Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Viruses ; 14(2)2022 01 19.
Article in English | MEDLINE | ID: mdl-35215786

ABSTRACT

The Varicella-zoster virus (VZV) or human herpes virus 3 is a neurotropic human alpha herpes virus responsible for chickenpox/varicella and shingles/Herpes zoster (HZ). This review will focus on HZ. Since HZ is secondary to varicella, its incidence increases with age. In children and youngsters, HZ is rare and associated to metabolic and neoplastic disorders. In adults, advanced age, distress, other infections (such as AIDS or COVID-19), and immunosuppression are the most common risk factors. HZ reactivation has recently been observed after COVID-19 vaccination. The disease shows different clinical stages of variable clinical manifestations. Some of the manifestations bear a higher risk of complications. Among the possible complications, postherpetic neuralgia, a chronic pain disease, is one of the most frequent. HZ vasculitis is associated with morbidity and mortality. Renal and gastrointestinal complications have been reported. The cornerstone of treatment is early intervention with acyclovir or brivudine. Second-line treatments are available. Pain management is essential. For (secondary) prophylaxis, currently two HZV vaccines are available for healthy older adults, a live attenuated VZV vaccine and a recombinant adjuvanted VZV glycoprotein E subunit vaccine. The latter allows vaccination also in severely immunosuppressed patients. This review focuses on manifestations of HZ and its management. Although several articles have been published on HZ, the literature continues to evolve, especially in regard to patients with comorbidities and immunocompromised patients. VZV reactivation has also emerged as an important point of discussion during the COVID-19 pandemic, especially after vaccination. The objective of this review is to discuss current updates related to clinical presentations, complications, and management of HZ.


Subject(s)
Disease Management , Herpes Zoster/drug therapy , Herpes Zoster/prevention & control , Herpesvirus 3, Human/pathogenicity , Herpesvirus Vaccines/immunology , Herpes Zoster/complications , Herpes Zoster/physiopathology , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/classification , Humans , Immunocompromised Host , Incidence , Latent Infection/virology , Morbidity , Neuralgia, Postherpetic/virology , Risk Factors , Vaccination , Vaccines, Synthetic/administration & dosage
2.
Virology ; 566: 98-105, 2022 01.
Article in English | MEDLINE | ID: mdl-34896902

ABSTRACT

The innate and acquired immune response induced by a commercial inactivated vaccine against Bovine Herpesvirus-1 (BoHV-1) and protection conferred against the virus were analyzed in cattle. Vaccination induced high levels of BoHV-1 antibodies at 30, 60, and 90 days post-vaccination (dpv). IgG1 and IgG2 isotypes were detected at 90 dpv, as well as virus-neutralizing antibodies. An increase of anti-BoHV-1 IgG1 in nasal swabs was detected 6 days post-challenge in vaccinated animals. After viral challenge, lower virus excretion and lower clinical score were observed in vaccinated as compared to unvaccinated animals, as well as BoHV-1-specific proliferation of lymphocytes and production of IFNγ, TNFα, and IL-4. Downregulation of the expression of endosome Toll-like receptors 8-9 was detected after booster vaccination. This is the first thorough study of the immunity generated by a commercial vaccine against BoHV-1 in cattle.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Herpesvirus 1, Bovine/immunology , Herpesvirus Vaccines/administration & dosage , Immunoglobulin G/biosynthesis , Infectious Bovine Rhinotracheitis/prevention & control , Toll-Like Receptor 8/immunology , Toll-Like Receptor 9/immunology , Adaptive Immunity/drug effects , Animals , Antibodies, Viral , Cattle , Cell Proliferation , Endosomes/immunology , Endosomes/metabolism , Gene Expression , Herpesvirus 1, Bovine/pathogenicity , Immunity, Innate/drug effects , Immunization, Secondary/methods , Infectious Bovine Rhinotracheitis/genetics , Infectious Bovine Rhinotracheitis/immunology , Infectious Bovine Rhinotracheitis/virology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocytes/immunology , Lymphocytes/virology , Male , Nasal Cavity/immunology , Nasal Cavity/virology , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/genetics , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccination/methods , Vaccines, Inactivated
3.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: mdl-33827944

