Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.754
Filter
1.
Can Vet J ; 65(6): 581-586, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38827595

ABSTRACT

Objective: Bovine respiratory disease (BRD) and overall postweaning treatment rates were compared among 3 groups of calves either differentially primed and boosted with commercially available bovine coronavirus (BCoV) vaccine or not vaccinated against BCoV. Animals: Commercial heifer and steer beef calves born in April and May 2022. Procedure: In June 2022, calves were randomly enrolled into 3 treatment groups. Those in 2 groups [V1 (n = 160) and V2 (n = 160)] were administered a mucosal priming dose of 1 of 2 commercial BCoV vaccines; those in the 3rd group [CTL (n = 151)] were unvaccinated against BCoV. The V1 and V2 groups were boosted by intramuscular injection pre-weaning with the same vaccine used for priming. Weaning occurred 3 wk after the last preweaning processing day. Ranch staff used a BRD case definition provided by their herd veterinarian to identify, treat, and record treatments for 45 d post-weaning. Results: Postweaning BRD treatment rates for V1, V2, and CTL were 7%, 9%, and 14%, respectively. The CTL calves had 2.2× greater odds of receiving treatment for BRD than V1 calves. There were no differences in odds of treatment between CTL and V2 calves or V1 and V2 calves. Conclusion: In a herd with previously diagnosed BCoV BRD cases, prime-boost vaccination of calves is associated with a difference in odds of BRD treatment post-weaning compared to not vaccinating calves against BCoV. Clinical relevance: Prime-boost vaccination with commercial BCoV vaccine may be an important management tool for herds with known BCoV BRD outbreaks.


Comparaison des taux de traitement des maladies respiratoires bovines après le sevrage entre des veaux de boucherie témoins non vaccinés et des veaux vaccinés amorce-rappel de manière variable à l'aide de vaccins contre le coronavirus bovin commercialement disponibles. Objectif: La maladie respiratoire bovine (BRD) et les taux globaux de traitement post-sevrage ont été comparés parmi 3 groupes de veaux soit vaccinés de manière différentielle et avec un rappel avec le vaccin contre le coronavirus bovin (BCoV) disponible commercialement, soit non vaccinés contre le BCoV. Animaux: Génisses et veaux de boucherie commerciaux nés en avril et mai 2022. Procédure: En juin 2022, les veaux ont été randomisés lors du recrutement dans 3 groupes de traitement. Ceux des 2 groupes [V1 (n = 160) et V2 (n = 160)] ont reçu une dose d'amorce par voie muqueuse de l'un des deux vaccins commerciaux BCoV; ceux du 3ème groupe [CTL (n = 151)] n'étaient pas vaccinés contre le BCoV. Les groupes V1 et V2 ont eu un rappel par injection intramusculaire avant le sevrage avec le même vaccin que celui utilisé pour l'amorçage. Le sevrage a eu lieu 3 semaines après le dernier jour de conditionnement pré-sevrage. Le personnel du ranch a utilisé une définition de cas de BRD fournie par le vétérinaire de leur troupeau pour identifier, traiter et enregistrer les traitements pendant 45 jours après le sevrage. Résultats: Les taux de traitement BRD post-sevrage pour V1, V2 et CTL étaient respectivement de 7 %, 9 % et 14 %. Les veaux CTL avaient 2,2 fois plus de chances de recevoir un traitement contre la BRD que les veaux V1. Il n'y avait aucune différence dans les probabilités de traitement entre les veaux CTL et V2 ou entre les veaux V1 et V2. Conclusion: Dans un troupeau avec des cas de BRD causés par le BCoV déjà diagnostiqués, la vaccination amorce-rappel des veaux est associée à une différence de probabilité de traitement par le BRD après le sevrage par rapport à la nonvaccination des veaux contre le BCoV. Pertinence clinique: La vaccination amorce-rappel avec le vaccin commercial BCoV peut être un outil de gestion important pour les troupeaux présentant des foyers connus de BCoV BRD.(Traduit par Dr Serge Messier).


