ABSTRACT

Developing influenza vaccines that protect against a broad range of viruses is a global health priority. Several conserved viral proteins or domains have been identified as promising targets for such vaccine development. However, none of the targets is sufficiently immunogenic to elicit complete protection, and vaccine platforms that can enhance immunogenicity and deliver multiple antigens are desperately needed. Here, we report proof-of-concept studies for the development of next-generation influenza vaccines using the bacteriophage T4 virus-like particle (VLP) platform. Using the extracellular domain of influenza matrix protein 2 (M2e) as a readout, we demonstrate that up to ~1,281 M2e molecules can be assembled on a 120 x 86 nanometer phage capsid to generate M2e-T4 VLPs. These M2e-decorated nanoparticles, without any adjuvant, are highly immunogenic, stimulate robust humoral as well as cellular immune responses, and conferred complete protection against lethal influenza virus challenge. Potentially, additional conserved antigens could be incorporated into the M2e-T4 VLPs and mass-produced in E. coli in a short amount of time to deal with an emerging influenza pandemic.


Subject(s)
Bacteriophage T4/immunology , Capsid Proteins/immunology , Influenza Vaccines , Vaccine Development/methods , Viral Matrix Proteins/immunology , Viroporin Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Bronchoalveolar Lavage Fluid/immunology , Capsid Proteins/genetics , Female , Humans , Immunogenicity, Vaccine , Influenza A virus/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Nanoparticle Drug Delivery System , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Peptide Library , Proof of Concept Study , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/genetics , Viroporin Proteins/genetics
3.
Int J Mol Sci ; 22(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669238

ABSTRACT

Protein assemblies provide unique structural features which make them useful as carrier molecules in biomedical and chemical science. Protein assemblies can accommodate a variety of organic, inorganic and biological molecules such as small proteins and peptides and have been used in development of subunit vaccines via display parts of viral pathogens or antigens. Such subunit vaccines are much safer than traditional vaccines based on inactivated pathogens which are more likely to produce side-effects. Therefore, to tackle a pandemic and rapidly produce safer and more effective subunit vaccines based on protein assemblies, it is necessary to understand the basic structural features which drive protein self-assembly and functionalization of portions of pathogens. This review highlights recent developments and future perspectives in production of non-viral protein assemblies with essential structural features of subunit vaccines.


Subject(s)
Ferritins/immunology , Vaccines, Subunit/immunology , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/immunology , Bacteriophage T4/immunology , Humans , Nanoparticles/chemistry , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand
4.
Microb Biotechnol ; 12(4): 730-741, 2019 07.
Article in English | MEDLINE | ID: mdl-31037835

ABSTRACT

Bacteriophages draw scientific attention in medicine and biotechnology, including phage engineering, widely used to shape biological properties of bacteriophages. We developed engineered T4-derived bacteriophages presenting seven types of tissue-homing peptides. We evaluated phage accumulation in targeted tissues, spleen, liver and phage circulation in blood (in mice). Contrary to expectations, accumulation of engineered bacteriophages in targeted organs was not observed, but instead, three engineered phages achieved tissue titres up to 2 orders of magnitude lower than unmodified T4. This correlated with impaired survival of these phages in the circulation. Thus, engineering of T4 phage resulted in the short-circulating phage phenotype. We found that the complement system inactivated engineered phages significantly more strongly than unmodified T4, while no significant differences in phages' susceptibility to phagocytosis or immunogenicity were found. The short-circulating phage phenotype of the engineered phages suggests that natural phages, at least those propagating on commensal bacteria of animals and humans, are naturally optimized to escape rapid neutralization by the immune system. In this way, phages remain active for longer when inside mammalian bodies, thus increasing their chance of propagating on commensal bacteria. The effect of phage engineering on phage pharmacokinetics should be considered in phage design for medical purposes.


