Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Vaccine ; 35(1): 77-83, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27890399

ABSTRACT

Anaplasma marginale is a devastating tick-borne pathogen causing anaplasmosis in cattle and results in significant economic loss to the cattle industry worldwide. Currently, there is no widely accepted vaccine against A. marginale. New generation subunit vaccines against A. marginale, which are much safer, more efficient and cost-effective, are in great need. The A. marginale outer membrane protein VirB9-1 is a promising antigen for vaccination. We previously have shown that soluble recombinant VirB9-1 protein can be expressed and purified from Escherichia coli and induce a high level of humoral and cellular immunity in mice. In this study, we re-formulated the nanovaccines using the partially-purified VirB9-1 protein as the antigen and hollow nano-size silica vesicles (SV-100) as the adjuvant. We simplified the purification method to obtain the partially-purified antigen VirB9-1 with a six-fold higher yield. The new formulations using the partially-purified VirB9-1 protein achieved higher antibody and cell-mediated immune responses compared to the purified ones. This finding suggests that the partially-purified VirB9-1 protein performs better than the purified ones in the vaccination against A. marginale, and a certain level of contaminants in the protein antigen can be self-adjuvant and boost immunogenicity together with the nanoparticle adjuvant. This may lead to finding a "Goldilocks" level of contaminants. The new nanovaccine formulation using partially-purified antigens along with nanoparticle adjuvants offers an alternative strategy for making cheaper veterinary vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Anaplasma marginale/immunology , Anaplasmosis/prevention & control , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Cattle Diseases/prevention & control , Silicon Dioxide/administration & dosage , Animals , Bacterial Outer Membrane Proteins/isolation & purification , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/isolation & purification , Cattle , Female , Mice, Inbred C57BL , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification
2.
PLoS One ; 11(4): e0154295, 2016.
Article in English | MEDLINE | ID: mdl-27115492

ABSTRACT

Anaplasma marginale is the most prevalent tick-borne livestock pathogen and poses a significant threat to cattle industry. In contrast to currently available live blood-derived vaccines against A. marginale, alternative safer and better-defined subunit vaccines will be of great significance. Two proteins (VirB9-1 and VirB9-2) from the Type IV secretion system of A. marginale have been shown to induce humoral and cellular immunity. In this study, Escherichia coli were used to express VirB9-1 and VirB9-2 proteins. Silica vesicles having a thin wall of 6 nm and pore size of 5.8 nm were used as the carrier and adjuvant to deliver these two antigens both as individual or mixed nano-formulations. High loading capacity was achieved for both proteins, and the mouse immunisation trial with individual as well as mixed nano-formulations showed high levels of antibody titres over 107 and strong T-cell responses. The mixed nano-formulation also stimulated high-level recall responses in bovine T-cell proliferation assays. These results open a promising path towards the development of efficient A. marginale vaccines and provide better understanding on the role of silica vesicles to deliver multivalent vaccines as mixed nano-formulations able to activate both B-cell and T-cell immunity, for improved animal health.


Subject(s)
Anaplasma marginale/drug effects , Anaplasmosis/prevention & control , Antibodies, Bacterial/biosynthesis , Bacterial Outer Membrane Proteins/immunology , Cattle Diseases/prevention & control , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Anaplasma marginale/immunology , Anaplasmosis/immunology , Anaplasmosis/microbiology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Cattle , Cattle Diseases/immunology , Cattle Diseases/microbiology , Cell Proliferation/drug effects , Cloning, Molecular , Drug Carriers/chemistry , Drug Carriers/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Immunization , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
4.
PLoS One ; 10(12): e0143507, 2015.
Article in English | MEDLINE | ID: mdl-26630001

