Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Front Vet Sci ; 10: 1208275, 2023.
Article in English | MEDLINE | ID: mdl-37404778

ABSTRACT

Introduction: African swine fever virus (ASFV) is a pathogen of great economic importance given that continues to threaten the pork industry worldwide, but there is no safe vaccine or treatment available. Development of a vaccine is feasible as immunization of pigs with some live attenuated ASFV vaccine candidates can confer protection, but safety concerns and virus scalability are challenges that must to be addressed. Identification of protective ASFV antigens is needed to inform the development of efficacious subunit vaccines. Methods: In this study, replication-incompetent adenovirus-vectored multicistronic ASFV antigen expression constructs that covered nearly 100% of the ASFV proteome were generated and validated using ASFV convalescent serum. Swine were immunized with a cocktail of the expression constructs, designated Ad5-ASFV, alone or formulated with either Montanide ISA-201™ (ASFV-ISA-201) or BioMize® adjuvant (ASFV-BioMize). Results: These constructs primed strong B cell responses as judged by anti-pp62-specific IgG responses. Notably, the Ad5-ASFV and the Ad5-ASFV ISA-201, but not the Ad5-ASFV BioMize®, immunogens primed significantly (p < 0.0001) higher anti-pp62-specific IgG responses compared with Ad5-Luciferase formulated with Montanide ISA-201™ adjuvant (Luc-ISA-201). The anti-pp62-specific IgG responses underwent significant (p < 0.0001) recall in all the vaccinees after boosting and the induced antibodies strongly recognized ASFV (Georgia 2007/1)-infected primary swine cells. However, following challenge by contact spreaders, only one pig nearly immunized with the Ad5-ASFV cocktail survived. The survivor had no typical clinical symptoms, but had viral loads and lesions consistent with chronic ASF. Discussion: Besides the limited sample size used, the outcome suggests that in vivo antigen expression, but not the antigen content, might be the limitation of this immunization approach as the replication-incompetent adenovirus does not amplify in vivo to effectively prime and expand protective immunity or directly mimic the gene transcription mechanisms of attenuated ASFV. Addressing the in vivo antigen delivery limitations may yield promising outcomes.

2.
Front Vet Sci ; 9: 921481, 2022.
Article in English | MEDLINE | ID: mdl-35711803

ABSTRACT

African Swine Fever Virus (ASFV) poses a serious threat to the pork industry worldwide; however, there is no safe vaccine or treatment available. The development of an efficacious subunit vaccine will require the identification of protective antigens. The ASFV pp220 polyprotein is essential for virus structural integrity. This polyprotein is processed to generate p5, p34, p14, p37, and p150 individual proteins. Immunization of pigs with a cocktail of adenoviruses expressing the proteins induced significant IgG, IFN-γ-secreting cells, and cytotoxic T lymphocyte responses. Four predicted SLA-I binding nonamer peptides, namely p34161-169, p37859-867, p1501363-1371, and p1501463-1471, recalled strong IFN-γ+ PBMC and splenocyte responses. Notably, peptide p34161-169 was recognized by PBMCs isolated from 7/10 pigs and by splenocytes isolated from 8/10 pigs. Peptides p37859-867 and p1501363-1371 stimulated recall IFN-γ+ responses in PBMCs and splenocytes isolated from 8/10 pigs, whereas peptide p1501463-1471 recalled responses in PBMCs and splenocytes isolated from 7/10 to 9/10 pigs, respectively. The results demonstrate that the pp220 polyprotein contains multiple epitopes that induce robust immune responses in pigs. Importantly, these epitopes are 100% conserved among different ASFV genotypes and were predicted to bind multiple SLA-I alleles. The outcomes suggest that pp220 is a promising candidate for inclusion in a prototype subunit vaccine.

