Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
2.
Methods Mol Biol ; 2225: 25-38, 2021.
Article in English | MEDLINE | ID: mdl-33108655

ABSTRACT

Various systems exist for the robust production of recombinant proteins. However, only a few systems are optimal for human vaccine protein production. Plant-based transient protein expression systems offer an advantageous alternative to costly mammalian cell culture-based systems and can perform posttranslational modifications due to the presence of an endomembrane system that is largely similar to that of the animal cell. Technological advances in expression vectors for transient expression in the last two decades have produced new plant expression systems with the flexibility and speed that cannot be matched by those based on mammalian or insect cell culture. The rapid and high-level protein production capability of transient expression systems makes them the optimal system to quickly and versatilely develop and produce vaccines against viruses such as 2019-nCoV that have sudden and unpredictable outbreaks. Here, expression of antiviral subunit vaccines in Nicotiana benthamiana plants via transient expression is demonstrated.


Subject(s)
Betacoronavirus/drug effects , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Plants/immunology , Pneumonia, Viral/prevention & control , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/biosynthesis , Betacoronavirus/immunology , Betacoronavirus/isolation & purification , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/transmission , Coronavirus Infections/virology , Genetic Vectors , Humans , Plants/genetics , Pneumonia, Viral/immunology , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , SARS-CoV-2
3.
Appl Microbiol Biotechnol ; 104(6): 2287-2296, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31980920

ABSTRACT

Infectious bursal disease is a widely spread threatening contagious viral infection of chickens that induces major damages to the Bursa of Fabricius and leads to severe immunosuppression in young birds causing significant economic losses for poultry farming. The etiological agent is the infectious bursal disease virus (IBDV), a non-enveloped virus belonging the family of Birnaviridae. At present, the treatment against the spread of this virus is represented by vaccination schedules mainly based on inactivated or live-attenuated viruses. However, these conventional vaccines present several drawbacks such as insufficient protection against very virulent strains and the impossibility to differentiate vaccinated animals from infected ones. To overcome these limitations, in the last years, several studies have explored the potentiality of recombinant subunit vaccines to provide an effective protection against IBDV infection. In this review, we will give an overview of these novel types of vaccines with special emphasis on current state-of-the-art in the use of plants as "biofactories" (plant molecular farming). In fact, plants have been thoroughly and successfully characterized as heterologous expression systems for the production of recombinant proteins for different applications showing several advantages compared with traditional expression systems (Escherichia coli, yeasts and insect cells) such as absence of animal pathogens in the production process, improved product quality and safety, reduction of manufacturing costs, and simplified scale-up.


Subject(s)
Birnaviridae Infections/veterinary , Infectious bursal disease virus/immunology , Plants, Genetically Modified , Vaccinology/methods , Viral Vaccines/immunology , Animals , Antibodies, Viral , Birnaviridae Infections/immunology , Birnaviridae Infections/prevention & control , Bursa of Fabricius/immunology , Bursa of Fabricius/virology , Chickens/immunology , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology , Viral Vaccines/biosynthesis
4.
Appl Microbiol Biotechnol ; 103(18): 7491-7504, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31332484

ABSTRACT

Infectious bursal disease virus (IBDV) is the cause of an economically important highly contagious disease of poultry, and vaccines are regarded as the most beneficial interventions for its prevention. In this study, plants were used to produce a recombinant chimeric IBDV antigen for the formulation of an innovative subunit vaccine. The fusion protein (PD-FcY) was designed to combine the immunodominant projection domain (PD) of the viral structural protein VP2 with the constant region of avian IgY (FcY), which was selected to enhance antigen uptake by avian immune cells. The gene construct encoding the fusion protein was transiently expressed in Nicotiana benthamiana plants and an extraction/purification protocol was set up, allowing to reduce the contamination by undesired plant compounds/proteins. Mass spectrometry analysis of the purified protein revealed that the glycosylation pattern of the FcY portion was similar to that observed in native IgY, while in vitro assays demonstrated the ability of PD-FcY to bind to the avian immunoglobulin receptor CHIR-AB1. Preliminary immunization studies proved that PD-FcY was able to induce the production of protective anti-IBDV-VP2 antibodies in chickens. In conclusion, the proposed fusion strategy holds promises for the development of innovative low-cost subunit vaccines for the prevention of avian viral diseases.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Immunoglobulins/immunology , Poultry Diseases/prevention & control , Viral Vaccines/biosynthesis , Animals , Antigens, Viral/biosynthesis , Chickens/immunology , Immunoglobulins/biosynthesis , Infectious bursal disease virus , Poultry Diseases/virology , Nicotiana/genetics , Vaccination , Vaccines, Subunit/biosynthesis , Viral Structural Proteins/biosynthesis , Viral Structural Proteins/immunology
5.
Biotechnol J ; 12(10)2017 Oct.
Article in English | MEDLINE | ID: mdl-28869356

