Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
J Immunol ; 206(4): 686-699, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33419770

ABSTRACT

East Coast fever (ECF), caused by Theileria parva, is the most important tick-borne disease of cattle in sub-Saharan Africa. Practical disadvantages associated with the currently used live-parasite vaccine could be overcome by subunit vaccines. An 80-aa polypeptide derived from the C-terminal portion of p67, a sporozoite surface Ag and target of neutralizing Abs, was the focus of the efforts on subunit vaccines against ECF and subjected to several vaccine trials with very promising results. However, the vaccination regimen was far from optimized, involving three inoculations of 450 µg of soluble p67C (s-p67C) Ag formulated in the Seppic adjuvant Montanide ISA 206 VG. Hence, an improved formulation of this polypeptide Ag is needed. In this study, we report on two nanotechnologies that enhance the bovine immune responses to p67C. Individually, HBcAg-p67C (chimeric hepatitis B core Ag virus-like particles displaying p67C) and silica vesicle (SV)-p67C (s-p67C adsorbed to SV-140-C18, octadecyl-modified SVs) adjuvanted with ISA 206 VG primed strong Ab and T cell responses to p67C in cattle, respectively. Coimmunization of cattle (Bos taurus) with HBcAg-p67C and SV-p67C resulted in stimulation of both high Ab titers and CD4 T cell response to p67C, leading to the highest subunit vaccine efficacy we have achieved to date with the p67C immunogen. These results offer the much-needed research depth on the innovative platforms for developing effective novel protein-based bovine vaccines to further the advancement.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Nanotechnology/methods , Protozoan Vaccines/immunology , Theileria parva/physiology , Theileriasis/immunology , Tick-Borne Diseases/immunology , Animals , Antibodies, Protozoan/blood , Cattle , Hepatitis B virus/chemistry , Hepatitis B virus/genetics , Mice , Mineral Oil/administration & dosage , Nanoparticles/chemistry , Protozoan Proteins/genetics , Protozoan Vaccines/genetics , RAW 264.7 Cells , Silicon Dioxide/chemistry , Ticks , Vaccination , Vaccines, Subunit , Viral Core Proteins/chemistry , Viral Core Proteins/genetics
2.
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
3.
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
4.
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
5.
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
SELECTION OF CITATIONS
SEARCH DETAIL