3.
J Immunol ; 206(8): 1709-1718, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33762324

ABSTRACT

Studies of immune responses elicited by bovine viral diarrhea virus (BVDV) vaccines have primarily focused on the characterization of neutralizing B cell and CD4+ T cell epitopes. Despite the availability of commercial vaccines for decades, BVDV prevalence in cattle has remained largely unaffected. There is limited knowledge regarding the role of BVDV-specific CD8+ T cells in immune protection, and indirect evidence suggests that they play a crucial role during BVDV infection. In this study, the presence of BVDV-specific CD8+ T cells that are highly cross-reactive in cattle was demonstrated. Most importantly, novel potent IFN-γ-inducing CD8+ T cell epitopes were identified from different regions of BVDV polyprotein. Eight CD8+ T cell epitopes were identified from the following structural BVDV Ags: Erns, E1, and E2 glycoproteins. In addition, from nonstructural BVDV Ags Npro, NS2-3, NS4A-B, and NS5A-B, 20 CD8+ T cell epitopes were identified. The majority of these IFN-γ-inducing CD8+ T cell epitopes were found to be highly conserved among more than 200 strains from BVDV-1 and -2 genotypes. These conserved epitopes were also validated as cross-reactive because they induced high recall IFN-γ+CD8+ T cell responses ex vivo in purified bovine CD8+ T cells isolated from BVDV-1- and -2-immunized cattle. Altogether, 28 bovine MHC class I-binding epitopes were identified from key BVDV Ags that can elicit broadly reactive CD8+ T cells against diverse BVDV strains. The data presented in this study will lay the groundwork for the development of a contemporary CD8+ T cell-based BVDV vaccine capable of addressing BVDV heterogeneity more effectively than current vaccines.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/immunology , CD8-Positive T-Lymphocytes/immunology , Diarrhea Viruses, Bovine Viral/physiology , Epitopes, T-Lymphocyte/metabolism , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Viral Vaccines/immunology , Animals , Cattle , Cells, Cultured , Conserved Sequence/genetics , Cross Reactions , Epitopes, T-Lymphocyte/genetics , Histocompatibility Antigens Class I/metabolism , Interferon-gamma/metabolism , Protein Binding , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/genetics
4.
Front Immunol ; 11: 589537, 2020.
Article in English | MEDLINE | ID: mdl-33281819

ABSTRACT

Bovine Viral Diarrhea Virus (BVDV) is an important pathogen that plays a significant role in initiating Bovine Respiratory Disease Complex (BRDC) in cattle. The disease causes multi-billion dollar losses globally due to high calf mortality and increased morbidity leading to heavy use of antibiotics. Current commercial vaccines provide limited cross-protection with several drawbacks such as safety, immunosuppression, potential reversion to virulence, and induction of neonatal pancytopenia. This study evaluates two prototype vaccines containing multiple rationally designed recombinant mosaic BVDV antigens for their potential to confer cross-protection against diverse BVDV strains. Genes encoding three novel mosaic antigens, designated E2123, NS2-31, and NS2-32, were designed in silico and expressed in mammalian cells for the formulation of a prototype protein-based vaccine. The mosaic antigens contain highly conserved protective epitopes from BVDV-1a, -1b, and -2, and included unique neutralizing epitopes from disparate strains to broaden coverage. We tested immunogenicity and protective efficacy of Expi293TM-expressed mosaic antigens (293F-E2123, 293F-NS2-31, and 293F-NS2-32), and baculovirus-expressed E2123 (Bac-E2123) mosaic antigen in calves. The Expi293TM-expressed antigen cocktail induced robust BVDV-specific cross-reactive IFN-γ responses, broadly neutralizing antibodies, and following challenge with a BVDV-1b strain, the calves had significantly (p < 0.05) reduced viremia and clinical BVD disease compared to the calves vaccinated with a commercial killed vaccine. The Bac-E2123 antigen was not as effective as the Expi293TM-expressed antigen cocktail, but it protected calves from BVD disease better than the commercial killed vaccine. The findings support feasibility for development of a broadly protective subunit BVDV vaccine for safe and effective management of BRD.


Subject(s)
Antigens, Viral/immunology , Bovine Virus Diarrhea-Mucosal Disease/therapy , Cattle/immunology , Diarrhea Viruses, Bovine Viral/immunology , Viral Vaccines/administration & dosage , Animals , Antigens, Viral/genetics , Bovine Virus Diarrhea-Mucosal Disease/immunology , Bovine Virus Diarrhea-Mucosal Disease/virology , Epitopes/immunology
5.
Front Vet Sci ; 7: 84, 2020.
Article in English | MEDLINE | ID: mdl-32154279

ABSTRACT

African swine fever is a major concern due to its negative impact on pork production in affected regions. Due to lack of treatment and a safe vaccine, it has been extremely difficult to control this devastating disease. The mechanisms of virus entry, replication within the host cells, immune evasion mechanisms, correlates of protection, and antigens that are effective at inducing host immune response, are now gradually being identified. This information is required for rational design of novel disease control strategies. Pigs which recover from infection with less virulent ASFV isolates can be protected from challenge with related virulent isolates. This strongly indicates that an effective vaccine against ASFV could be developed. Nonetheless, it is clear that effective immunity depends on both antibody and cellular immune responses. This review paper summarizes the key studies that have evaluated three major approaches for development of African Swine Fever virus vaccines. Recent immunization strategies have involved development and in vivo evaluation of live attenuated virus, and recombinant protein- and DNA-based and virus-vectored subunit vaccine candidates. The limitations of challenge models for evaluating ASFV vaccine candidates are also discussed.