ABSTRACT

Bovine Viral Diarrhoea Virus (BVDV) is one of the most serious pathogen, which causes tremendous economic loss to the cattle industry worldwide, meriting the development of improved subunit vaccines. Structural glycoprotein E2 is reported to be a major immunogenic determinant of BVDV virion. We have developed a novel hollow silica vesicles (SV) based platform to administer BVDV-1 Escherichia coli-expressed optimised E2 (oE2) antigen as a nanovaccine formulation. The SV-140 vesicles (diameter 50 nm, wall thickness 6 nm, perforated by pores of entrance size 16 nm and total pore volume of 0.934 cm3 g(-1)) have proven to be ideal candidates to load oE2 antigen and generate immune response. The current study for the first time demonstrates the ability of freeze-dried (FD) as well as non-FD oE2/SV140 nanovaccine formulation to induce long-term balanced antibody and cell mediated memory responses for at least 6 months with a shortened dosing regimen of two doses in small animal model. The in vivo ability of oE2 (100 µg)/SV-140 (500 µg) and FD oE2 (100 µg)/SV-140 (500 µg) to induce long-term immunity was compared to immunisation with oE2 (100 µg) together with the conventional adjuvant Quil-A from the Quillaja saponira (10 µg) in mice. The oE2/SV-140 as well as the FD oE2/SV-140 nanovaccine generated oE2-specific antibody and cell mediated responses for up to six months post the final second immunisation. Significantly, the cell-mediated responses were consistently high in mice immunised with oE2/SV-140 (1,500 SFU/million cells) at the six-month time point. Histopathology studies showed no morphological changes at the site of injection or in the different organs harvested from the mice immunised with 500 µg SV-140 nanovaccine compared to the unimmunised control. The platform has the potential for developing single dose vaccines without the requirement of cold chain storage for veterinary and human applications.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/immunology , Diarrhea Virus 2, Bovine Viral/immunology , Immunity, Innate/immunology , Nanoparticles/administration & dosage , Silicon Dioxide/chemistry , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Adjuvants, Immunologic , Animals , Antibody Formation , Blotting, Western , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Cattle , Drug Carriers/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunization , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanoparticles/chemistry , Viral Vaccines/immunology
5.
PLoS One ; 10(11): e0141870, 2015.
Article in English | MEDLINE | ID: mdl-26535891

ABSTRACT

Bovine viral diarrhoea virus 1 (BVDV-1) is arguably the most important viral disease of cattle. It is associated with reproductive, respiratory and chronic diseases in cattle across the world. In this study we have investigated the capacity of the major immunological determinant of BVDV-1, the E2 protein combined with hollow type mesoporous silica nanoparticles with surface amino functionalisation (HMSA), to stimulate immune responses in sheep. The current work also investigated the immunogenicity of the E2 nanoformulation before and after freeze-drying processes. The optimal excipient formulation for freeze-drying of the E2 nanoformulation was determined to be 5% trehalose and 1% glycine. This excipient formulation preserved both the E2 protein integrity and HMSA particle structure. Sheep were immunised three times at three week intervals by subcutaneous injection with 500 µg E2 adsorbed to 6.2 mg HMSA as either a non-freeze-dried or freeze-dried nanoformulation. The capacity of both nanovaccine formulations to generate humoral (antibody) and cell-mediated responses in sheep were compared to the responses in sheep immunisation with Opti-E2 (500 µg) together with the conventional adjuvant Quil-A (1 mg), a saponin from the Molina tree (Quillaja saponira). The level of the antibody responses detected to both the non-freeze-dried and freeze-dried Opti-E2/HMSA nanoformulations were similar to those obtained for Opti-E2 plus Quil-A, demonstrating the E2 nanoformulations were immunogenic in a large animal, and freeze-drying did not affect the immunogenicity of the E2 antigen. Importantly, it was demonstrated that the long term cell-mediated immune responses were detectable up to four months after immunisation. The cell-mediated immune responses were consistently high in all sheep immunised with the freeze-dried Opti-E2/HMSA nanovaccine formulation (>2,290 SFU/million cells) compared to the non-freeze-dried nanovaccine formulation (213-500 SFU/million cells). This study is the first to demonstrate that a freeze-dried silica mesoporous nanovaccine formulation gives balanced immune responses in a production animal.