Subject(s)
Bacteriophage T4/immunology , Blood/virology , Recombinant Proteins/metabolism , Viral Proteins/metabolism , Viral Tropism , Administration, Intravenous , Animals , Bacteriophage T4/genetics , Complement System Proteins/metabolism , Mice , Microbial Viability , Recombinant Proteins/genetics , Viral Proteins/genetics
5.
Biomed Res Int ; 2017: 3612015, 2017.
Article in English | MEDLINE | ID: mdl-29201902

ABSTRACT

Phage preparations used for phage therapy may have not only direct antibacterial action but also immunomodulating effects mediated by phages themselves as well as by bacterial antigens. Therefore phage application in patients with immune disorders, and especially with autoimmune diseases, requires special attention. The aim of this study was to investigate the effect of phage lysates (staphylococcal phages A3/R, phi200, and MS-1 cocktail, enterococcal phage 15/P, Pseudomonas phage 119x, and E. coli T4 phage) as well as purified T4 phage on the course of murine collagen-induced arthritis (CIA), commonly used as an animal model of rheumatoid arthritis. Intraperitoneal application of phage lysates or purified T4 phage did not aggravate the course of autoimmune joint disease. Moreover, although endotoxins are known to potentiate CIA, the systemic administration of phage lysate of Pseudomonas aeruginosa, which contains debris of this Gram-negative bacillus, did not significantly influence CIA although the sonicate of the corresponding bacterial strain did. Interestingly, a purified T4 phage revealed some anti-inflammatory activity when applied under the therapeutic scheme. Our preliminary results do not suggest that phages may aggravate the symptoms of rheumatoid arthritis. In contrast T4 phage may even exert an immunosuppressive effect.


Subject(s)
Arthritis, Experimental/therapy , Autoimmune Diseases/immunology , Bacteriophage T4/immunology , Phage Therapy/methods , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/immunology , Autoimmune Diseases/etiology , Autoimmune Diseases/virology , Bacteriophage T4/pathogenicity , Disease Models, Animal , Escherichia coli/immunology , Escherichia coli/virology , Humans , Immunomodulation/immunology , Mice , Phage Therapy/adverse effects , Pseudomonas Phages/immunology , Pseudomonas Phages/pathogenicity , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/virology , Staphylococcus Phages/immunology , Staphylococcus Phages/pathogenicity
6.
Viruses ; 7(8): 4783-99, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26308042

ABSTRACT

A specific humoral response to bacteriophages may follow phage application for medical purposes, and it may further determine the success or failure of the approach itself. We present a long-term study of antibody induction in mice by T4 phage applied per os: 100 days of phage treatment followed by 112 days without the phage, and subsequent second application of phage up to day 240. Serum and gut antibodies (IgM, IgG, secretory IgA) were analyzed in relation to microbiological status of the animals. T4 phage applied orally induced anti-phage antibodies when the exposure was long enough (IgG day 36, IgA day 79); the effect was related to high dosage. Termination of phage treatment resulted in a decrease of IgA again to insignificant levels. Second administration of phage induces secretory IgA sooner than that induced by the first administrations. Increased IgA level antagonized gut transit of active phage. Phage resistant E. coli dominated gut flora very late, on day 92. Thus, the immunological response emerges as a major factor determining phage survival in the gut. Phage proteins Hoc and gp12 were identified as highly immunogenic. A low response to exemplary foreign antigens (from Ebola virus) presented on Hoc was observed, which suggests that phage platforms can be used in oral vaccine design.


Subject(s)
Antibodies, Viral/analysis , Bacteriophage T4/immunology , Blood/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/virology , Immunity, Mucosal , Administration, Oral , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/immunology , Ebolavirus/genetics , Ebolavirus/immunology , Escherichia coli/isolation & purification , Escherichia coli/virology , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Longitudinal Studies , Male , Mice, Inbred C57BL , Viral Structural Proteins/immunology
7.
PLoS One ; 10(6): e0128779, 2015.
Article in English | MEDLINE | ID: mdl-26098893

ABSTRACT

Recombinant subunit vaccines should contain minimal non-pathogen motifs to reduce potential off-target reactivity. We recently developed a vaccine antigen against respiratory syncytial virus (RSV), which comprised the fusion (F) glycoprotein stabilized in its pre-fusion trimeric conformation by "DS-Cav1" mutations and by an appended C-terminal trimerization motif or "foldon" from T4-bacteriophage fibritin. Here we investigate the creation of a cysteine zipper to allow for the removal of the phage foldon, while maintaining the immunogenicity of the parent DS-Cav1+foldon antigen. Constructs without foldon yielded RSV F monomers, and enzymatic removal of the phage foldon from pre-fusion F trimers resulted in their dissociation into monomers. Because the native C terminus of the pre-fusion RSV F ectodomain encompasses a viral trimeric coiled-coil, we explored whether introduction of cysteine residues capable of forming inter-protomer disulfides might allow for stable trimers. Structural modeling indicated the introduced cysteines to form disulfide "rings", with each ring comprising a different set of inward facing residues of the coiled-coil. Three sets of rings could be placed within the native RSV F coiled-coil, and additional rings could be added by duplicating portions of the coiled-coil. High levels of neutralizing activity in mice, equivalent to that of the parent DS-Cav1+foldon antigen, were elicited by a 4-ring stabilized RSV F trimer with no foldon. Structure-based alteration of a viral coiled-coil to create a cysteine zipper thus allows a phage trimerization motif to be removed from a candidate vaccine antigen.