6.
Vet Microbiol ; 235: 10-20, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31282366

ABSTRACT

African Swine Fever Virus (ASFV) causes a hemorrhagic disease in swine and wild boars with a fatality rate close to 100%. Less virulent strains cause subchronic or chronic forms of the disease. The virus is endemic in sub-Saharan Africa and an outbreak in Georgia in 2007 spread to Armenia, Russia, Ukraine, Belarus, Poland, Lithuania, and Latvia. In August 2018, there was an outbreak in China and in April 2019, ASFV was reported in Vietnam and Cambodia. Since no vaccine or treatment exists, a vaccine is needed to safeguard the swine industry. Previously, we evaluated immunogenicity of two adenovirus-vectored cocktails containing ASFV antigens and demonstrated induction of unprecedented robust antibody and T cell responses, including cytotoxic T lymphocytes. In the present study, we evaluated protective efficacy of both cocktails by intranasal challenge of pigs with ASFV-Georgia 2007/1. A nine antigen cocktail-(I) formulated in BioMize adjuvant induced strong IgG responses, but when challenged, the vaccinees had more severe reaction relative to the controls. A seven antigen cocktail-(II) was evaluated using two adjuvants: BioMize and ZTS-01. The BioMize formulation induced stronger antibody responses, but 8/10 vaccinees and 4/5 controls succumbed to the disease or reached experimental endpoint at 17 days post-challenge. In contrast, the ZTS-01 formulation induced weaker antibody responses, but 4/9 pigs succumbed to the disease while the 5 survivors exhibited low clinical scores and no viremia at 17 days post-challenge, whereas 4/5 controls succumbed to the disease or reached experimental endpoint. Overall, none of the immunogens conferred statistically significant protection.


Subject(s)
African Swine Fever/prevention & control , Antibodies, Viral/blood , Antigens, Viral/immunology , Viral Vaccines/immunology , Adenoviridae , Administration, Intranasal , African Swine Fever/immunology , African Swine Fever Virus , Animals , Antigens, Viral/genetics , Immunoglobulin G/blood , Swine , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/genetics , Viremia , Virulence
7.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31085705

ABSTRACT

Lyme disease (LD), the most prevalent vector-borne illness in the United States and Europe, is caused by Borreliella burgdorferi No vaccine is available for humans. Dogmatically, B. burgdorferi can establish a persistent infection in the mammalian host (e.g., mice) due to a surface antigen, VlsE. This antigenically variable protein allows the spirochete to continually evade borreliacidal antibodies. However, our recent study has shown that the B. burgdorferi spirochete is effectively cleared by anti-B. burgdorferi antibodies of New Zealand White rabbits, despite the surface expression of VlsE. Besides homologous protection, the rabbit antibodies also cross-protect against heterologous B. burgdorferi spirochetes and significantly reduce the pathology of LD arthritis in persistently infected mice. Thus, this finding that NZW rabbits develop a unique repertoire of very potent antibodies targeting the protective surface epitopes, despite abundant VlsE, prompted us to identify the specificities of the protective rabbit antibodies and their respective targets. By applying subtractive reverse vaccinology, which involved the use of random peptide phage display libraries coupled with next-generation sequencing and our computational algorithms, repertoires of nonprotective (early) and protective (late) rabbit antibodies were identified and directly compared. Consequently, putative surface epitopes that are unique to the protective rabbit sera were mapped. Importantly, the relevance of newly identified protection-associated epitopes for their surface exposure has been strongly supported by prior empirical studies. This study is significant because it now allows us to systematically test the putative epitopes for their protective efficacy with an ultimate goal of selecting the most efficacious targets for development of a long-awaited LD vaccine.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Vaccines/immunology , Borrelia burgdorferi/immunology , Epitopes , Animals , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Lipoproteins/immunology , Male , Mice , Mice, Inbred C3H , Rabbits , Vaccines, Subunit/immunology
8.
Infect Immun ; 87(7)2019 07.
Article in English | MEDLINE | ID: mdl-30988058