Subject(s)
Diarrhea Viruses, Bovine Viral/metabolism , Diarrhea/prevention & control , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Viral Envelope Proteins/immunology , Adjuvants, Immunologic , Adsorption , Animals , Antibody Formation/immunology , Cattle , Diarrhea/immunology , Diarrhea/veterinary , Diarrhea Viruses, Bovine Viral/immunology , Drug Compounding , Enzyme-Linked Immunospot Assay , Freeze Drying , Immunity, Cellular , Immunity, Humoral , Interferon-gamma/blood , Leukocytes, Mononuclear/metabolism , Nanoparticles/ultrastructure , Porosity , Quillaja Saponins/chemistry , Sheep , Viral Vaccines/immunology
6.
Biomaterials ; 35(37): 9972-9983, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25239045

ABSTRACT

Bovine Viral Diarrhoea Virus (BVDV) is widely distributed in cattle industries and causes significant economic losses worldwide annually. A limiting factor in the development of subunit vaccines for BVDV is the need to elicit both antibody and T-cell-mediated immunity as well as addressing the toxicity of adjuvants. In this study, we have prepared novel silica vesicles (SV) as the new generation antigen carriers and adjuvants. With small particle size of 50 nm, thin wall (~6 nm), large cavity (~40 nm) and large entrance size (5.9 nm for SV-100 and 16 nm for SV-140), the SV showed high loading capacity (∼ 250 µg/mg) and controlled release of codon-optimised E2 (oE2) protein, a major immunogenic determinant of BVDV. The in vivo functionality of the system was validated in mice immunisation trials comparing oE2 plus Quil A (50 µg of oE2 plus 10 µg of Quil A, a conventional adjuvant) to the oE2/SV-140 (50 µg of oE2 adsorbed to 250 µg of SV-140) or oE2/SV-140 together with 10 µg of Quil A. Compared to the oE2 plus Quil A, which generated BVDV specific antibody responses at a titre of 10(4), the oE2/SV-140 group induced a 10 times higher antibody response. In addition, the cell-mediated response, which is essential to recognise and eliminate the invading pathogens, was also found to be higher [1954-2628 spot forming units (SFU)/million cells] in mice immunised with oE2/SV-140 in comparison to oE2 plus Quil A (512-1369 SFU/million cells). Our study has demonstrated that SV can be used as the next-generation nanocarriers and adjuvants for enhanced veterinary vaccine delivery.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bovine Virus Diarrhea-Mucosal Disease/immunology , Cattle/virology , Diarrhea Virus 2, Bovine Viral/immunology , Drug Carriers/chemistry , Silicon Dioxide/chemistry , Viral Vaccines/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibody Formation , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Female , Immunity, Cellular , Immunization , Mice , Mice, Inbred C57BL , Quillaja Saponins , Saponins/administration & dosage , Saponins/immunology , T-Lymphocytes/immunology , Viral Vaccines/immunology
7.
Int J Pharm ; 465(1-2): 325-32, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24583208

ABSTRACT

Amino functionalised mesoporous silica nanoparticles (AM-41) have been identified as a promising vaccine delivery material. The capacity of AM-41 to stabilise vaccine components at ambient temperature (23-27°C) was determined by adsorbing the model antigen ovalbumin (OVA) to AM-41 particles (OVA-41). The OVA-41 was successfully freeze-dried using the excipients 5% trehalose and 1% PEG8000. The immunological activity of OVA and the nanoparticle structure were maintained following two months storage at ambient temperature. The results of immunisation studies in mice with reconstituted OVA-41 demonstrated the induction of humoral and cell-meditated immune responses. The capacity of AM-41 particles to facilitate ambient storage of vaccine components without the loss of immunological potency will underpin the further development of this promising vaccine delivery platform.