Subject(s)
Antigens, Viral/immunology , Cysteine/chemistry , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Neutralizing/immunology , Antigens, Viral/chemistry , Bacteriophage T4/immunology , Caveolin 1/chemistry , Caveolin 1/genetics , Caveolin 1/immunology , Cell Line , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Respiratory Syncytial Viruses/genetics , Vaccines, Subunit/immunology , Viral Proteins/immunology
8.
J Virol ; 88(21): 12551-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142581

ABSTRACT

UNLABELLED: Advances in phage therapy and novel applications of phages in biotechnology encourage interest in phage impact on human and animal immunity. Here we present comparative studies of immunogenic properties of T4 phage head surface proteins gp23*, gp24*, Hoc, and Soc, both as elements of the phage capsid and as isolated agents. Studies comprise evaluation of specific antibodies in the human population, analysis of the proteins' impact on the primary and secondary responses in mice, and the effect of specific antibodies on phage antibacterial activity in vitro and in vivo in mice. In humans, natural antibodies specific to T4-like phages were abundant (81% of investigated sera). Among those, significantly elevated levels of IgG antibodies only against major head protein (gp23*) were found, which probably reflected cross-reactions of T4 with antibodies induced by other T4-like phages. Both IgM and IgG antibodies were induced mostly by gp23* and Hoc, while weak (gp24*) and very weak (Soc) reactivities of other head proteins were noticed. Thus, T4 head proteins that markedly contribute to immunological memory to the phage are highly antigenic outer capsid protein (Hoc) and major capsid protein (gp23*). Specific anti-gp23* and anti-Hoc antibodies substantially decreased T4 phage activity in vitro and to some extent in vivo. Cooperating with antibodies, the immune complement system also contributed to annihilating phages. IMPORTANCE: Current descriptions of phage immunogenicity and its biological consequences are still vague and incomplete; thus, the central problem of this work is timely and may have strong practical implications. Here is presented the very first description of the contribution of bacteriophage proteins to immunological memory of the phage. Understanding of interactions between phages and mammalian immunology may help in biotechnological adaptations of phages for therapeutic requirements as well as for better appreciation of phage ecology and their role in the biosphere.


Subject(s)
Antibodies, Viral/blood , Bacteriophage T4/immunology , Viral Proteins/immunology , Adolescent , Adult , Animals , Complement System Proteins/immunology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Mice , Mice, Inbred C57BL , Young Adult
9.
Mol Immunol ; 57(2): 66-73, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24091293

ABSTRACT

Human and equine rabies immunoglobulins are currently available for passive immunization against rabies. However, these are hampered by the limited supply and some drawbacks. Advances in antibody engineering have led to overcome issues of clinical applications and to improve the protective efficacy. In the present study, we report the generation of a trivalent single-chain Fv (scFv50AD1-Fd), that recognizes the rabies virus glycoprotein, genetically fused to the trimerization domain of the bacteriophage T4 fibritin, termed 'foldon' (Fd). scFv50AD1-Fd was expressed as soluble recombinant protein in bacterial periplasmic space and purified through affinity chromatography. The molecular integrity and stability were analyzed by polyacrylamide gradient-gel electrophoresis, size-exclusion chromatography and incubation in human sera. The antigen-binding properties of the trimeric scFv were analyzed by direct and competitive-ELISA. Its apparent affinity constant was estimated at 1.4 ± 0.25 × 10(9)M(-1) and was 75-fold higher than its monovalent scFv (1.9 ± 0.68 × 10(7)M(-1)). The scFv50AD1-Fd neutralized rabies virus in a standard in vitro and in vivo neutralization assay. We showed a high neutralization activity up to 75-fold compared with monovalent format and the WHO standard serum. The gain in avidity resulting from multivalency along with an improved biological activity makes the trivalent scFv50AD1-Fd construct an important reagent for rabies protection. The antibody engineering approach presented here may serve as a strategy for designing a new generation of anti-rabies for passive immunotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Viral/immunology , Glycoproteins/immunology , Rabies virus/immunology , Rabies/therapy , Single-Chain Antibodies/therapeutic use , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Affinity/immunology , Bacteriophage T4/immunology , Bacteriophage T4/metabolism , Cell Line , Cricetinae , Immunization, Passive , Neutralization Tests , Rabies/immunology , Rabies/prevention & control , Rabies Vaccines/immunology , Recombinant Proteins/immunology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism
10.
PLoS Pathog ; 9(7): e1003495, 2013.
Article in English | MEDLINE | ID: mdl-23853602