ABSTRACT

Borrelia burgdorferi is a tick-borne bacterium responsible for approximately 300,000 annual cases of Lyme disease (LD) in the United States, with increasing incidences in other parts of the world. The debilitating nature of LD is mainly attributed to the ability of B. burgdorferi to persist in patients for many years despite strong anti-Borrelia antibody responses. Antimicrobial treatment of persistent infection is challenging. Similar to infection of humans, B. burgdorferi establishes long-term infection in various experimental animal models except for New Zealand White (NZW) rabbits, which clear the spirochete within 4 to 12 weeks. LD spirochetes have a highly evolved antigenic variation vls system, on the lp28-1 plasmid, where gene conversion results in surface expression of the antigenically variable VlsE protein. VlsE is required for B. burgdorferi to establish persistent infection by continually evading otherwise potent antibodies. Since the clearance of B. burgdorferi is mediated by humoral immunity in NZW rabbits, the previously reported results that LD spirochetes lose lp28-1 during rabbit infection could potentially explain the failure of B. burgdorferi to persist. However, the present study unequivocally disproves that previous finding by demonstrating that LD spirochetes retain the vls system. However, despite the vls system being fully functional, the spirochete fails to evade anti-Borrelia antibodies of NZW rabbits. In addition to being protective against homologous and heterologous challenges, the rabbit antibodies significantly ameliorate LD-induced arthritis in persistently infected mice. Overall, the current data indicate that NZW rabbits develop a protective antibody repertoire, whose specificities, once defined, will identify potential candidates for a much-anticipated LD vaccine.


Subject(s)
Antigenic Variation/physiology , Antigens, Bacterial/immunology , Borrelia burgdorferi/genetics , Lyme Disease/immunology , Lyme Disease/microbiology , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/genetics , Lipoproteins/genetics , Plasmids , Rabbits
9.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29866906

ABSTRACT

The tick-borne pathogen Borrelia burgdorferi is responsible for approximately 300,000 Lyme disease (LD) cases per year in the United States. Recent increases in the number of LD cases, in addition to the spread of the tick vector and a lack of a vaccine, highlight an urgent need for designing and developing an efficacious LD vaccine. Identification of protective epitopes that could be used to develop a second-generation (subunit) vaccine is therefore imperative. Despite the antigenicity of several lipoproteins and integral outer membrane proteins (OMPs) on the B. burgdorferi surface, the spirochetes successfully evade antibodies primarily due to the VlsE-mediated antigenic variation. VlsE is thought to sterically block antibody access to protective epitopes of B. burgdorferi However, it is highly unlikely that VlsE shields the entire surface epitome. Thus, identification of subdominant epitope targets that induce protection when they are made dominant is necessary to generate an efficacious vaccine. Toward the identification, we repeatedly immunized immunocompetent mice with live-attenuated VlsE-deleted B. burgdorferi and then challenged the animals with the VlsE-expressing (host-adapted) wild type. Passive immunization and Western blotting data suggested that the protection of 50% of repeatedly immunized animals against the highly immune-evasive B. burgdorferi was antibody mediated. Comparison of serum antibody repertoires identified in protected and nonprotected animals permitted the identification of several putative epitopes significantly associated with the protection. Most linear putative epitopes were conserved between the main pathogenic Borrelia genospecies and found within known subdominant regions of OMPs. Currently, we are performing immunization studies to test whether the identified protection-associated epitopes are protective for mice.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Vaccines/immunology , Borrelia burgdorferi/immunology , Epitopes/immunology , Lipoproteins/metabolism , Lyme Disease/immunology , Animals , Bacterial Vaccines/administration & dosage , Blotting, Western , Disease Models, Animal , Epitope Mapping , Immunization, Passive , Lipoproteins/deficiency , Lyme Disease/prevention & control , Male , Mice , Mice, Inbred C3H , Mice, SCID , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
10.
PLoS One ; 12(5): e0177007, 2017.
Article in English | MEDLINE | ID: mdl-28481911

ABSTRACT

African Swine Fever Virus (ASFV) is a high-consequence transboundary animal pathogen that often causes hemorrhagic disease in swine with a case fatality rate close to 100%. Lack of treatment or vaccine for the disease makes it imperative that safe and efficacious vaccines are developed to safeguard the swine industry. In this study, we evaluated the immunogenicity of seven adenovirus-vectored novel ASFV antigens, namely A151R, B119L, B602L, EP402RΔPRR, B438L, K205R and A104R. Immunization of commercial swine with a cocktail of the recombinant adenoviruses formulated in adjuvant primed strong ASFV antigen-specific IgG responses that underwent rapid recall upon boost. Notably, most vaccinees mounted robust IgG responses against all the antigens in the cocktail. Most importantly and relevant to vaccine development, the induced antibodies recognized viral proteins from Georgia 2007/1 ASFV-infected cells by IFA and by western blot analysis. The recombinant adenovirus cocktail also induced ASFV-specific IFN-γ-secreting cells that were recalled upon boosting. Evaluation of local and systemic effects of the recombinant adenovirus cocktail post-priming and post-boosting in the immunized animals showed that the immunogen was well tolerated and no serious negative effects were observed. Taken together, these outcomes showed that the adenovirus-vectored novel ASFV antigen cocktail was capable of safely inducing strong antibody and IFN-γ+ cell responses in commercial swine. The data will be used for selection of antigens for inclusion in a multi-antigen prototype vaccine to be evaluated for protective efficacy.