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) are commonly present in the gastrointestinal tract of cattle and cause serious infectious disease in humans. Immunizing cattle against EHEC is a promising strategy to decrease the risk of food contamination; however, veterinary vaccines against EHEC such as Econiche have not been widely adopted by the agricultural industry, and have been discontinued, prompting the need for more cost-effective EHEC vaccines. The objective of this project is to develop a platform to produce plant-made antigens for oral vaccination of ruminants against EHEC. Five recombinant proteins were designed as vaccine candidates and expressed transiently in Nicotiana benthamiana and transplastomically in Nicotiana tabacum. Three of these EHEC proteins, NleA, Stx2b, and a fusion of EspA accumulated when transiently expressed. Transient protein accumulation was the highest when EHEC proteins were fused to an elastin-like polypeptide (ELP) tag. In the transplastomic lines, EspA accumulated up to 479 mg kg-1 in lyophilized leaf material. Sheep that were administered leaf tissue containing recombinant EspA shed less E. coli O157:H7 when challenged, as compared to control animals. These results suggest that plant-made, transgenic EspA has the potential to reduce EHEC shedding in ruminants.


Subject(s)
Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Vaccines/biosynthesis , Nicotiana/genetics , Plants, Genetically Modified/genetics , Ruminants/microbiology , Vaccines, Subunit/biosynthesis , Administration, Oral , Animals , Disease Models, Animal , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli O157/immunology , Escherichia coli Vaccines/administration & dosage , Escherichia coli Vaccines/genetics , Escherichia coli Vaccines/immunology , Feces/microbiology , Gene Expression Regulation, Plant , Immunization , Male , Plant Leaves/chemistry , Plants, Genetically Modified/metabolism , RNA, Messenger/biosynthesis , Recombinant Proteins , Sheep , Shiga Toxin 2/genetics , Nicotiana/chemistry , Vaccination , Virulence Factors/genetics
6.
Curr Opin Biotechnol ; 48: 42-53, 2017 12.
Article in English | MEDLINE | ID: mdl-28365472

ABSTRACT

There is an unmet demand for safe and efficient vaccines for prevention of various infectious diseases. Subunit vaccines comprise selected pathogen specific antigens are a safe alternative to whole organism vaccines. However they often lack immunogenicity. Natural and synthetic self-assembling polymers and proteins will be reviewed in view their use to encapsulate and/or display antigens to serve as immunogenic antigen carriers for induction of protective immunity. Recent advances made in in vivo assembly of antigen-displaying polyester inclusions will be a focus. Particulate vaccines are inherently immunogenic due to enhanced uptake by antigen presenting cells which process antigens mediating adaptive immune responses. Bioengineering approaches enable the design of tailor-made particulate vaccines to fine tune immune responses towards protective immunity.


Subject(s)
Bioengineering/trends , Polymers/metabolism , Vaccines, Subunit/biosynthesis , Vaccines/biosynthesis , Virion/immunology , Virion/metabolism , Animals , Antigens/immunology , Bioengineering/methods , Humans , Polymerization , Polymers/chemical synthesis , Vaccines/immunology , Vaccines, Subunit/metabolism
7.
Microb Pathog ; 105: 273-279, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259674

ABSTRACT

Streptococcus uberis is one of the most prevalent pathogens causing clinical and subclinical mastitis worldwide. Among bacterial factors involved in intramammary infections caused by this organism, S. uberis adhesion molecule (SUAM) is one of the main virulence factors identified. This molecule is involved in S. uberis internalization to mammary epithelial cells through lactoferrin (Lf) binding. The objective of this study was to evaluate SUAM properties as a potential subunit vaccine component for prevention of S. uberis mastitis. B epitope prediction analysis of SUAM sequence was used to identify potentially immunogenic regions. Since these regions were detected all along the gene, this criterion did not allow selecting a specific region as a potential immunogen. Hence, four fractions of SUAM (-1fr, 2fr, 3fr and 4fr), comprising most of the protein, were cloned and expressed. Every fraction elicited a humoral immune response in mice as predicted by bioinformatics analysis. SUAM-1fr generated antibodies with the highest recognition ability towards SUAM native protein. Moreover, antibodies against SUAM-1fr produced the highest proportion of internalization inhibition of S. uberis to mammary epithelial cells. In conclusion, SUAM immunogenic and functionally relevant regions were identified and allowed to propose SUAM-1fr as a potential candidate for a subunit vaccine for S. uberis mastitis prevention.