ABSTRACT

Koi herpesvirus (KHV) is highly contagious and lethal to cyprinid fish, causing significant economic losses to the carp aquaculture industry, particularly to koi carp breeders. Vaccines delivered through intramuscular needle injection or gene gun are not suitable for mass vaccination of carp. So, the development of cost-effective oral vaccines that are easily applicable at a farm level is highly desirable. In this study, we utilized chitosan-alginate capsules as an oral delivery system for a live probiotic (Lactobacillus rhamnosus) vaccine, pYG-KHV-ORF81/LR CIQ249, expressing KHV ORF81 protein. The tolerance of the encapsulated recombinant Lactobacillus to various digestive environments and the ability of the probiotic strain to colonize the intestine of carp was tested. The immunogenicity and the protective efficacy of the encapsulated probiotic vaccine was evaluated by determining IgM levels, lymphocyte proliferation, expression of immune-related genes, and viral challenge to vaccinated fish. It was clear that the chitosan-alginate capsules protected the probiotic vaccine effectively against extreme digestive environments, and a significant level (P < 0.01) of antigen-specific IgM with KHV-neutralizing activity was detected, which provided a protection rate of ca. 85% for koi carp against KHV challenge. The strategy of using chitosan-alginate capsules to deliver probiotic vaccines is easily applicable for mass oral vaccination of fish.IMPORTANCE An oral probiotic vaccine, pYG-KHV-ORF81/LR CIQ249, encapsulated by chitosan-alginate capsules as an oral delivery system was developed for koi carp against koi herpesvirus (KHV) infection. This encapsulated probiotic vaccine can be protected from various digestive environments and maintain effectively high viability, showing a good tolerance to digestive environments. This encapsulated probiotic vaccine has a good immunogenicity in koi carp via oral vaccination, and a significant level of antigen-specific IgM was effectively induced after oral vaccination, displaying effective KHV-neutralizing activity. This encapsulated probiotic vaccine can provide effective protection for koi carp against KHV challenge, which is handling-stress free for the fish, cost effective, and suitable for the mass oral vaccination of koi carp at a farm level, suggesting a promising vaccine strategy for fish.


Subject(s)
Carps , Fish Diseases/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae/immunology , Herpesvirus Vaccines/administration & dosage , Probiotics , Viral Proteins/immunology , Administration, Oral , Alginates , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/immunology , Capsules , Cell Proliferation , Chitosan , Herpesviridae Infections/prevention & control , Herpesvirus Vaccines/immunology , Immunogenicity, Vaccine , Immunoglobulin M/blood , Lacticaseibacillus rhamnosus , Lymphocytes/physiology , Mass Vaccination/veterinary , Recombinant Fusion Proteins , Spleen/immunology , Spleen/metabolism , Vaccines, Synthetic/administration & dosage , Viral Proteins/genetics
4.
Value Health ; 24(1): 41-49, 2021 01.
Article in English | MEDLINE | ID: mdl-33431152

ABSTRACT

OBJECTIVES: Cost-effectiveness analyses (CEA) are based on the value judgment that health outcomes (eg, quantified in quality-adjusted life-years; QALYs) are all equally valuable irrespective of their context. Whereas most published CEAs perform extensive sensitivity analysis on various parameters and assumptions, only rarely is the influence of the QALY-equivalence assumption on cost-effectiveness results investigated. We illustrate how the integration of alternative social value judgments in CEA can be a useful form of sensitivity analysis. METHODS: Because varicella-zoster virus (VZV) vaccination affects 2 distinct diseases (varicella zoster and herpes zoster) and likely redistributes infections across different age groups, the program has an important equity dimension. We used a cost-effectiveness model and disentangled the share of direct protection and herd immunity within the total projected QALYs resulting from a 50-year childhood VZV program in the UK. We use the UK population's preferences for QALYs in the vaccine context to revalue QALYs accordingly. RESULTS: Revaluing different types of QALYs for different age groups in line with public preferences leads to a 98% change in the projected net impact of the program. The QALYs gained among children through direct varicella protection become more important, whereas the QALYs lost indirectly through zoster in adults diminish in value. Weighting of vaccine-related side effects made a large difference. CONCLUSIONS: Our study shows that a sensitivity analysis in which alternative social value judgments about the value of health outcomes are integrated into CEA of vaccines is relatively straightforward and provides important additional information for decision makers to interpret cost-effectiveness results.