Subject(s)
Coronavirus, Bovine , Viral Vaccines , Animals , Cattle , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Coronavirus, Bovine/immunology , Male , Female , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Weaning , Vaccination/veterinary , Bovine Respiratory Disease Complex/prevention & control
2.
Hum Vaccin Immunother ; 20(1): 2346390, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38691025

ABSTRACT

Middle East respiratory coronavirus (MERS-CoV) is a newly emergent, highly pathogenic coronavirus that is associated with 34% mortality rate. MERS-CoV remains listed as priority pathogen by the WHO. Since its discovery in 2012 and despite the efforts to develop coronaviruses vaccines to fight against SARS-CoV-2, there are currently no MERS-CoV vaccine that has been approved. Therefore, there is high demand to continue on the development of prophylactic vaccines against MERS-CoV. Current advancements in vaccine developments can be adapted for the development of improved MERS-CoV vaccines candidates. Nucleic acid-based vaccines, including pDNA and mRNA, are relatively new class of vaccine platforms. In this work, we developed pDNA and mRNA vaccine candidates expressing S.FL gene of MERS-CoV. Further, we synthesized a silane functionalized hierarchical aluminosilicate to encapsulate each vaccine candidates. We tested the nucleic acid vaccine candidates in mice and evaluated humoral antibodies response. Interestingly, we determined that the non-encapsulated, codon optimized S.FL pDNA vaccine candidate elicited the highest level of antibody responses against S.FL and S1 of MERS-CoV. Encapsulation of mRNA with nanoporous aluminosilicate increased the humoral antibody responses, whereas encapsulation of pDNA did not. These findings suggests that MERS-CoV S.FL pDNA vaccine candidate induced the highest level of humoral responses. This study will enhance further optimization of nanosilica as potential carrier for mRNA vaccines. In conclusion, this study suggests MERS-CoV pDNA vaccine candidate as a suitable vaccine platform for further pivotal preclinical testings.


Subject(s)
Antibodies, Viral , Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , Nanoparticles , Silicon Dioxide , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Vaccines, DNA/administration & dosage , Middle East Respiratory Syndrome Coronavirus/immunology , Middle East Respiratory Syndrome Coronavirus/genetics , Mice , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Antibodies, Viral/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Silicon Dioxide/chemistry , Mice, Inbred BALB C , Female , Humans , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Vaccine Development
3.
BMC Infect Dis ; 24(1): 476, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714948

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne viral disease caused by the SFTS virus (Dabie bandavirus), which has become a substantial risk to public health. No specific treatment is available now, that calls for an effective vaccine. Given this, we aimed to develop a multi-epitope DNA vaccine through the help of bioinformatics. The final DNA vaccine was inserted into a special plasmid vector pVAX1, consisting of CD8+ T cell epitopes, CD4+ T cell epitopes and B cell epitopes (six epitopes each) screened from four genome-encoded proteins--nuclear protein (NP), glycoprotein (GP), RNA-dependent RNA polymerase (RdRp), as well as nonstructural protein (NSs). To ascertain if the predicted structure would be stable and successful in preventing infection, an immunological simulation was run on it. In conclusion, we designed a multi-epitope DNA vaccine that is expected to be effective against Dabie bandavirus, but in vivo trials are needed to verify this claim.


Subject(s)
Epitopes, T-Lymphocyte , Phlebovirus , Severe Fever with Thrombocytopenia Syndrome , Vaccines, DNA , Viral Vaccines , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Phlebovirus/immunology , Phlebovirus/genetics , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Severe Fever with Thrombocytopenia Syndrome/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Humans , Computer-Aided Design , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Animals , Computational Biology
5.
Viruses ; 16(5)2024 05 10.
Article in English | MEDLINE | ID: mdl-38793639

ABSTRACT

African Swine Fever Virus (ASFV) is a large dsDNA virus that encodes at least 150 proteins. The complexity of ASFV and lack of knowledge of effector immune functions and protective antigens have hindered the development of safe and effective ASF vaccines. In this study, we constructed four Orf virus recombinant vectors expressing individual ASFV genes B602L, -CP204L, E184L, and -I73R (ORFVΔ121-ASFV-B602L, -CP204L, -E184L, and -I73R). All recombinant viruses expressed the heterologous ASFV proteins in vitro. We then evaluated the immunogenicity of the recombinants by immunizing four-week-old piglets. In two independent animal studies, we observed high antibody titers against ASFV p30, encoded by CP204L gene. Using Pepscan ELISA, we identified a linear B-cell epitope of 12 amino acids in length (Peptide 15) located in an exposed loop region of p30 as an immunodominant ASFV epitope. Additionally, antibodies elicited against ASFV p30 presented antibody-dependent cellular cytotoxicity (ADCC) activity. These results underscore the role of p30 on antibody responses elicited against ASFV and highlight an important functional epitope that contributes to p30-specific antibody responses.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antibodies, Viral , Antibody-Dependent Cell Cytotoxicity , Epitopes, B-Lymphocyte , Immunodominant Epitopes , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Animals , Swine , Antibodies, Viral/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Immunodominant Epitopes/immunology , Immunodominant Epitopes/genetics , African Swine Fever/immunology , African Swine Fever/virology , Viral Proteins/immunology , Viral Proteins/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics
6.
Viruses ; 16(5)2024 05 16.
Article in English | MEDLINE | ID: mdl-38793675