Subject(s)
Bacterial Adhesion/immunology , Bacterial Vaccines/immunology , Mastitis/prevention & control , Streptococcus/immunology , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Base Sequence , Cattle , DNA, Bacterial/genetics , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , Immunoglobulin G/blood , Lactoferrin/metabolism , Mice , Models, Animal , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Streptococcal Infections/microbiology , Streptococcus/genetics , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Virulence Factors/genetics
8.
Virus Res ; 232: 41-47, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28159612

ABSTRACT

Highly pathogenic avian influenza viruses cause severe disease and huge economic losses in domestic poultry and might pose a serious threat to people because of the high mortality rates in case of an accidental transmission to humans. The main goal of this work was to evaluate the immune responses and hemagglutination inhibition potential elicited by a combined DNA/recombinant protein prime/boost vaccination compared to DNA/DNA and protein/protein regimens in chickens. A plasmid encoding hemagglutinin (HA) from the A/swan/Poland/305-135V08/2006 (H5N1) virus, or the recombinant HA protein produced in Pichia pastoris system, both induced H5 HA-specific humoral immune responses in chickens. In two independent experiments, anti-HA antibodies were detected in sera collected two weeks after the first dose and the response was enhanced by the second dose of a vaccine, regardless of the type of subunit vaccine (DNA or recombinant protein) administered. The serum collected from chickens two weeks after the second dose was characterized by three types of assays: indirect ELISA, hemagglutination inhibition (HI) and a diagnostic test based on H5 antibody competition. Although the indirect ELISA failed to detect superiority of any of the three vaccine regimens, the other two tests clearly indicated that priming of chickens with the DNA vaccine significantly enhanced the protective potential of the recombinant protein vaccine produced in P. pastoris.


Subject(s)
Antibodies, Viral/biosynthesis , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Poultry Diseases/prevention & control , Vaccination/methods , Animals , Chickens/virology , Gene Expression , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization, Secondary , Immunogenicity, Vaccine , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/biosynthesis , Influenza Vaccines/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Pichia/genetics , Pichia/metabolism , Plasmids/chemistry , Plasmids/metabolism , Poultry Diseases/immunology , Poultry Diseases/virology , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/biosynthesis , Vaccines, DNA/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/genetics
9.
Virus Res ; 231: 148-165, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27932207

ABSTRACT

Human papillomavirus (HPV) is known to be a necessary factor for many gynecologic malignancies and is also associated with a subset of head and neck malignancies. This knowledge has created the opportunity to control these HPV-associated cancers through vaccination. However, despite the availability of prophylactic HPV vaccines, HPV infections remain extremely common worldwide. In addition, while prophylactic HPV vaccines have been effective in preventing infection, they are ineffective at clearing pre-existing HPV infections. Thus, there is an urgent need for therapeutic and T cell-based vaccines to treat existing HPV infections and HPV-associated lesions and cancers. Unlike prophylactic vaccines, which generate neutralizing antibodies, therapeutic, and T cell-based vaccines enhance cell-mediated immunity against HPV antigens. Our review will cover various therapeutic and T cell-based vaccines in development for the treatment of HPV-associated diseases. Furthermore, we review the strategies to enhance the efficacy of therapeutic vaccines and the latest clinical trials on therapeutic and T cell-based HPV vaccines.


Subject(s)
Dendritic Cells/transplantation , Head and Neck Neoplasms/prevention & control , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , T-Lymphocytes/transplantation , Uterine Cervical Neoplasms/prevention & control , Vaccination , Adoptive Transfer , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Clinical Trials as Topic , Dendritic Cells/immunology , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Humans , Immunity, Cellular/drug effects , Papillomaviridae/drug effects , Papillomaviridae/growth & development , Papillomaviridae/immunology , Papillomaviridae/pathogenicity , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/biosynthesis , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/biosynthesis , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology
10.
Virus Res ; 231: 166-175, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27889616