Subject(s)
Adenoviridae/genetics , African Swine Fever Virus/genetics , African Swine Fever/immunology , Antigens, Viral/immunology , Swine/immunology , African Swine Fever Virus/immunology , Animals , Antigens, Viral/genetics , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Genetic Vectors
11.
PLoS One ; 12(1): e0170504, 2017.
Article in English | MEDLINE | ID: mdl-28107431

ABSTRACT

Lack of safe and effective adjuvants is a major hindrance to the development of efficacious vaccines. Signaling via CD40 pathway leads to enhanced antigen processing and presentation, nitric oxide expression, pro-inflammatory cytokine expression by antigen presenting cells, and stimulation of B-cells to undergo somatic hypermutation, immunoglobulin class switching, and proliferation. Agonistic anti-CD40 antibodies have shown promising adjuvant qualities in human and mouse vaccine studies. An anti-CD40 monoclonal antibody (mAb), designated 2E4E4, was identified and shown to have strong agonistic effects on primary cells from multiple livestock species. The mAb recognize swine, bovine, caprine, and ovine CD40, and evoked 25-fold or greater proliferation of peripheral blood mononuclear cells (PBMCs) from these species relative to cells incubated with an isotype control (p<0.001). In addition, the mAb induced significant nitric oxide (p<0.0001) release by bovine macrophages. Furthermore, the mAb upregulated the expression of MHC-II by PBMCs, and stimulated significant (p<0.0001) IL-1α, IL6, IL-8, and TNF-α expression by PBMCs. These results suggest that the mAb 2E4E4 can target and stimulate cells from multiple livestock species and thus, it is a potential candidate for adjuvant development. This is the first study to report an anti-swine CD40 agonistic mAb that is also broadly reactive against multiple species.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , Animals , Cattle/immunology , Cross Reactions/immunology , Cytokines/metabolism , Female , Flow Cytometry , Genes, MHC Class II/immunology , Goats/immunology , HEK293 Cells , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Sheep/immunology , Swine/immunology
12.
PLoS One ; 12(1): e0170425, 2017.
Article in English | MEDLINE | ID: mdl-28099492

ABSTRACT

Bovine viral diarrhea virus (BVDV) plays a key role in bovine respiratory disease complex, which can lead to pneumonia, diarrhea and death of calves. Current vaccines are not very effective due, in part, to immunosuppressive traits and failure to induce broad protection. There are diverse BVDV strains and thus, current vaccines contain representative genotype 1 and 2 viruses (BVDV-1 & 2) to broaden coverage. BVDV modified live virus (MLV) vaccines are superior to killed virus vaccines, but they are susceptible to neutralization and complement-mediated destruction triggered by passively acquired antibodies, thus limiting their efficacy. We generated three novel mosaic polypeptide chimeras, designated NproE2123; NS231; and NS232, which incorporate protective determinants that are highly conserved among BVDV-1a, 1b, and BVDV-2 genotypes. In addition, strain-specific protective antigens from disparate BVDV strains were included to broaden coverage. We confirmed that adenovirus constructs expressing these antigens were strongly recognized by monoclonal antibodies, polyclonal sera, and IFN-γ-secreting T cells generated against diverse BVDV strains. In a proof-of-concept efficacy study, the multi-antigen proto-type vaccine induced higher, but not significantly different, IFN-γ spot forming cells and T-cell proliferation compared to a commercial MLV vaccine. In regards to the humoral response, the prototype vaccine induced higher BVDV-1 specific neutralizing antibody titers, whereas the MLV vaccine induced higher BVDV-2 specific neutralizing antibody titers. Following BVDV type 2a (1373) challenge, calves immunized with the proto-type or the MLV vaccine had lower clinical scores compared to naïve controls. These results support the hypothesis that a broadly protective subunit vaccine can be generated using mosaic polypeptides that incorporate rationally selected and validated protective determinants from diverse BVDV strains. Furthermore, regarding biosafety of using a live vector in cattle, we showed that recombinant human adenovirus-5 was cleared within one week following intradermal inoculation.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/immunology , Bovine Virus Diarrhea-Mucosal Disease/immunology , Cross Protection/immunology , Cross-Priming/immunology , Diarrhea Virus 1, Bovine Viral/immunology , Diarrhea Virus 2, Bovine Viral/immunology , Vaccines, Subunit/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/genetics , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Cell Line , Chimera/genetics , Chimera/immunology , Diarrhea Virus 1, Bovine Viral/genetics , Diarrhea Virus 2, Bovine Viral/genetics , HEK293 Cells , Humans , Interferon-gamma/immunology , T-Lymphocytes/immunology , Vaccination
13.
Res Vet Sci ; 108: 112-5, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27663378