Subject(s)
Antigens/chemistry , Drug Carriers , Freeze Drying , Nanoparticles , Ovalbumin/chemistry , Silicon Dioxide/chemistry , Technology, Pharmaceutical/methods , Vaccines/chemistry , Animals , Antigens/administration & dosage , Antigens/immunology , Cattle , Cell Line , Cell Survival/drug effects , Chemistry, Pharmaceutical , Drug Stability , Drug Storage , Excipients/chemistry , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunization , Injections, Subcutaneous , Mice, Inbred C57BL , Nanotechnology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Polyethylene Glycols/chemistry , Porosity , Protein Stability , Silicon Dioxide/administration & dosage , Silicon Dioxide/toxicity , Temperature , Trehalose/chemistry , Vaccines/administration & dosage , Vaccines/immunology
8.
Nanoscale ; 5(12): 5167-79, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23657437

ABSTRACT

Vaccines have been at the forefront of improving human health for over two centuries. The challenges faced in developing effective vaccines flow from complexities associated with the immune system and requirement of an efficient and safe adjuvant to induce a strong adaptive immune response. Development of an efficient vaccine formulation requires careful selection of a potent antigen, efficient adjuvant and route of delivery. Adjuvants are immunological agents that activate the antigen presenting cells (APCs) and elicit a strong immune response. In the past decade, the use of mesoporous silica nanoparticles (MSNs) has gained significant attention as potential delivery vehicles for various biomolecules. In this review, we aim to highlight the potential of MSNs as vaccine delivery vehicles and their ability to act as adjuvants. We have provided an overview on the latest progress on synthesis, adsorption and release kinetics and biocompatibility of MSNs as next generation antigen carriers and adjuvants. A comprehensive summary on the ability of MSNs to deliver antigens and elicit both humoral and cellular immune responses is provided. Finally, we give insight on fundamental challenges and some future prospects of these nanoparticles as adjuvants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/administration & dosage , Drug Carriers/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , Antigen-Presenting Cells/metabolism , Cell Survival/drug effects , Humans , Nanoparticles/toxicity , Porosity
9.
Small ; 9(18): 3138-46, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23625779

ABSTRACT

Immunization to the model protein antigen ovalbumin (OVA) is investigated using MCM-41 mesoporous silica nanoparticles as a novel vaccine delivery vehicle and adjuvant system in mice. The effects of amino surface functionalization and adsorption time on OVA adsorption to nanoparticles are assessed. Amino-functionalized MCM-41 (AM-41) shows an effect on the amount of OVA binding, with 2.5-fold increase in binding capacity (72 mg OVA/g AM-41) compared to nonfunctionalized MCM-41 (29 mg OVA/g MCM-41). Immunization studies in mice with a 10 µg dose of OVA adsorbed to AM-41 elicits both antibody and cell-mediated immune responses following three subcutaneous injections. Immunizations at a lower 2 µg dose of OVA adsorbed to AM-41 particles results in an antibody response but not cell-mediated immunity. The level of antibody responses following immunization with nanoformulations containing either 2 µg or 10 µg of OVA are only slightly lower than that in mice which receive 50 µg OVA adjuvanted with QuilA, a crude mixture of saponins extracted from the bark of the Quillaja saponaria Molina tree. This is a significant result, since it demonstrates that AM-41 nanoparticles are self-adjuvanting and elicit immune responses at reduced antigen doses in vivo compared to a conventional delivery system. Importantly, there are no local or systemic negative effects in animals injected with AM-41. Histopathological studies of a range of tissue organs show no changes in histopathology of the animals receiving nanoparticles over a six week period. These results establish the biocompatible MCM-41 silica nanoparticles as a new method for vaccine delivery which incorporates a self-adjuvant effect.