ABSTRACT

Infections with sexually transmitted high-risk Human Papillomavirus (hrHPV), of which there are at least 15 genotypes, are responsible for a tremendous disease burden by causing cervical, and subsets of other ano-genital and oro-pharyngeal carcinomas, together representing 5% of all cancer cases worldwide. HPV subunit vaccines consisting of virus-like particles (VLP) self-assembled from major capsid protein L1 plus adjuvant have been licensed. Prophylactic vaccinations with the 2-valent (HPV16/18), 4-valent (HPV6/11/16/18), or 9-valent (HPV6/11/16/18/31/33/45/52/58) vaccine induce high-titer neutralizing antibodies restricted to the vaccine types that cause up to 90% of cervical carcinomas, a subset of other ano-genital and oro-pharyngeal cancers and 90% of benign ano-genital warts (condylomata). The complexity of manufacturing multivalent L1-VLP vaccines limits the number of included VLP types and thus the vaccines' spectrum of protection, leaving a panel of oncogenic mucosal HPV unaddressed. In addition, current vaccines do not protect against cutaneous HPV types causing benign skin warts, or against beta-papillomavirus (betaPV) types implicated in the development of non-melanoma skin cancer (NMSC) in immunosuppressed patients. In contrast with L1-VLP, the minor capsid protein L2 contains type-common epitopes that induce low-titer yet broadly cross-neutralizing antibodies to heterologous PV types and provide cross-protection in animal challenge models. Efforts to increase the low immunogenicity of L2 (poly)-peptides and thereby to develop broader-spectrum HPV vaccines are the focus of this review.


Subject(s)
Antibodies, Viral/biosynthesis , Capsid Proteins/immunology , Head and Neck Neoplasms/prevention & control , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/prevention & control , Vaccination , Animals , Antibodies, Neutralizing/biosynthesis , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cross Protection , Epitopes/chemistry , Epitopes/immunology , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Humans , Immunity, Humoral/drug effects , Immunogenicity, Vaccine , Mice , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/genetics , Papillomaviridae/drug effects , Papillomaviridae/growth & development , Papillomaviridae/immunology , Papillomaviridae/pathogenicity , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology , Vaccines, Virus-Like Particle
11.
Mol Biol Rep ; 43(9): 911-22, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27251218

ABSTRACT

As an ancient disease, tuberculosis (TB) is a major global health threat. Therefore, there is an urgent need for an effective and safe anti-TB vaccine. In the current study, a delivery system of Fc domain of mouse IgG2a and early secreted antigenic target protein 6 (ESAT-6) was evaluated for the selective uptake of antigens by antigen-presenting cells (APCs). Thus, it was based on the immunogenicity of a fusion protein. The study was initiated by the transfer of recombinant expression vectors of pPICZαA-ESAT-6:Fcγ2a and pPICZαA-ESAT-6: His into Pichia pastoris (P. pastoris). Recombinant proteins were assessed for immunogenicity following the immunoblotting analysis. High levels of IFN-γ and IL-12 were produced to induce Th1-type cellular responses through vaccination with both recombinant proteins [ESAT-6:Fcγ2a (EF) and ESAT-6:His (EH)]. The Fc-tagged recombinant protein induced more effective Th1-type cellular responses with a low increment in IL-4 compared to PBS, BCG, and EH groups. Although in all the immunized groups, the ratio of IFN-γ/IL-4 was in favor of Th1 responses, the highest Th1/Th2 balance was observed in EF immunized group. Fc fragment of mouse IgG2a may induce a selective uptake of APCs towards the cross-presentation and formation of Th1 responses in favor of an appropriate protective anti-tuberculosis reaction. Thus, further research on Fc-fusion proteins is required to develop Fc-based TB vaccines.


Subject(s)
Immunoglobulin Fc Fragments/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccines, Subunit/immunology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/biosynthesis , Bacterial Proteins/immunology , Female , Immunity, Cellular , Interferon-gamma/blood , Interleukin-12/blood , Interleukin-4/blood , Mice, Inbred C57BL , Pichia , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/biosynthesis , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Vaccination , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/biosynthesis
12.
Nutr Hosp ; 32(5): 2274-9, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26545688