ABSTRACT

Many pathogens enter the host through mucosal surfaces and spread rapidly via the circulation. The most effective way to prevent disease is to establish mucosal and systemic immunity against the pathogen. However, current vaccination programs in poultry industry require repeated administrations of live-attenuated virus or large amounts (10 to 100µg) of antigen together with adjuvant to induce specific secretory IgA immune responses at the mucosal effector sites. In the present study, we show that a single administration of 0.4µg of oligopeptide complexed with an agonistic anti-chicken CD40 (chCD40) monoclonal antibody (Mab) effectively targets antigen-presenting cells of the bird's mucosa-associated lymphoid tissue in vivo, and induces peptide-specific secretory IgA (sIgA) in the trachea 7days post administration. Anti-chCD40 Mab-peptide complex was administered once to four-week old male Leghorns via various mucosal routes (orally, via cloacal drinking, or oculo-nasally) or via subcutaneous (s.c.) immunization. Immunization through any of the three mucosal induction routes induced significant peptide-specific mucosal sIgA responses 7 and 14days after immunization. Interestingly, s.c. injection of the complex also induced mucosal sIgA. Our data suggest in vivo targeting of CD40 as a potential adjuvant platform, particularly for the purpose of enhancing and speeding up mucosal vaccine responses in chickens, and potentially other food animals. This is the first study able to elicit specific sIgA immune responses in remote mucosal sites with a single administration of only 0.4µg of antigen.


Subject(s)
Avian Proteins/metabolism , CD40 Antigens/immunology , Chickens/immunology , Immunoglobulin A, Secretory/metabolism , Poultry Diseases/prevention & control , Vaccination/veterinary , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal/veterinary , Administration, Oral , Animals , CD40 Antigens/administration & dosage , Injections, Subcutaneous/veterinary , Male , Mucous Membrane/immunology
14.
Clin Vaccine Immunol ; 23(11): 888-900, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27628166

ABSTRACT

The African swine fever virus (ASFV) causes a fatal hemorrhagic disease in domestic swine, and at present no treatment or vaccine is available. Natural and gene-deleted, live attenuated strains protect against closely related virulent strains; however, they are yet to be deployed and evaluated in the field to rule out chronic persistence and a potential for reversion to virulence. Previous studies suggest that antibodies play a role in protection, but induction of cytotoxic T lymphocytes (CTLs) could be the key to complete protection. Hence, generation of an efficacious subunit vaccine depends on identification of CTL targets along with a suitable delivery method that will elicit effector CTLs capable of eliminating ASFV-infected host cells and confer long-term protection. To this end, we evaluated the safety and immunogenicity of an adenovirus-vectored ASFV (Ad-ASFV) multiantigen cocktail formulated in two different adjuvants and at two immunizing doses in swine. Immunization with the cocktail rapidly induced unprecedented ASFV antigen-specific antibody and cellular immune responses against all of the antigens. The robust antibody responses underwent rapid isotype switching within 1 week postpriming, steadily increased over a 2-month period, and underwent rapid recall upon boost. Importantly, the primed antibodies strongly recognized the parental ASFV (Georgia 2007/1) by indirect fluorescence antibody (IFA) assay and Western blotting. Significant antigen-specific gamma interferon-positive (IFN-γ+) responses were detected postpriming and postboosting. Furthermore, this study is the first to demonstrate induction of ASFV antigen-specific CTL responses in commercial swine using Ad-ASFV multiantigens. The relevance of the induced immune responses in regard to protection needs to be evaluated in a challenge study.


Subject(s)
African Swine Fever Virus/immunology , Antigens, Viral/immunology , Immunity, Cellular , Immunogenicity, Vaccine , Viral Vaccines/immunology , Adenoviridae/genetics , Animals , Antigens, Viral/chemistry , Genetic Vectors , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Swine , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Viral Vaccines/adverse effects , Virulence
15.
Vet Res Commun ; 39(4): 229-36, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26521056

ABSTRACT

Neonatal calf colibacillosis caused by enterotoxigenic Escherichia coli (ETEC) is an economically significant problem in most parts of the world. The most common ETEC found in calves express the F5 (K99) fimbriae, which are necessary for the attachment of the bacteria to the ganglioside receptors on enterocytes. It is known that prevention of ETEC F5(+) adhesion to its ganglioside receptors with specific antibodies protects calves from colibacillosis. Previously we have described the development and characterization of a mouse recombinant antibody fragment (moRAb) that prevents F5 fimbrial protein induced agglutination of horse red blood cells (HRBC), which exhibit the same gangloside receptor for F5 fimbriae. Here we demonstrate that this recombinant antibody fragment inhibits in vitro the attachment of ETEC F5(+) bacteria to HRBC as well as isolated calf enterocytes, and in vivo it decreases fluid accumulation in intestinal loops of calves. Thus, correct oral administration of this anti-F5 moRAb may serve as an immunoprophylactic for cost effective control of colibacillosis in calves.