ABSTRACT

The emergence of new virulent genotypes and the continued genetic drift of Newcastle disease virus (NDV) implies that distinct genotypes of NDV are simultaneously evolving in different geographic locations across the globe, including throughout Africa, where NDV is an important veterinary pathogen. Expanding the genomic diversity of NDV increases the possibility of diagnostic and vaccine failures. In this review, we systematically analyzed the genetic diversity of NDV genotypes in Africa using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Information published between 1999 and 2022 were used to obtain the genetic background of different genotypes of NDV and their geographic distributions in Africa. The following genotypes were reported in Africa: I, II, III, IV, V, VI, VII, VIII, XI, XIII, XIV, XVII, XVIII, XX, and XXI. A new putative genotype has been detected in the Democratic Republic of the Congo. However, of 54 African countries, only 26 countries regularly report information on NDV outbreaks, suggesting that this number may be vastly underestimated. With eight different genotypes, Nigeria is the country with the greatest genotypic diversity of NDV among African countries. Genotype VII is the most prevalent group of NDV in Africa, which was reported in 15 countries. A phylogeographic analysis of NDV sequences revealed transboundary transmission of the virus in Eastern Africa, Western and Central Africa, and in Southern Africa. A regional and continental collaboration is recommended for improved NDV risk management in Africa.


Subject(s)
Genetic Variation , Genotype , Newcastle Disease , Newcastle disease virus , Phylogeny , Newcastle disease virus/genetics , Newcastle disease virus/classification , Newcastle disease virus/isolation & purification , Newcastle Disease/virology , Newcastle Disease/epidemiology , Africa/epidemiology , Animals , Genome, Viral , Vaccination/veterinary , Chickens/virology , Viral Vaccines/genetics , Viral Vaccines/immunology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Phylogeography
7.
Vet Q ; 44(1): 1-12, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38726839

ABSTRACT

Duck plague (DP) is an acute, contagious and fatal disease, caused by duck enteritis virus (DEV), with worldwide distribution causing several outbreaks and posing severe economic losses. The present study was carried out with a goal of development of a live attenuated cell culture based DP vaccine using an Indian strain of DEV and evaluation of its safety, efficacy along with complete genome analysis. The live attenuated DP vaccine (DPvac/IVRI-19) was developed by serial propagation of a virulent isolate of DEV (DEV/India/IVRI-2016) in the chicken embryo fibroblast (CEF) primary cell culture. Adaptation of DEV in CEF cell culture was indicated by more rapid appearance of cytopathic effects (CPE) and gradual increase of virus titre, which reached up to 107.5 TCID50/mL after 41 passages. The safety, immunogenicity and efficacy of the vaccine were determined by immunization trials in ducklings. The DPvac/IVRI-19 was found to be avirulent and completely safe in the ducklings. Further, the vaccine induced both humoral and cell mediated immune responses and afforded 100% protection against the virulent DEV challenge. A comparison of the whole genome of DPvac/IVRI-19 (MZ911871) and DEV/India/IVRI-2016 (MZ824102) revealed significant number of mutations, which might be associated with viral attenuation. Phylogenetic tree of DEV/India/IVRI-2016 revealed its evolutionary relationship with other DEV isolates, but it formed a separate cluster with certain unique mutations. Thus, with the proven safety and 100% efficacy, the DPvac/IVRI-19 is suitable for large scale production with precisely pure form of vaccine and has potential utility at national and global levels.


Subject(s)
Ducks , Fibroblasts , Mardivirus , Poultry Diseases , Vaccines, Attenuated , Viral Vaccines , Animals , Vaccines, Attenuated/immunology , Ducks/virology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Fibroblasts/virology , Chick Embryo , Viral Vaccines/immunology , Mardivirus/immunology , Mardivirus/pathogenicity , Herpesviridae Infections/veterinary , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , India
8.
Front Immunol ; 15: 1373656, 2024.
Article in English | MEDLINE | ID: mdl-38742108