ABSTRACT

Pneumonic plague is a highly virulent infectious disease with 100% mortality rate, and its causative organism Yersinia pestis poses a serious threat for deliberate use as a bioterror agent. Currently, there is no FDA approved vaccine against plague. The polymeric bacterial capsular protein F1, a key component of the currently tested bivalent subunit vaccine consisting, in addition, of low calcium response V antigen, has high propensity to aggregate, thus affecting its purification and vaccine efficacy. We used two basic approaches, structure-based immunogen design and phage T4 nanoparticle delivery, to construct new plague vaccines that provided complete protection against pneumonic plague. The NH2-terminal ß-strand of F1 was transplanted to the COOH-terminus and the sequence flanking the ß-strand was duplicated to eliminate polymerization but to retain the T cell epitopes. The mutated F1 was fused to the V antigen, a key virulence factor that forms the tip of the type three secretion system (T3SS). The F1mut-V protein showed a dramatic switch in solubility, producing a completely soluble monomer. The F1mut-V was then arrayed on phage T4 nanoparticle via the small outer capsid protein, Soc. The F1mut-V monomer was robustly immunogenic and the T4-decorated F1mut-V without any adjuvant induced balanced TH1 and TH2 responses in mice. Inclusion of an oligomerization-deficient YscF, another component of the T3SS, showed a slight enhancement in the potency of F1-V vaccine, while deletion of the putative immunomodulatory sequence of the V antigen did not improve the vaccine efficacy. Both the soluble (purified F1mut-V mixed with alhydrogel) and T4 decorated F1mut-V (no adjuvant) provided 100% protection to mice and rats against pneumonic plague evoked by high doses of Y. pestis CO92. These novel platforms might lead to efficacious and easily manufacturable next generation plague vaccines.


Subject(s)
Antigens, Bacterial/metabolism , Antigens, Viral/metabolism , Bacteriophage T4/immunology , Capsid/immunology , Plague/immunology , Vaccines, Virus-Like Particle/immunology , Yersinia pestis/virology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriophage T4/chemistry , Bacteriophage T4/metabolism , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Female , Mice , Mice, Inbred BALB C , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Particle Size , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plague/microbiology , Plague/prevention & control , Plague/virology , Plague Vaccine/chemistry , Plague Vaccine/immunology , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Protein Interaction Domains and Motifs , Random Allocation , Rats , Rats, Inbred BN , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Vaccines, Virus-Like Particle/chemistry , Yersinia pestis/immunology
11.
Proc Natl Acad Sci U S A ; 110(26): 10771-6, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23690590

ABSTRACT

Mucosal surfaces are a main entry point for pathogens and the principal sites of defense against infection. Both bacteria and phage are associated with this mucus. Here we show that phage-to-bacteria ratios were increased, relative to the adjacent environment, on all mucosal surfaces sampled, ranging from cnidarians to humans. In vitro studies of tissue culture cells with and without surface mucus demonstrated that this increase in phage abundance is mucus dependent and protects the underlying epithelium from bacterial infection. Enrichment of phage in mucus occurs via binding interactions between mucin glycoproteins and Ig-like protein domains exposed on phage capsids. In particular, phage Ig-like domains bind variable glycan residues that coat the mucin glycoprotein component of mucus. Metagenomic analysis found these Ig-like proteins present in the phages sampled from many environments, particularly from locations adjacent to mucosal surfaces. Based on these observations, we present the bacteriophage adherence to mucus model that provides a ubiquitous, but non-host-derived, immunity applicable to mucosal surfaces. The model suggests that metazoan mucosal surfaces and phage coevolve to maintain phage adherence. This benefits the metazoan host by limiting mucosal bacteria, and benefits the phage through more frequent interactions with bacterial hosts. The relationships shown here suggest a symbiotic relationship between phage and metazoan hosts that provides a previously unrecognized antimicrobial defense that actively protects mucosal surfaces.