ABSTRACT

UNLABELLED: Backgound and aims: Dermatophagoides peteronyssinus is one of the important house dust mites responsible for allergic asthma that can be tentatively managed by specific immunotherapy. The present study was to construct a vector encoding T-cell epitopes of major allergen group 1 of Dermatophagoides pteronyssinus as a vaccine delivered by MHC class II pathway. METHODS: the nucleotide sequences of the 3 target genes were synthesized, including TAT, IhC and the recombinant fragment of Der p 1 encoding 3 T-cell epitopes. After amplification of the 3 target fragments by PCR and digestion with corresponding restriction endonucleases, the recombinant gene TAT-IhC-Der p 1-3T was ligated using T4 DNA ligase and inserted into the prokaryotic expression vector pET28a(+) to construct the recombinant plasmid pET- 28a(+)-TAT-IhC-Der p 1-3T, which was confirmed by digestion with restriction endonucleases and sequencing. The recombinant vector was transformed into E. coli strain BL21 (DE3) and induced with IPTG, and the induced protein TAT-IhC-Der p1-3T was detected by SDS-PAGE. After purification, the recombinant protein was confirmed by Western blotting and its allergenicity tested using IgE-binding assay. RESULTS: the recombinant plasmid pET-28a-TAT-IhCDer p1-3T was successfully constructed as confirmed by restriction endonuclease digestion and sequencing, and the expression of the recombinant protein TAT-IhC-Der p1-3T was induced in E. coli. Western blotting verified successfull purification of the target protein, which showed a stronger IgE-binding ability than Der p1. CONCLUSION: we successfully constructed the recombinant expression vector pET-28a-TAT-IhC-Der p1-3T expressing a T-cell epitope vaccine delivered by MHC II pathway with strong IgE-binding ability, which provides a basis for further study on specific immunotherapy via MHC class II pathway.


Antecedentes y objetivo: el Dermatophagoides peteronyssinus es uno de los principales ácaros del polvo doméstico responsables del asma alérgica que se pueden administrar provisionalmente para una inmunoterapia específica. El presente estudio busca construir un vector que codifique epítopos de células T del grupo de alérgenos principal, el Grupo 1 de Dermatophagoides pteronyssinus como una vacuna suministrada mediante la vía MHC de clase II. Métodos: se sintetizaron las secuencias de nucleótidos de los 3 genes objetivo, incluyendo TAT, IhC y el fragmento recombinante de Der p 1 encargado de codificar 3 epítopos de célula T. Después de la amplificación de los 3 fragmentos objetivo por PCR y digestión con endonucleasas de restricción correspondientes, el gen recombinante TAT-IhC-Der p 1-3T se ligó usando T4 DNA ligasa y se insertó en el vector de expresión procariota pET28a (+) para construir el plásmido recombinante pET 28a (+)-TAT-IHC-Der p 1-3T, que se confirmó por digestión con endonucleasas de restricción y secuenciación. El vector recombinante se transformó en E. coli cepa BL21 (DE3) y se indujo con IPTG, y la proteína inducida TATIHC- Der p1-3T se detectó mediante SDS-PAGE. Después de la purificación, la proteina recombinante se confirmó por análisis de inmunotransferencia (Western blot) y se probó su alergenicidad usando el ensayo de unión a IgE. Resultados: el plásmido recombinante pET-28a-TATIHCDer p1-3T se construyó con éxito, se confirmó por digestión con endonucleasas de restricción y la secuenciación y la expresión de la proteína recombinante TAT-IHCDer p1-3T fue inducida en E. coli. Purificación con éxito verificada mediante Western blot de la proteína objetivo, que mostró una capacidad de unión a IgE más fuerte que Der p1. Conclusión: hemos construido con éxito el vector de expresión recombinante pET-28a-TAT-IHC-Der p1-3T que expresa una vacuna de epítopo de células T administrada por vía MHC II con fuerte capacidad de union a IgE. Este trabajo proporciona una base para seguir estudiando la inmunoterapia específica mediante la vía MHC de clase II.


Subject(s)
Allergens/biosynthesis , Allergens/genetics , Antigens, Dermatophagoides/biosynthesis , Antigens, Dermatophagoides/genetics , Arthropod Proteins/biosynthesis , Arthropod Proteins/genetics , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Epitopes, T-Lymphocyte/genetics , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/genetics , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics , Animals , Cell Fusion , DNA Primers , Genes, MHC Class II/genetics , Genetic Vectors , Humans , Immunoglobulin E/chemistry , Plasmids/genetics
13.
Rev. argent. microbiol ; 47(1): 4-8, Mar. 2015. ilus, graf.
Article in English | LILACS, BINACIS | ID: biblio-1171812

ABSTRACT

Bovine viral diarrhea virus (BVDV) is an important cause of economic losses worldwide. E2 is an immunodominant protein and a promising candidate to develop subunit vaccines. To improve its immunogenicity, a truncated E2 (tE2) was fused to a single chain antibody named APCH, which targets to antigen-presenting cells. APCH-tE2 and tE2 proteins were expressed in the baculovirus system and their immunogenicity was firstly compared in guinea pigs. APCH-tE2 vaccine was the best one to evoke a humoral response, and for this reason, it was selected for a cattle vaccination experiment. All the bovines immunized with 1.5Ag of APCH-tE2 developed high levels of neutralizing antibodies against BVDV up to a year post-immunization, demonstrating its significant potential as a subunit vaccine. This novel vaccine is undergoing scale-up and was transferred to the private sector. Nowadays, it is being evaluated for registration as the first Argentinean subunit vaccine for cattle