ABSTRACT

African swine fever virus (ASFV) is one of the most complex viruses. ASFV is a serious threat to the global swine industry because no commercial vaccines against this virus are currently available except in Vietnam. Moreover, ASFV is highly stable in the environment and can survive in water, feed, and aerosols for a long time. ASFV is transmitted through the digestive and respiratory tract. Mucosal immunity is the first line of defense against ASFV. Saccharomyces cerevisiae (SC), which has been certified by the U.S. Food and Drug Administration and has a generally recognized as safe status in the food industry, was used for oral immunization in this study. ASFV antigens were effectively expressed in recombinant SC strains with high DNA copy numbers and stable growth though surface display technology and chromosome engineering (δ-integration). The recombinant SC strains containing eight ASFV antigens-KP177R, E183L, E199L, CP204L, E248R, EP402R, B602L, and B646L- induced strong humoral and mucosal immune responses in mice. There was no antigenic competition, and these antigens induced Th1 and Th2 cellular immune responses. Therefore, the oral immunization strategy using recombinant SC strains containing multiple ASFV antigens demonstrate potential for future testing in swine, including challenge studies to evaluate its efficacy as a vaccine against ASFV.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antigens, Viral , Immunization , Saccharomyces cerevisiae , Viral Vaccines , Animals , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Saccharomyces cerevisiae/immunology , Saccharomyces cerevisiae/genetics , Administration, Oral , Mice , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antigens, Viral/immunology , African Swine Fever/immunology , African Swine Fever/prevention & control , Swine , Immunity, Mucosal , Antibodies, Viral/blood , Antibodies, Viral/immunology , Mice, Inbred BALB C , Female , Immunity, Humoral
9.
PLoS One ; 19(5): e0300507, 2024.
Article in English | MEDLINE | ID: mdl-38728300

ABSTRACT

According to the 2018 WHO R&D Blueprint, Nipah virus (NiV) is a priority disease, and the development of a vaccine against NiV is strongly encouraged. According to criteria used to categorize zoonotic diseases, NiV is a stage III disease that can spread to people and cause unpredictable outbreaks. Since 2001, the NiV virus has caused annual outbreaks in Bangladesh, while in India it has caused occasional outbreaks. According to estimates, the mortality rate for infected individuals ranges from 70 to 91%. Using immunoinformatic approaches to anticipate the epitopes of the MHC-I, MHC-II, and B-cells, they were predicted using the NiV glycoprotein and nucleocapsid protein. The selected epitopes were used to develop a multi-epitope vaccine construct connected with linkers and adjuvants in order to improve immune responses to the vaccine construct. The 3D structure of the engineered vaccine was anticipated, optimized, and confirmed using a variety of computer simulation techniques so that its stability could be assessed. According to the immunological simulation tests, it was found that the vaccination elicits a targeted immune response against the NiV. Docking with TLR-3, 7, and 8 revealed that vaccine candidates had high binding affinities and low binding energies. Finally, molecular dynamic analysis confirms the stability of the new vaccine. Codon optimization and in silico cloning showed that the proposed vaccine was expressed to a high degree in Escherichia coli. The study will help in identifying a potential epitope for a vaccine candidate against NiV. The developed multi-epitope vaccine construct has a lot of potential, but they still need to be verified by in vitro & in vivo studies.


Subject(s)
Glycoproteins , Nipah Virus , Viral Vaccines , Nipah Virus/immunology , Viral Vaccines/immunology , Glycoproteins/immunology , Glycoproteins/chemistry , Humans , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Computer Simulation , Epitopes/immunology , Epitopes/chemistry , Molecular Dynamics Simulation , Nucleocapsid/immunology , Molecular Docking Simulation
10.
Antiviral Res ; 226: 105900, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705200

ABSTRACT

BACKGROUND & AIMS: The spread of foot-and-mouth disease virus (FMDV) through aerosol droplets among cloven-hoofed ungulates in close contact is a major obstacle for successful animal husbandry. Therefore, the development of suitable mucosal vaccines, especially nasal vaccines, to block the virus at the initial site of infection is crucial. PATIENTS AND METHODS: Here, we constructed eukaryotic expression plasmids containing the T and B-cell epitopes (pTB) of FMDV in tandem with the molecular mucosal adjuvant Fms-like tyrosine kinase receptor 3 ligand (Flt3 ligand, FL) (pTB-FL). Then, the constructed plasmid was electrostatically attached to mannose-modified chitosan-coated poly(lactic-co-glycolic) acid (PLGA) nanospheres (MCS-PLGA-NPs) to obtain an active nasal vaccine targeting the mannose-receptor on the surface of antigen-presenting cells (APCs). RESULTS: The MCS-PLGA-NPs loaded with pTB-FL not only induced a local mucosal immune response, but also induced a systemic immune response in mice. More importantly, the nasal vaccine afforded an 80% protection rate against a highly virulent FMDV strain (AF72) when it was subcutaneously injected into the soles of the feet of guinea pigs. CONCLUSIONS: The nasal vaccine prepared in this study can effectively induce a cross-protective immune response against the challenge with FMDV of same serotype in animals and is promising as a potential FMDV vaccine.