Subject(s)
Adjuvants, Immunologic/chemistry , Nanoparticles/chemistry , Ovalbumin/chemistry , Silicon Dioxide/chemistry , Animals , Mice
10.
Microb Cell Fact ; 10: 57, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21787435

ABSTRACT

BACKGROUND: Protein expression in Escherichia coli may result in the recombinant protein being expressed as insoluble inclusion bodies. In addition, proteins purified from E. coli contain endotoxins which need to be removed for in vivo applications. The structural protein, E2, from Bovine Viral Diarrhoea Virus (BVDV) is a major immunogenic determinant, and is an ideal candidate as a subunit vaccine. The E2 protein contains 17 cysteine residues creating difficulties in E. coli expression. In this report we outline a procedure for successfully producing soluble and endotoxin-free BVDV E2 protein from inclusion bodies (IB). RESULTS: The expression of a truncated form of BVDV-E2 protein (E2-T1) in E. coli resulted in predominantly aggregated insoluble IB. Solubilisation of E2-T1 with high purity and stability from IB aggregates was achieved using a strong reducing buffer containing 100 mM Dithiothreitol. Refolding by dialysis into 50 mM Tris (pH 7.0) containing 0.2% Igepal CA630 resulted in a soluble but aggregated protein solution. The novel application of a two-phase extraction of inclusion body preparations with Triton X-114 reduced endotoxin in solubilised E2-T1 to levels suitable for in vivo use without affecting protein yields. Dynamic light scattering analyses showed 37.5% of the protein was monomeric, the remaining comprised of soluble aggregates. Mice immunised with E2-T1 developed a high titre antibody response by ELISA. Western hybridisation analysis showed E2-T1 was recognised by sera from immunised mice and also by several BVDV-E2 polyclonal and monoclonal antibodies. CONCLUSION: We have developed a procedure using E. coli to produce soluble E2-T1 protein from IB, and due to their insoluble nature we utilised a novel approach using Triton X-114 to efficiently remove endotoxin. The resultant protein is immunogenic and detectable by BVDV-E2 specific antibodies indicating its usefulness for diagnostic applications and as a subunit vaccine. The optimised E. coli expression system for E2-T1 combined with methodologies for solubilisation, refolding and integrated endotoxin removal presented in this study should prove useful for other vaccine applications.


Subject(s)
Diarrhea Viruses, Bovine Viral/metabolism , Endotoxins/isolation & purification , Inclusion Bodies/metabolism , Viral Envelope Proteins/biosynthesis , Animals , Antibodies/immunology , Antibodies/metabolism , Cattle , Cloning, Molecular , Escherichia coli/metabolism , Inclusion Bodies/genetics , Mice , Octoxynol , Polyethylene Glycols/chemistry , Protein Refolding , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Reducing Agents/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
11.
Planta ; 226(6): 1525-33, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17653759

ABSTRACT

Root-knot nematodes (Meloidogyne spp.) are obligate, sedentary endoparasites that infect many plant species causing large economic losses worldwide. Available nematicides are being banned due to their toxicity or ozone-depleting properties and alternative control strategies are urgently required. We have produced transgenic tobacco (Nicotiana tabacum) plants expressing different dsRNA hairpin structures targeting a root-knot nematode (Meloidogyne javanica) putative transcription factor, MjTis11. We provide evidence that MjTis11 was consistently silenced in nematodes feeding on the roots of transgenic plants. The observed silencing was specific for MjTis11, with other sequence-unrelated genes being unaffected in the nematodes. Those transgenic plants able to induce silencing of MjTis11, also showed the presence of small interfering RNAs. Even though down-regulation of MjTis11 did not result in a lethal phenotype, this study demonstrates the feasibility of silencing root-knot nematode genes by expressing dsRNA in the host plant. Host-delivered RNA interference-triggered (HD-RNAi) silencing of parasite genes provides a novel disease resistance strategy with wide biotechnological applications. The potential of HD-RNAi is not restricted to parasitic nematodes but could be adapted to control other plant-feeding pests.


Subject(s)
Nematoda/genetics , Nicotiana/genetics , RNA Interference , RNA, Double-Stranded/genetics , Animals , Blotting, Northern , Caenorhabditis elegans/classification , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Genes, Helminth/genetics , Host-Pathogen Interactions/genetics , Nematoda/growth & development , Plant Roots/genetics , Plant Roots/parasitology , Plants, Genetically Modified , RNA, Double-Stranded/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Nicotiana/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...