Subject(s)
Bacteriophages/immunology , Bacteriophages/physiology , Mucus/immunology , Mucus/virology , Adhesiveness , Animals , Bacterial Adhesion/immunology , Bacteriophage T4/genetics , Bacteriophage T4/immunology , Bacteriophage T4/physiology , Bacteriophages/genetics , Cell Line , Escherichia coli/immunology , Escherichia coli/virology , Host-Pathogen Interactions/immunology , Humans , Mice , Models, Immunological , Mucus/microbiology , Symbiosis/immunology
12.
Vaccine ; 29(34): 5802-11, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21482223

ABSTRACT

Vascular endothelial growth factor (VEGF) has been known as a potential vasculogenic and angiogenic factor and its receptor (VEGFR2) is a major receptor to response to the angiogenic activity of VEGF. The technique that to break the immune tolerance of "self-antigens" associated with angiogenesis is an attractive approach for cancer therapy with T4 phage display system. In this experiment, mouse VEGFR2 was constructed on T4 phage nanometer-particle surface as a recombinant vaccine. T4-mVEGFR2 recombinant vaccine was identified by PCR and western blot assay. Immunotherapy with T4-mVEGFR2 was confirmed by protective immunity against Lewis lung carcinoma (LLC) in mice. The antibody against mVEGFR2 was detected by ELISPOT, ELISA and Dot ELISA. The inhibitive effects against angiogenesis were studied using CD31 and CD105 via histological analysis. VEGF-mediated endothelial cells proliferation and tube formation were inhibited in vitro by immunoglobulin induced by T4-mVEGFR2. The antitumor activity was substantiated from the adoptive transfer of the purified immunoglobulin. Antitumor activity and autoantibody production of mVEGFR2 could be neutralized by the depletion of CD4+T lymphocytes. These studies strongly suggest that T4-mVEGFR2 recombinant vaccine might be a promising antitumor approach.


Subject(s)
Carcinoma, Lewis Lung , Neovascularization, Pathologic , Vaccines, Synthetic , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/immunology , Adoptive Transfer , Angiogenesis Inhibitors/administration & dosage , Animals , Bacteriophage T4/genetics , Bacteriophage T4/immunology , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Cell Proliferation , Endoglin , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Immunotherapy/methods , Intracellular Signaling Peptides and Proteins/blood , Lung Neoplasms/blood supply , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/prevention & control , Neovascularization, Pathologic/therapy , Platelet Endothelial Cell Adhesion Molecule-1/blood , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
13.
Mol Microbiol ; 77(2): 444-55, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20497329

ABSTRACT

Bacteriophage T4 is decorated with 155 copies of the highly antigenic outer capsid protein, Hoc. The Hoc molecule (40 kDa) is present at the centre of each hexameric capsomer and provides a good platform for surface display of pathogen antigens. Biochemical and modelling studies show that Hoc consists of a string of four domains, three immunoglobulin (Ig)-like and one non-Ig domain at the C-terminus. Biochemical data suggest that the Hoc protein has two functional modules, a capsid binding module containing domains 1 and 4 and a solvent-exposed module containing domains 2 and 3. This model is consistent with the dumbbell-shaped cryo-EM density of Hoc observed in the reconstruction of the T4 capsid. Mutagenesis localized the capsid binding site to the C-terminal 25 amino acids, which are predicted to form two beta-strands flanking a capsid binding loop. Mutations in the loop residues, ESRNG, abolished capsid binding, suggesting that these residues might interact with the major capsid protein, gp23*. With the conserved capsid binding module forming a foothold on the virus and the solvent-exposed module able to adapt to bind to a variety of surfaces, Hoc probably provides survival advantages to the phage, such as increasing the virus concentration near the host, efficient dispersion of the virus and exposing the tail for more efficient contact with the host cell surface prior to infection.


Subject(s)
Bacteriophage T4/chemistry , Capsid Proteins/chemistry , Amino Acid Sequence , Bacteriophage T4/immunology , Binding Sites , Capsid Proteins/immunology , Models, Molecular , Molecular Sequence Data , Mutation , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Sequence Alignment
14.
Acta Pharmacol Sin ; 30(5): 637-45, 2009 May.
Article in English | MEDLINE | ID: mdl-19417736