Subject(s)
Administration, Intranasal , Chitosan , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Nanospheres , Polylactic Acid-Polyglycolic Acid Copolymer , Viral Vaccines , Animals , Chitosan/chemistry , Chitosan/administration & dosage , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/genetics , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/immunology , Mice , Nanospheres/chemistry , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Mice, Inbred BALB C , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Nucleic Acids/administration & dosage , Immunity, Mucosal , Drug Delivery Systems
11.
PLoS One ; 19(5): e0300778, 2024.
Article in English | MEDLINE | ID: mdl-38758816

ABSTRACT

Mpox (formerly known as monkeypox) virus and some related poxviruses including smallpox virus pose a significant threat to public health, and effective prevention and treatment strategies are needed. This study utilized a reverse vaccinology approach to retrieve conserved epitopes for monkeypox virus and construct a vaccine that could provide cross-protection against related viruses with similar antigenic properties. The selected virulent proteins of monkeypox virus, MPXVgp165, and Virion core protein P4a, were subjected to epitope mapping for vaccine construction. Two vaccines were constructed using selected T cell epitopes and B cell epitopes with PADRE and human beta-defensins adjuvants conjugated in the vaccine sequence. Both constructs were found to be highly antigenic, non-allergenic, nontoxic, and soluble, suggesting their potential to generate an adequate immune response and be safe for humans. Vaccine construct 1 was selected for molecular dynamic simulation studies. The simulation studies revealed that the TLR8-vaccine complex was more stable than the TLR3-vaccine complex. The lower RMSD and RMSF values of the TLR8 bound vaccine compared to the TLR3 bound vaccine suggested better stability and consistency of hydrogen bonds. The Rg values of the vaccine chain bound to TLR8 indicated overall stability, whereas the vaccine chain bound to TLR3 showed deviations throughout the simulation. These results suggest that the constructed vaccine could be a potential preventive measure against monkeypox and related viruses however, further experimental validation is required to confirm these findings.


Subject(s)
Molecular Dynamics Simulation , Monkeypox virus , Humans , Monkeypox virus/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Computer Simulation , Poxviridae/immunology , Viral Vaccines/immunology , Epitope Mapping , Mpox (monkeypox)/prevention & control , Mpox (monkeypox)/immunology , Animals , Toll-Like Receptor 8/immunology
12.
Sci Rep ; 14(1): 11783, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38782944

ABSTRACT

Cyprinid herpesvirus is a causative agent of a destructive disease in common and koi carp (Cyprinus carpio), which leads to substantial global financial losses in aquaculture industries. Among the strains of C. herpesvirus, C. herpesvirus 1 (CyHV-1) and C. herpesvirus 3 (CyHV-3) are known as highly pathogenic to carp fishes in Europe, Asia, and Africa. To date, no effective vaccine has been developed to combat these viruses. This study aimed to develop unique multi-epitope subunit vaccines targeting the CyHV-1 and CyHV-3 using a reverse vaccinology approach. The study began with a comprehensive literature review to identify the most critical proteins, which were then subjected to in silico analyses to predict highly antigenic epitopes. These analyses involved assessing antigenicity, transmembrane topology screening, allergenecity, toxicity, and molecular docking approaches. We constructed two multi-epitope-based vaccines incorporating a suitable adjuvant and appropriate linkers. It revealed that both the vaccines are non-toxic and immunogenic. The tertiary structures of the vaccine proteins were generated, refined, and validated to ensure their suitability. The binding affinity between the vaccine constructs and TLR3 and TLR5 receptors were assessed by molecular docking studies. Molecular dynamics simulations indicated that vaccine construct V1 exhibited greater stability with both TLR3 and TLR5 based on RMSD analysis. Hydrogen bond analysis revealed a stronger binding affinity between the vaccine constructs and TLR5 compared to TLR3. Furthermore, MM-PBSA analysis suggested that both vaccine constructs exhibited a better affinity for TLR5. Considering all aspects, the results suggest that in silico development of CyHV vaccines incorporating multiple epitopes holds promise for management of diseases caused by CyHV-1 and CyHV-3. However, further in vivo trials are highly recommended to validate the efficacies of these vaccines.


Subject(s)
Carps , Fish Diseases , Herpesviridae Infections , Herpesviridae , Molecular Docking Simulation , Vaccines, Subunit , Animals , Vaccines, Subunit/immunology , Carps/virology , Carps/immunology , Herpesviridae/immunology , Fish Diseases/prevention & control , Fish Diseases/immunology , Fish Diseases/virology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/immunology , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Viral Vaccines/immunology , Epitopes/immunology , Epitopes/chemistry , Computational Biology/methods , Herpesvirus Vaccines/immunology , Immunoinformatics
13.
Hum Vaccin Immunother ; 20(1): 2351664, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38757508