Subject(s)
Antibodies, Bacterial/pharmacology , Bacterial Adhesion/drug effects , Cattle Diseases/prevention & control , Enterocytes/drug effects , Escherichia coli Infections/veterinary , Animals , Animals, Newborn , Antibodies, Bacterial/immunology , Antibodies, Bacterial/therapeutic use , Cattle , Cattle Diseases/drug therapy , Cattle Diseases/pathology , Enterotoxins/toxicity , Erythrocytes/drug effects , Escherichia coli , Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , Escherichia coli Infections/prevention & control , Escherichia coli Proteins/immunology , Fimbriae, Bacterial/pathology , Horses , Ileum/pathology , Male , Recombinant Proteins/immunology
16.
J Immunol Methods ; 378(1-2): 116-20, 2012 Apr 30.
Article in English | MEDLINE | ID: mdl-22366632

ABSTRACT

Producing diagnostic antibodies in chicken egg yolk represents an alternate animal system that offers many advantages including high productivity at low cost. Despite being an excellent counterpart to mammalian antibodies, chicken IgG from yolk still represents an underused resource. The potential of agonistic monoclonal anti-CD40 antibodies (mAb) as a powerful immunological adjuvant has been demonstrated in mammals, but not in chickens. We recently reported an agonistic anti-chicken CD40 mAb (designated mAb 2C5) and showed that it may have potential as an immunological adjuvant. In this study, we examined the efficacy of targeting a short peptide to chicken CD40 [expressed by the antigen-presenting cells (APCs)] in enhancing an effective IgG response in chickens. For this purpose, an immune complex consisting of one streptavidin molecule, two directionally biotinylated mAb 2C5 molecules, and two biotinylated peptide molecules was produced. Chickens were immunized subcutaneously with doses of this complex ranging from 10 to 90 µg per injection once, and relative quantification of the peptide-specific IgG response showed that the mAb 2C5-based complex was able to elicit a strong IgG response as early as four days post-immunization. This demonstrates that CD40-targeting antigen to chicken APCs can significantly enhance antibody responses and induce immunoglobulin isotype-switching. This immunization strategy holds promise for rapid production of hapten-specific IgG in chickens.


Subject(s)
Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , CD40 Antigens/immunology , Haptens/immunology , Immunization/methods , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibody Formation/immunology , Antigen-Presenting Cells/immunology , Chickens , Egg Yolk/immunology , Injections, Subcutaneous/methods , Streptavidin/immunology
17.
Dev Comp Immunol ; 34(11): 1139-43, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20599554

ABSTRACT

CD40 is mainly expressed by professional antigen-presenting cells (APCs). Its ligand, CD40L, is transiently expressed on activated CD4(+) T-cells. CD40-CD40L interactions mediate T-cell help to APCs and provide crucial signals for affinity maturation and B-cell class switching. In mammals, agonistic monoclonal anti-CD40 antibodies (mAbs) mimic the effects of CD40L on APCs, leading to enhanced T-cell priming and expansion, increased antibody production and class switching. In this study, we describe agonistic anti-chicken CD40 mAb 2C5. This mAb detected CD40 on primary chicken B-cells and macrophages, DT40 B-cells, and HD11 macrophages, induced NO synthesis in HD11 macrophages, and stimulated DT40 B-cell proliferation. These observations demonstrated at least partial functional equivalence of 2C5 to chicken CD154. This mAb may therefore constitute a new tool to study the role of CD40 in the chicken immune system, and its agonistic effects suggest that it could also be used as an adjuvant.