El virus de la diarrea viral bovina (BVDV) es causante de importantes pérdidas económicas a nivel mundial. La proteína E2 es la inmunodominante del virus y es la candidata para desarrollar vacunas de subunidad. Para mejorar su inmunogenicidad, una versión truncada de la E2 (tE2) se fusionó a un anticuerpo de cadena simple (APCH), que se dirige a las células presentadoras de antígeno. Se expresaron las proteínas APCH-tE2 y tE2 en el sistema de baculovirus y su inmunogenicidad fue evaluada y comparada en cobayos; la proteína APCH-tE2 fue la que indujo la mejor respuesta humoral. Por dicha razón se la evaluó en bovinos utilizando 1,5µg de antígeno. Los animales presentaron altos títulos de anticuerpos neutralizantes contra BVDV hasta un año posinmunización. Esta nueva vacuna está en proceso de escalado y se transfirió al sector privado. Actualmente se está evaluando para su registro como la primera vacuna argentina de subunidad para bovinos


Subject(s)
Animals , Cattle , Guinea Pigs , Diarrhea Viruses, Bovine Viral/immunology , Vaccines, Subunit/biosynthesis , Antigen-Presenting Cells/drug effects , Baculoviridae/immunology , Immunization/veterinary , Adenovirus E2 Proteins/immunology , Diarrhea Viruses, Bovine Viral/drug effects , Antibodies, Neutralizing/analysis
14.
J Pharm Pharmacol ; 67(3): 319-28, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25556638

ABSTRACT

OBJECTIVES: Recombinant protein subunit vaccines are formulated using protein antigens that have been synthesized in heterologous host cells. Several host cells are available for this purpose, ranging from Escherichia coli to mammalian cell lines. This article highlights the benefits of using yeast as the recombinant host. KEY FINDINGS: The yeast species, Saccharomyces cerevisiae and Pichia pastoris, have been used to optimize the functional yields of potential antigens for the development of subunit vaccines against a wide range of diseases caused by bacteria and viruses. Saccharomyces cerevisiae has also been used in the manufacture of 11 approved vaccines against hepatitis B virus and one against human papillomavirus; in both cases, the recombinant protein forms highly immunogenic virus-like particles. SUMMARY: Advances in our understanding of how a yeast cell responds to the metabolic load of producing recombinant proteins will allow us to identify host strains that have improved yield properties and enable the synthesis of more challenging antigens that cannot be produced in other systems. Yeasts therefore have the potential to become important host organisms for the production of recombinant antigens that can be used in the manufacture of subunit vaccines or in new vaccine development.


Subject(s)
Antigens/biosynthesis , Bacterial Infections/prevention & control , Protein Biosynthesis , Recombinant Proteins/biosynthesis , Vaccines, Subunit/biosynthesis , Virus Diseases/prevention & control , Yeasts/metabolism , Humans , Pichia/metabolism , Saccharomyces cerevisiae/metabolism
15.
Sheng Wu Gong Cheng Xue Bao ; 30(6): 920-30, 2014 Jun.
Article in Chinese | MEDLINE | ID: mdl-25212009

ABSTRACT

Chloroplast-based expression system is promising for the hyper-expression of plant-derived recombinant therapeutic proteins and vaccines. To verify the feasibility of obtaining high-level expression of the SARS subunit vaccine and to provide a suitable plant-derived vaccine production platform against the severe acute respiratory syndrome coronavirus (SARS-CoV), a 193-amino acid fragment of SARS CoV spike protein receptor-binding domain (RBD), fused with the peptide vector cholera toxin B subunit (CTB), was expressed in tobacco chloroplasts. Codon-optimized CTB-RBD sequence was integrated into the chloroplast genome and homoplasmy was obtained, as confirmed by PCR and Southern blot analysis. Western blot showed expression of the recombinant fusion protein mostly in soluble monomeric form. Quantification of the recombinant fusion protein CTB-RBD was conducted by ELISA analysis from the transplastomic leaves at different developmental stages, attachment positions and time points in a day and the different expression levels of the CTB-RBD were observed with the highest expression of 10.2% total soluble protein obtained from mature transplastomic leaves. Taken together, our results demonstrate the feasibility of highly expressing SARS subunit vaccine RBD, indicating its potential in subsequent development of a plant-derived recombinant subunit vaccine and reagents production for antibody detection in SARS serological tests.