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) is a lethal beta-coronavirus that emerged in 2012. The virus is part of the WHO blueprint priority list with a concerning fatality rate of 35%. Scientific efforts are ongoing for the development of vaccines, anti-viral and biotherapeutics, which are majorly directed toward the structural spike protein. However, the ongoing effort is challenging due to conformational instability of the spike protein and the evasion strategy posed by the MERS-CoV. In this study, we have expressed and purified the MERS-CoV pre-fusion spike protein in the Expi293F mammalian expression system. The purified protein was extensively characterized for its biochemical and biophysical properties. Thermal stability analysis showed a melting temperature of 58°C and the protein resisted major structural changes at elevated temperature as revealed by fluorescence spectroscopy and circular dichroism. Immunological assessment of the MERS-CoV spike immunogen in BALB/c mice with AddaVaxTM and Imject alum adjuvants showed elicitation of high titer antibody responses but a more balanced Th1/Th2 response with AddaVaxTM squalene like adjuvant. Together, our results suggest the formation of higher-order trimeric pre-fusion MERS-CoV spike proteins, which were able to induce robust immune responses. The comprehensive characterization of MERS-CoV spike protein warrants a better understanding of MERS spike protein and future vaccine development efforts.


Subject(s)
Antibodies, Viral , Mice, Inbred BALB C , Middle East Respiratory Syndrome Coronavirus , Spike Glycoprotein, Coronavirus , Viral Vaccines , Middle East Respiratory Syndrome Coronavirus/immunology , Animals , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Mice , Female , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Immunogenicity, Vaccine , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Adjuvants, Immunologic/administration & dosage , Adjuvants, Vaccine , Humans
14.
Viral Immunol ; 37(4): 216-219, 2024 05.
Article in English | MEDLINE | ID: mdl-38717823

ABSTRACT

In May 2022, mpox began to spread worldwide, posing a serious threat to human public health. Modified Vaccinia Ankara-Bavaria Nordic (MVA-BN) is a live attenuated orthopoxvirus vaccine that has been authorized by the U.S. Food and Drug Administration as the vaccine of choice for the prevention of mpox. In this study, we conducted a meta-analysis of all currently published literature on the efficacy and safety of the MVA-BN vaccine in the real world, showing that the MVA-BN vaccine is effective and safe, with efficacy of up to 75% with a single dose and up to 80% with a two-dose vaccine. Meanwhile, we found that subcutaneous injection has lower local and systemic adverse events than intradermal injection, regardless of single- or two-dose vaccination, and subcutaneous injection is better tolerated in children, the elderly, or people with underlying medical conditions. These results have important reference value for clinical practice.


Subject(s)
Vaccine Efficacy , Vaccines, Attenuated , Humans , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Poxviridae Infections/prevention & control , Poxviridae Infections/immunology , Vaccinia virus/immunology , Vaccinia virus/genetics , Vaccination , Injections, Subcutaneous , Injections, Intradermal , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Orthopoxvirus/immunology , Orthopoxvirus/genetics , Child
15.
ACS Nano ; 18(19): 12235-12260, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38696217

ABSTRACT

Variants of coronavirus porcine epidemic diarrhea virus (PEDV) frequently emerge, causing an incomplete match between the vaccine and variant strains, which affects vaccine efficacy. Designing vaccines with rapidly replaceable antigens and high efficacy is a promising strategy for the prevention of infection with PEDV variant strains. In our study, three different types of self-assembled nanoparticles (nps) targeting receptor-binding N-terminal domain (NTD) and C-terminal domain (CTD) of S1 protein, named NTDnps, CTDnps, and NTD/CTDnps, were constructed and evaluated as vaccine candidates against PEDV. NTDnps and CTDnps vaccines mediated significantly higher neutralizing antibody (NAb) titers than NTD and CTD recombinant proteins in mice. The NTD/CTDnps in varying ratios elicited significantly higher NAb titers when compared with NTDnps and CTDnps alone. The NTD/CTDnps (3:1) elicited NAb with titers up to 92.92% of those induced by the commercial vaccine. Piglets immunized with NTD/CTDnps (3:1) achieved a passive immune protection rate of 83.33% of that induced by the commercial vaccine. NTD/CTDnps (3:1) enhanced the capacity of mononuclear macrophages and dendritic cells to take up and present antigens by activating major histocompatibility complex I and II molecules to stimulate humoral and cellular immunity. These data reveal that a combination of S1-NTD and S1-CTD antigens targeting double receptor-binding domains strengthens the protective immunity of nanoparticle vaccines against PEDV. Our findings will provide a promising vaccine candidate against PEDV.