ABSTRACT

AIM: Flt4 plays a key role in promoting tumor metastasis by stimulating solid tumor lymphangiogenesis. In this study, mouse Flt4 (mFlt4) was displayed on T4 phage in order to explore the feasibility of breaking immune tolerance to "self-antigens" and to evaluate the phage's antitumor activity. METHODS: A T4 phage nanometer particle expressing mFlt4 on the surface was constructed for evaluation as a recombinant vaccine. The presence of the mFlt4 gene in the T4-mFlt4 recombinant vaccine was verified by PCR and Western blot analysis. The immunotherapeutic potential of T4-mFlt4 was tested in mice injected with Lewis lung carcinoma (LLC) cells. Anti-Flt4 antibody producing B cells were detected by ELISPOT. The effects of T4-mFlt4 on lymphatic metastasis and lymphangiogenesis were investigated in a mouse antimetastasis assay and by Flt4 and CD105 immunohistochemistry. RESULTS: The T4-mFlt4 recombinant vaccine demonstrated antitumor activity and elicited autoantibodies against mFlt4. Mice carrying LLC-derived tumors exhibited prolonged survival when given the vaccine compared with control-treated animals. The vaccine also inhibited lymphangiogenesis and tumor metastasis in the mouse models. However, T4-mFlt4 was not observed to inhibit tumor growth. CONCLUSION: The T4-mFlt4 recombinant vaccine induced protective antitumor immunity and antimetastasis against LLC. Induction of an autoimmune response directed against tumor progression merits further study as a new strategy for immunotherapy in cancer.


Subject(s)
Bacteriophage T4/immunology , Cancer Vaccines/therapeutic use , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Nanoparticles , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , Vascular Endothelial Growth Factor Receptor-3/administration & dosage , Vascular Endothelial Growth Factor Receptor-3/therapeutic use , Animals , Autoantibodies/metabolism , B-Lymphocytes/metabolism , Lymphangiogenesis/immunology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/prevention & control , Vascular Endothelial Growth Factor Receptor-3/immunology
15.
Biotechnol Prog ; 25(2): 552-8, 2009.
Article in English | MEDLINE | ID: mdl-19334277

ABSTRACT

Salmonella typhimurium antigens were displayed on the capsid of a T2 bacteriophage to explore the potential of phage display for an oral vaccine. Segments of the flagellin proteins FliC (H1 antigen) and FljB (H2) were fused to the N-terminal of T2 phage SOC to give two recombinant phages, T2FliCm and T2FljBm. Over 14 days, 19 BALB/c mice were orally administered twice, either with purified recombinant FliCm and FljBm protein, or T2FliCm and T2FljBm with or without host Escherichia coli. Feces were sampled over 10 weeks and examined for phage by plaque assay and for the presence of mucosal IgA by ELISA. Relatively few phages were detected relative to the amount administered (up to 8.21 x 10(3) PFU/g faeces) and none were detected five days after initial administration. The administration of a large number of phages appeared to cause no clinical symptoms. IgA concentration in feces peaked around four weeks after the second administration and subsided after eight weeks. The highest relative titers were observed in the protein group (0.37% for anti-FliCm and 0.22% for anti-FljBm) and the mouse group which received no E. coli (0.33% and 0.35%) despite the theoretical amount of protein contained in a phage dose being at least 80-465 times lower than the protein dose administered. The possibility that the immuno-stimulatory properties of the phage create an adjuvant effect to enhance the immunogenic properties of the displayed proteins is discussed. We conclude that phage may be valuable as a vector for oral vaccines.


Subject(s)
Bacterial Proteins/immunology , Bacteriophage T4/genetics , Flagellin/immunology , Immunoglobulin A/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Administration, Oral , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacteriophage T4/immunology , Feces/chemistry , Female , Flagellin/administration & dosage , Flagellin/genetics , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Immunoglobulin A/analysis , Mice , Mice, Inbred BALB C , Salmonella Infections/microbiology , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/genetics , Salmonella typhimurium/genetics
16.
Vaccine ; 26(11): 1471-81, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18289743