Subject(s)
Chloroplasts/metabolism , Nicotiana/metabolism , Spike Glycoprotein, Coronavirus/biosynthesis , Cholera Toxin , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/biosynthesis , Severe acute respiratory syndrome-related coronavirus , Vaccines, Subunit/biosynthesis
16.
PLoS One ; 9(1): e85401, 2014.
Article in English | MEDLINE | ID: mdl-24465551

ABSTRACT

Schistosomiasis is caused by several worm species of the genus Schistosoma and afflicts up to 600 million people in 74 tropical and sub-tropical countries in the developing world. Present disease control depends on treatment with the only available drug praziquantel. No vaccine exists despite the intense search for molecular candidates and adjuvant formulations over the last three decades. Cysteine peptidases such as papain and Der p 1 are well known environmental allergens that sensitize the immune system driving potent Th2-responses. Recently, we showed that the administration of active papain to mice induced significant protection (P<0.02, 50%) against an experimental challenge infection with Schistosoma mansoni. Since schistosomes express and secrete papain-like cysteine peptidases we reasoned that these could be employed as vaccines with inbuilt adjuvanticity to protect against these parasites. Here we demonstrate that sub-cutaneous injection of functionally active S. mansoni cathepsin B1 (SmCB1), or a cathepsin L from a related parasite Fasciola hepatica (FhCL1), elicits highly significant (P<0.0001) protection (up to 73%) against an experimental challenge worm infection. Protection and reduction in worm egg burden were further increased (up to 83%) when the cysteine peptidases were combined with other S. mansoni vaccine candidates, glyceraldehyde 3-phosphate dehydrogenase (SG3PDH) and peroxiredoxin (PRX-MAP), without the need to add chemical adjuvants. These studies demonstrate the capacity of helminth cysteine peptidases to behave simultaneously as immunogens and adjuvants, and offer an innovative approach towards developing schistosomiasis vaccines.


Subject(s)
Antigens, Helminth/immunology , Cathepsin B/immunology , Cathepsins/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/prevention & control , Vaccines, Subunit/administration & dosage , Adjuvants, Immunologic , Animals , Antibodies, Helminth/biosynthesis , Antigens, Helminth/chemistry , Cathepsin B/chemistry , Cathepsins/chemistry , Fasciola hepatica/chemistry , Fasciola hepatica/enzymology , Female , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/chemistry , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/immunology , Immunity, Active/drug effects , Mice , Peroxiredoxins/chemistry , Peroxiredoxins/immunology , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Schistosoma mansoni/chemistry , Schistosoma mansoni/enzymology , Schistosomiasis mansoni/immunology , Vaccination , Vaccines, Subunit/biosynthesis
17.
J Biotechnol ; 168(4): 710-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994481

ABSTRACT

The twin-arginine translocation (Tat) system is a major pathway for transmembrane translocation of fully folded proteins. In this study, a multivalent vaccine to present foreign antigens on live attenuated vaccine Edwardsiella tarda WED using screened Tat signal peptide was constructed. Because the Tat system increases the yields of folded antigens in periplasmic space or extracellular milieu, it is expected to contribute to the production of conformational epitope-derived specific antibodies. E. tarda Tat signal peptides fused with the green fluorescent protein (GFP) was constructed under the control of an in vivo inducible dps promoter. The resulting plasmids were electroporated into WED and the subcellular localizations of GFP were analyzed with Western blotting. Eight signal peptides with optimized GFP translocation efficiency were further fused to a protective antigen glyceraldehyde-3-phosphate dehydrogenase (GapA) from a fish pathogen Aeromonas hydrophila. Signal peptides of DmsA, NapA, and SufI displayed high efficiency for GapA translocation. The relative percent survival (RPS) of turbot was measured with a co-infection of E. tarda and A. hydrophila, and the strain with DmsA signal peptide showed the maximal protection. This study demonstrated a new platform to construct multivalent vaccines using optimized Tat signal peptide in E. tarda.