Subject(s)
Nanoparticles , Porcine epidemic diarrhea virus , Viral Vaccines , Porcine epidemic diarrhea virus/immunology , Animals , Nanoparticles/chemistry , Swine , Mice , Viral Vaccines/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Mice, Inbred BALB C , Antigens, Viral/immunology , Antigens, Viral/chemistry , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Protein Domains/immunology , Female , Nanovaccines
16.
Methods Mol Biol ; 2786: 51-87, 2024.
Article in English | MEDLINE | ID: mdl-38814390

ABSTRACT

Vectored RNA vaccines offer a variety of possibilities to engineer targeted vaccines. They are cost-effective and safe, but replication competent, activating the humoral as well as the cellular immune system.This chapter focuses on RNA vaccines derived from negative-strand RNA viruses from the order Mononegavirales with special attention to Newcastle disease virus-based vaccines and their generation. It shall provide an overview on the advantages and disadvantages of certain vector platforms as well as their scopes of application, including an additional section on experimental COVID-19 vaccines.


Subject(s)
Genetic Vectors , Newcastle disease virus , mRNA Vaccines , Animals , Humans , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Genetic Vectors/genetics , Newcastle disease virus/genetics , Newcastle disease virus/immunology , RNA Viruses/genetics , RNA Viruses/immunology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , mRNA Vaccines/genetics , mRNA Vaccines/immunology
17.
Int J Biol Macromol ; 270(Pt 1): 132432, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761609

ABSTRACT

The African swine fever virus (ASFV) continues to pose significant economic and pandemic risks. Consequently, discovering new, efficient vaccines is crucial. Messenger RNA (mRNA) vaccines have emerged as promising candidates, providing minimal risk of insertional mutagenesis, high safety profiles, effectiveness, rapid scalability in production, and cost-effectiveness. In this study, we have developed an ASF p30 mRNA vaccine candidate (mRNA/Man-LNP) employing mannose-modified lipid nanoparticles (LNPs). The mRNA/Man-LNP exhibited effective antigen presentation and facilitated dendritic cells (DCs) maturation. Notably, it elicited strong IgG titers and activated CD4+ and CD8+ T-cells in immunized mice, all while adhering to stringent biosafety standards. This investigation demonstrates that mRNA/Man-LNP can trigger both humoral and cellular immune responses, suggesting its potential as a potent and promising vaccine candidate for controlling African swine fever (ASF).


Subject(s)
African Swine Fever Virus , African Swine Fever , Mannose , Nanoparticles , Viral Vaccines , Animals , Nanoparticles/chemistry , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , African Swine Fever/prevention & control , African Swine Fever/immunology , Mice , Viral Vaccines/immunology , Swine , Mannose/chemistry , Dendritic Cells/immunology , Lipids/chemistry , Vaccine Development , RNA, Messenger/genetics , RNA, Messenger/immunology , mRNA Vaccines , Female , Antibodies, Viral/immunology , Antibodies, Viral/blood , Liposomes
18.
Med Sci (Basel) ; 12(2)2024 May 22.
Article in English | MEDLINE | ID: mdl-38804384

ABSTRACT

mRNA vaccines have emerged as an optimistic technological platform for vaccine innovation in this new scientific era. mRNA vaccines have dramatically altered the domain of vaccinology by offering a versatile and rapid approach to combating infectious diseases and virus-induced cancers. Clinical trials have demonstrated efficacy rates of 94-95% in preventing COVID-19, and mRNA vaccines have been increasingly recognized as a powerful vaccine platform. Although mRNA vaccines have played an essential role in the COVID-19 pandemic, they still have several limitations; their instability and degradation affect their storage, delivery, and over-all efficiency. mRNA is typically enclosed in a transport mechanism to facilitate its entry into the target cell because it is an unstable and negatively charged molecule. For instance, mRNA that is given using lipid-nanoparticle-based vaccine delivery systems (LNPs) solely enters cells through endocytosis, establishing an endosome without damaging the cell membrane. The COVID-19 pandemic has accelerated the development of mRNA vaccine platforms used to treat and prevent several infectious diseases. This technology has the potential to change the future course of the disease by providing a safe and effective way to combat infectious diseases and cancer. A single-stranded genetic sequence found in mRNA vaccines instructs host cells to produce proteins inside ribosomes to elicit immunological responses and prepare the immune system to fight infections or cancer cells. The potential applications of mRNA vaccine technology are vast and can lead to the development of a preferred vaccine pattern. As a result, a new generation of vaccinations has gradually gained popularity and access to the general population. To adapt the design of an antigen, and even combine sequences from different variations in response to new changes in the viral genome, mRNA vaccines may be used. Current mRNA vaccines provide adequate safety and protection, but the duration of that protection can only be determined if further clinical research is conducted.