ABSTRACT

An orally delivered foot-and-mouth disease (FMD) vaccine has not previously been reported. By using a T4 bacteriophage nanoparticle surface gene-protein display system (T4-S-GPDS), we created a foot-and-mouth disease virus (FMDV) entire capsid protein vaccine candidate. On the T4 phage surface SOC site, a full length FMDV capsid precursor polyprotein (P1, 755 aa) and proteinase 3C (213 aa) derived from an infected pig of serotype O strain GD-10 (1999), were separately displayed on different T4 phage particle surfaces through inserting their coding region DNAs into the T4 phage genome, yielding phage strains T4-P1 and T4-3C. We also constructed a series of FMDV sub-full length capsid structural protein (subunit) containing T4 phage recombinant vaccines. Both sucking and young BALB/c mice were used as two kinds of FMDV vaccine potency evaluation models. Many groups of both model mice were vaccinated orally or by subcutaneous injection with varying FMDV-T4 phage recombinant vaccines, with and without addition of adjuvant, then challenged with a lethal dose of cattle source virulent FMDV. In the case of immunization with a mixture of phage T4-P1 and phage T4-3C particles without any adjuvant added, all mice were 100% protected following either oral or injection immunization, whereas 100% of the control, non-immunized mice and mice immunized with only T4 phage vector Z1/Zh(-) or wild-type T4(+)D phage died; in contrast, with FMDV subunit vaccine, less than 75% protection followed the same potency challenge in both mice model groups. In addition, two pigs immunized with a phage T4-P1 and phage T4-3C mix were protected upon housing together with infected pigs. This study represents a clear example of how FMD and other pathogenic disease vaccines can be prepared by a simple and efficient bacteriophage route.


Subject(s)
Bacteriophage T4/immunology , Capsid/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Viral Vaccines/immunology , Animals , Animals, Newborn , DNA, Viral/biosynthesis , DNA, Viral/genetics , DNA, Viral/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Escherichia coli/ultrastructure , Escherichia coli/virology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/pathogenicity , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron , Peptide Library , Promoter Regions, Genetic/genetics , Protein Engineering , Serotyping , Swine , Vaccines, Synthetic/therapeutic use
17.
Immunol Lett ; 116(1): 24-32, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18166233

ABSTRACT

Bacteriophages in eukaryotic hosts may behave as particulate antigens able to activate the innate immune system and generate adaptive immunity. Dendritic cells (DCs) play a key role in the initiation of the immune response, mainly by priming T cell-mediated immunity. For this reason, they are increasingly applied as an adjuvant for effective anti-tumor therapies in animal models as well as in a few clinical trials. The presented study focused on the application of mouse DCs which were activated with T4 bacteriophages (T4 phages, T4) and further loaded with tumor antigens (TAg) in inducing an anti-tumor response. The activation of bone marrow-derived DCs with T4 phages and TAg resulted in augmentation of their differentiation marker expression accompanied by an enhanced ability to prime T cells for IFN-gamma production. These activated DCs (BM-DC/T4+TAg) were used in experimental immunotherapy of C57BL/6 mice bearing advanced MC38 colon carcinoma tumors. As a result of their triple application, a significant tumor growth delay, up to 19 days, was observed compared with the controls - treated with BM-DCs activated only with T4 phages, TAg, or lipopolysaccharide solution ["solvent"], where the tumor growth delay did not exceed 7 days. The percentage of tumor growth inhibition estimated 10 days after the third cell injection ranged from 32% (for animals treated with BM-DC/TAg cells) to 76% (for animals treated with BM-DC/T4+TAg cells) over the tumor-bearing untreated control mice. The obtained data indicate that in vitro interactions between T4 phages and BM-DCs followed by TAg activation caused augmentation of the anti-tumor effect when DCs were used as a vaccine for tumor-bearing mice treatment. Therefore, pretreatment of DCs with the phages may be considered as a beneficial element of a novel strategy in anti-tumor immunotherapy.


Subject(s)
Bacteriophage T4 , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/virology , Immunotherapy, Adoptive/methods , Animals , Antigen Presentation , Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Bacteriophage T4/immunology , Cell Line, Tumor , Colonic Neoplasms/therapy , Female , Immunity, Cellular , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Neoplasm Transplantation
18.
Virus Res ; 131(2): 233-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17996972

ABSTRACT

It has been known that administration of antibiotics may lead to excessive release of bacterial endotoxins and complicate clinical course of patients with Gram-negative infections. This concern may also apply to phages. Endotoxin may in turn activate neutrophils to produce reactive oxygen species (ROS) that are believed to play an important role in the pathogenesis of multiple organ dysfunction in the course of sepsis. We showed that a purified T4 phage preparation with low-endotoxin content could significantly diminish the luminol-dependent chemiluminescence (CL) of peripheral blood polymorphonuclear leukocytes (PMNs) both stimulated by lipopolysaccharides (LPSs) isolated from different Escherichia coli strains. This effect was also observed for live bacteria used for PMNs stimulation and was independent of bacterial susceptibility for T4-mediated lysis. Our data suggest, that phage-mediated inhibition of LPS- or bacteria-stimulated ROS production by PMNs may be attributed not only to phage-PMNs interactions, but also to phage-LPS interactions and bacterial lysis (in case of homologous phage). Interestingly, the T4 preparation did not influence ROS formation by PMNs stimulated with PMA. This suggests that the observed phenomena are also dependent upon the nature of activator. Bacteriophage-mediated inhibition of ROS formation by cells exposed to endotoxin provides new evidence for possible interactions between phages and mammalian cells. It helps in understanding the role of phages in our environment and may also be of important clinical significance.