Subject(s)
Antibodies, Monoclonal/metabolism , Avian Proteins/metabolism , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , Macrophages/metabolism , Adjuvants, Immunologic , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Avian Proteins/agonists , Avian Proteins/genetics , Avian Proteins/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD40 Antigens/agonists , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Ligand/immunology , Cell Proliferation/drug effects , Cells, Cultured , Chickens , Genetic Engineering , Ligands , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Nitric Oxide/metabolism , Protein Structure, Tertiary/genetics
18.
Transgenic Res ; 18(3): 347-60, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18830803

ABSTRACT

As a first step towards manufacturing functional anti-K99 single chain variable antibody fragment (scFv) in a plant system to prevent colibacillosis in neonatal calves, we investigated the feasibility of producing these antibodies in rice plants. Two scFv constructs, with or without the endoplasmic reticulum (ER) targeting KDEL sequence, were introduced into rice for either ER-retention of the recombinant antibody or its secretion. In agreement with several other published reports, extremely low-levels of scFv were produced in rice plants transformed with the construct lacking the ER-targeting sequence. Constructs containing the KDEL sequence resulted in significantly higher levels of the antibody in rice leaves. Although scFv transcripts were found in all three rice tissues analyzed, scFv protein was detected only in the leaf and embryo tissues and not in the endosperm portion of the seed. Functionality of the rice-produced scFv was tested in two in vitro assays, i.e., inhibition of K99-induced horse red blood cell agglutination and inhibition of the attachment of enterotoxigenic Escherichia coli (ETEC) to calf enterocytes. Rice-scFv was found to be functionally equivalent to anti-K99 monoclonal antibody (mAb) in both the assays. The results obtained in this investigation provide valuable information and in combination with other studies of this kind, will be helpful in devising strategies to improve production of useful recombinant proteins in the seeds.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antigens, Surface/immunology , Bacterial Toxins/immunology , Immunoglobulin Variable Region/biosynthesis , Oryza/metabolism , Plants, Genetically Modified/metabolism , Agglutination Tests , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Bacterial Adhesion/drug effects , Cattle , Erythrocytes/drug effects , Erythrocytes/microbiology , Escherichia coli/physiology , Horses , Immunoglobulin Variable Region/isolation & purification , Immunoglobulin Variable Region/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
19.
Vet Immunol Immunopathol ; 121(1-2): 58-67, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17897723

ABSTRACT

A single-chain antibody library against Eimeria tenella sporozoites was constructed by phage display. Antibody-displaying phage was selected in five panning rounds against cryopreserved E. tenella sporozoites. A 1000-fold increase in phage output and a 3000-fold enrichment were obtained after three rounds of panning, as the binding clones became the dominant population in the library. Ten clones were randomly selected from the last selection round, and their nucleotide sequences were aligned and compared to chicken germ-line sequences. Analysis of the light chain variable regions revealed possible donor pseudogenes which act as donors in gene conversion events, and contribute to the diversification of the V(L) immune repertoire. Possible somatic hypermutation events, a consequence of affinity maturation, were also identified. Soluble antibody was produced in a non-suppressor E. coli strain, purified by nickel affinity chromatography, and characterized by immunoblotting. In an immunofluorescence assay, this recombinant antibody showed specific binding to E. tenella sporozoites.


Subject(s)
Antibodies, Protozoan/biosynthesis , Chickens , Coccidiosis/veterinary , Eimeria tenella/immunology , Immunoglobulin Variable Region/immunology , Poultry Diseases/parasitology , Amino Acid Sequence , Animals , Antibodies, Protozoan/genetics , Antibodies, Protozoan/immunology , Antibody Specificity , Base Sequence , Coccidiosis/parasitology , Enzyme-Linked Immunosorbent Assay/veterinary , Microscopy, Fluorescence/veterinary , Molecular Sequence Data , Peptide Library , Sequence Alignment
20.
Vet Parasitol ; 137(1-2): 28-35, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16442735

ABSTRACT

Recombinant antigen-based enzyme-linked immunosorbent assay (ELISA) was developed for the serological diagnosis of Babesia bigemina infection by using a full-length B. bigemina rhoptry-associated protein 1 (rRAP-1) and the truncated C-terminal RAP-1 (rRAP-1/CT). While the rRAP-1 showed cross reactivity between B. bigemina- and Babesia bovis-infected bovine sera, the rRAP-1/CT was highly specific to B. bigemina-infected bovine sera and proved useful in the detection of sequential sera collected from an experimentally infected cow during the acute and latent infection. The high yield of soluble rRAP-1/CT and its diagnostic specificity demonstrate its potential in the diagnosis of B. bigemina infection. Its usefulness for epidemiological investigation is currently being evaluated.


Subject(s)
Antigens, Protozoan , Babesia/immunology , Babesiosis/veterinary , Cattle Diseases/diagnosis , Cattle Diseases/parasitology , Protozoan Proteins/immunology , Animals , Babesiosis/diagnosis , Babesiosis/parasitology , Cattle , Cross Reactions , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/veterinary , Fluorescent Antibody Technique, Indirect/methods , Fluorescent Antibody Technique, Indirect/veterinary , Gene Expression , Mice , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...