Subject(s)
COVID-19 , SARS-CoV-2 , mRNA Vaccines , Humans , COVID-19/prevention & control , SARS-CoV-2/immunology , Pandemics/prevention & control , Oncogenic Viruses , Vaccines, Synthetic , Vaccine Development , COVID-19 Vaccines/immunology , Pneumonia, Viral/prevention & control , Coronavirus Infections/prevention & control , Betacoronavirus , Viral Vaccines/immunology , RNA, Messenger , Neoplasms
19.
Open Vet J ; 14(4): 1019-1028, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38808294

ABSTRACT

Background: Canine distemper (CD) is a worldwide spread disease that has been described in 12 families of mammals, especially in the Carnivora order, being better studied in domestic canines where vaccination represents the best means of control. CD is controlled by vaccination, but many cases of the disease still occur in vaccinated animals. Aim: The aim of this work was to study antigen-specific epitopes that can subsidize the development of a new vaccine approach. Methods: Mapping of T cell reactive epitopes for CD virus (CDV) was carried out through enzyme-linked immunospot assays using 119 overlapped synthetic peptides from the viral hemagglutinin protein, grouped in 22 pools forming a matrix to test the immune response of 32 animals. Results: Evaluations using the criteria established to identify reactive pools, demonstrated that 26 animals presented at least one reactive pool, that one pool was not reactive to any animal, and six pools were the most frequent among the reactive peptides. The crisscrossing of the most reactive pools in the matrix revealed nine peptides considered potential candidate epitopes for T cell stimulation against the CDV and those were used to design an in-silico protein, containing also predicted epitopes for B cell stimulation, and further analyzed using immune epitope databases to ensure protein quality and stability. Conclusion: The final in silico optimized protein presents characteristics that qualify it to be used to develop a new prototype epitope-based anti-CDV vaccine.


Subject(s)
Distemper Virus, Canine , Distemper , Epitope Mapping , Viral Vaccines , Distemper Virus, Canine/immunology , Animals , Distemper/prevention & control , Distemper/immunology , Dogs , Viral Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Enzyme-Linked Immunospot Assay/veterinary
20.
Adv Exp Med Biol ; 1451: 273-287, 2024.
Article in English | MEDLINE | ID: mdl-38801584

ABSTRACT

Smallpox was a significant cause of mortality for over three thousand years, amounting to 10% of deaths yearly. Edward Jenner discovered smallpox vaccination in 1796, which rapidly became a smallpox infection preventive practice throughout the world and eradicated smallpox infection by 1980. After smallpox eradication, monkeypox vaccines have been used primarily in research and in outbreaks in Africa, where the disease is endemic. In the present, the vaccines are being used for people who work with animals or in high-risk areas, as well as for healthcare workers treating patients with monkeypox. Among all orthopoxviruses (OPXV), monkeypox viral (MPXV) infection occurs mainly in cynomolgus monkeys, natural reservoirs, and occasionally causes severe multi-organ infection in humans, who were the incidental hosts. The first case of the present epidemic of MXPV was identified on May 7, 2022, and rapidly increased the number of cases. In this regard, the WHO declared the outbreak, an international public health emergency on July 23, 2022. The first monkeypox vaccine was developed in the 1960s by the US Army and was based on the vaccinia virus, which is also used in smallpox vaccines. In recent years, newer monkeypox vaccines have been developed based on other viruses such as Modified Vaccinia Ankara (MVA). These newer vaccines are safer and can provide longer-lasting immunity with fewer side effects. For the future, there is ongoing research to improve the current vaccines and to develop new ones. One notable advance has been the development of a recombinant vaccine that uses a genetically modified vaccinia virus to express monkeypox antigens. This vaccine has shown promising results in pre-clinical trials and is currently undergoing further testing in clinical trials. Another recent development has been the use of a DNA vaccine, which delivers genetic material encoding monkeypox antigens directly into cells. This type of vaccine has shown effectiveness in animal studies and is also undergoing clinical testing in humans. Overall, these recent advances in monkeypox vaccine development hold promise for protecting individuals against this potentially serious disease.


Subject(s)
Smallpox Vaccine , Humans , Animals , Smallpox Vaccine/immunology , Smallpox/prevention & control , Smallpox/immunology , Smallpox/epidemiology , Smallpox/history , History, 21st Century , History, 20th Century , Mpox (monkeypox)/prevention & control , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/immunology , Poxviridae Infections/prevention & control , Poxviridae Infections/immunology , Poxviridae Infections/epidemiology , Poxviridae/immunology , Poxviridae/genetics , Monkeypox virus/immunology , Monkeypox virus/genetics , Vaccination , Viral Vaccines/immunology , Vaccine Development
SELECTION OF CITATIONS
SEARCH DETAIL
...