Subject(s)
Bacteriophage T4/immunology , Neutrophils/immunology , Neutrophils/virology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Endotoxins/immunology , Escherichia coli/immunology , Humans , Luminescent Measurements , Luminol/metabolism , Neutrophil Activation
19.
Vaccine ; 25(7): 1225-35, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17069938

ABSTRACT

We describe a multicomponent antigen display and delivery system using bacteriophage T4. Two dispensable outer capsid proteins, Hoc (highly antigenic outer capsid protein, 155 copies) and Soc (small outer capsid protein, 810 copies), decorate phage T4 capsid. These proteins bind to the symmetrically localized capsid sites, which appear following prohead assembly and expansion. We hypothesized that multiple antigens fused to Hoc can be displayed on the same capsid and such particles can elicit broad immunological responses. Anthrax toxin proteins, protective antigen (PA), lethal factor (LF), and edema factor (EF), and their functional domains, were fused to Hoc with an N-terminal hexa-histidine tag and the recombinant proteins were over-expressed in E. coli and purified. Using a defined in vitro assembly system, the anthrax-Hoc fusion proteins were efficiently displayed on T4 capsid, either individually or in combinations. All of the 155 Hoc binding sites can be occupied by one antigen, or they can be split among two or more antigens by varying their molar ratio in the binding reaction. Immunization of mice with T4 phage carrying PA, LF, and EF elicited strong antigen-specific antibodies against all antigens as well as lethal toxin neutralization titers. The triple antigen T4 phage elicited stronger PA-specific immune responses than the phage displaying PA alone. These features offer novel avenues to develop customized multicomponent vaccines against anthrax and other pathogenic diseases.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Bacteriophage T4/genetics , Bacteriophage T4/immunology , Animals , Anthrax Vaccines/genetics , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/genetics , Bacterial Toxins/genetics , Binding Sites , Blotting, Western , Capsid/immunology , Capsid Proteins/immunology , Drug Delivery Systems , Female , Gene Dosage , Mice , Mice, Inbred CBA , Peptide Library , Plasmids/genetics , Plasmids/immunology , Protein Conformation , Protein Folding , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/immunology
20.
J Virol ; 80(15): 7688-98, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16840347

ABSTRACT

Bacteriophage T4 capsid is an elongated icosahedron decorated with 155 copies of Hoc, a nonessential highly antigenic outer capsid protein. One Hoc monomer is present in the center of each major capsid protein (gp23*) hexon. We describe an in vitro assembly system which allows display of HIV antigens, p24-gag, Nef, and an engineered gp41 C-peptide trimer, on phage T4 capsid surface through Hoc-capsid interactions. In-frame fusions were constructed by splicing the human immunodeficiency virus (HIV) genes to the 5' or 3' end of the Hoc gene. The Hoc fusion proteins were expressed, purified, and displayed on hoc(-) phage particles in a defined in vitro system. Single or multiple antigens were efficiently displayed, leading to saturation of all available capsid binding sites. The displayed p24 was highly immunogenic in mice in the absence of any external adjuvant, eliciting strong p24-specific antibodies, as well as Th1 and Th2 cellular responses with a bias toward the Th2 response. The phage T4 system offers new direction and insights for HIV vaccine development with the potential to increase the breadth of both cellular and humoral immune responses.


Subject(s)
AIDS Vaccines , Bacteriophage T4/metabolism , Capsid Proteins/metabolism , Capsid/metabolism , HIV Antigens/immunology , HIV Core Protein p24/immunology , HIV-1/immunology , Recombinant Fusion Proteins/metabolism , Animals , Bacteriophage T4/genetics , Bacteriophage T4/immunology , Capsid/immunology , Capsid Proteins/genetics , Female , HIV Antigens/genetics , HIV Antigens/metabolism , HIV-1/genetics , Humans , Mice , Mice, Inbred BALB C , Peptide Fragments/immunology , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...