Subject(s)
Antigen Presentation/genetics , Escherichia coli Proteins/immunology , Fishes/microbiology , Membrane Transport Proteins/immunology , Vaccines, Subunit/genetics , Aeromonas hydrophila/enzymology , Animals , Edwardsiella tarda/genetics , Edwardsiella tarda/metabolism , Enterobacteriaceae Infections/prevention & control , Escherichia coli Proteins/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/immunology , Membrane Transport Proteins/genetics , Promoter Regions, Genetic , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology
18.
Vaccine ; 31(23): 2623-7, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23583894

ABSTRACT

Infectious Bursal Disease Virus (IBDV) is the etiological agent of an immunosuppressive and highly contagious disease that affects young birds. This disease causes important economic losses in the poultry industry worldwide. The VP2 protein has been used for the development of subunit vaccines in a variety of heterologous platforms. In this context, the aim of this study was to investigate VP2 expression and immunogenicity using an experimental plant-based vaccine against IBDV. We determined that the agroinfiltration of N. benthamiana leaves allowed the production of VP2 with no apparent change on its conformational epitopes. Chickens intramuscularly immunized in a dose/boost scheme with crude concentrated extracts developed a specific humoral response with viral neutralizing ability. Given these results, it seems plausible for a plant-based vaccine to have a niche in the veterinary field. Thus, plants can be an adequate system of choice to produce immunogens against IBDV.


Subject(s)
Birnaviridae Infections/veterinary , Infectious bursal disease virus/immunology , Nicotiana/microbiology , Poultry Diseases/prevention & control , Viral Structural Proteins/biosynthesis , Viral Structural Proteins/immunology , Viral Vaccines/biosynthesis , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Birnaviridae Infections/immunology , Birnaviridae Infections/prevention & control , Chick Embryo , Chickens , Infectious bursal disease virus/genetics , Poultry Diseases/immunology , Poultry Diseases/virology , T-Lymphocytes/immunology , Nicotiana/genetics , Nicotiana/immunology , Nicotiana/metabolism , Vaccination/veterinary , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/immunology , Viral Structural Proteins/genetics , Viral Vaccines/immunology
19.
Curr Pharm Des ; 19(31): 5574-86, 2013.
Article in English | MEDLINE | ID: mdl-23394571

ABSTRACT

Plants have a demonstrated potential for large-scale, rapid production of recombinant proteins for diverse product applications, including subunit vaccines and monoclonal antibodies. In this field, the accent has recently shifted from the engineering of "edible" vaccines based on stable expression of target protein in transgenic or transplastomic plants to the development of purified formulated vaccines that are delivered via injection. The injectable vaccines are commonly produced using transient expression of target gene delivered into genetically unmodified plant host via viral or bacterial vectors. Most viral vectors are based on plant RNA viruses, where nonessential sequences are replaced with the gene of interest. Utilization of viral hybrids that consist of genes and regulatory elements of different virus species, or transcomplementation systems (vector/transgene) had a substantial impact on the level of target protein expression. Development and introduction of agroviral hybrid vectors that combine genetic elements of bacterial binary plasmids and plant viral vectors, and agroinfiltration as a tool of the vector delivery have resulted in significant progress in large-scale production of recombinant vaccines and monoclonal antibodies in plants. This article presents an overview of plant hybrid viral vector expression systems developed so far.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Genetic Vectors , Recombinant Proteins/biosynthesis , Animals , Humans , Plant Proteins/metabolism , Plant Viruses/genetics , Vaccines, Subunit/biosynthesis , Vaccines, Synthetic/biosynthesis
20.
Vaccine ; 30(43): 6121-6, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22885016

ABSTRACT

Two currently licensed live oral rotavirus vaccines (Rotarix® and RotaTeq®) are highly efficacious against severe rotavirus diarrhea. However, the efficacy of such vaccines in selected low-income African and Asian countries is much lower than that in middle or high-income countries. Additionally, these two vaccines have recently been associated with rare case of intussusception in vaccinated infants. We developed a novel recombinant subunit parenteral rotavirus vaccine which may be more effective in low-income countries and also avert the potential problem of intussusception. Truncated recombinant VP8* (ΔVP8*) protein of human rotavirus strain Wa P[8], DS-1 P[4] or 1076 P[6] expressed in Escherichia coli was highly soluble and was generated in high yield. Guinea pigs hyperimmunized intramuscularly with each of the ΔVP8* proteins (i.e., P[8], P[4] or P[6]) developed high levels of homotypic as well as variable levels of heterotypic neutralizing antibodies. Moreover, the selected ΔVP8* proteins when administered to mice at a clinically relevant dosage, route and schedule, elicited high levels of serum anti-VP8* IgG and/or neutralizing antibodies. Our data indicated that the ΔVP8* proteins may be a plausible additional candidate as new parenteral rotavirus vaccines.


Subject(s)
RNA-Binding Proteins/immunology , Rotavirus Infections/prevention & control , Rotavirus Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chlorocebus aethiops , Escherichia coli , Female , Guinea Pigs , Immunoglobulin G/blood , Mice , Plasmids , Recombinant Proteins/immunology , Rotavirus/immunology , Rotavirus Vaccines/biosynthesis , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/immunology , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...