Subject(s)
Cost-Benefit Analysis/methods , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/economics , Social Values , Varicella Zoster Virus Infection/prevention & control , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , Consumer Behavior , Decision Support Techniques , Herpesvirus Vaccines/adverse effects , Humans , Immunity, Herd , Infant , Middle Aged , Models, Economic , Quality-Adjusted Life Years , United Kingdom/epidemiology , Varicella Zoster Virus Infection/economics , Varicella Zoster Virus Infection/epidemiology , Young Adult
5.
Transbound Emerg Dis ; 68(2): 587-597, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32643286

ABSTRACT

New technologies in the field of vaccinology arise as a necessity for the treatment and control of many diseases. Whole virus inactivated vaccines and modified live virus ones used against Bovine Herpesvirus-1 (BoHV-1) infection have several disadvantages. Previous works on DNA vaccines against BoHV-1 have demonstrated the capability to induce humoral and cellular immune responses. Nevertheless, 'naked' DNA induces low immunogenic response. Thus, loading of antigen encoding DNA sequences in liposomal formulations targeting dendritic cell receptors could be a promising strategy to better activate these antigen-presenting cells (APC). In this work, a DNA-based vaccine encoding the truncated version of BoHV-1 glycoprotein D (pCIgD) was evaluated alone and encapsulated in a liposomal formulation containing LPS and decorated with MANα1-2MAN-PEG-DOPE (pCIgD-Man-L). The vaccinations were performed in mice and bovines. The results showed that the use of pCIgD-Man-L enhanced the immune response in both animal models. For humoral immunity, significant differences were achieved when total antibody titres and isotypes were assayed in sera. Regarding cellular immunity, a significant increase in the proliferative response against BoHV-1 was detected in animals vaccinated with pCIgD-Man-L when compared to the response induced in animals vaccinated with pCIgD. In addition, upregulation of CD40 molecules on the surface of bovine dendritic cells (DCs) was observed when cells were stimulated and activated with the vaccine formulations. When viral challenge was performed, bovines vaccinated with MANα1-2MAN-PEG-DOPE elicited better protection which was evidenced by a lower viral excretion. These results demonstrate that the dendritic cell targeting using MANα1-2MAN decorated liposomes can boost the immunogenicity resulting in a long-lasting immunity. Liposomes decorated with MANα1-2MAN-PEG-DOPE were tested for the first time as a DNA vaccine nanovehicle in cattle as a preventive treatment against BoHV-1. These results open new perspectives for the design of vaccines for the control of bovine rhinotracheitis.


Subject(s)
Cattle Diseases/prevention & control , Herpesviridae Infections/veterinary , Herpesvirus 1, Bovine/immunology , Herpesvirus Vaccines/administration & dosage , Vaccination/veterinary , Animals , Cattle , Herpesviridae Infections/prevention & control , Male , Mice , Vaccines, DNA/administration & dosage
6.
J Glob Health ; 10(1): 010404, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32257152

ABSTRACT

BACKGROUND: Epstein-Barr virus (EBV) is an important human pathogen; it infects >90% people globally and is linked to infectious mononucleosis and several types of cancer. Vaccines against EBV are in development. In this study we present the first systematic review of the literature on risk factors for EBV infection, and discuss how they differ between settings, in order to improve our understanding of EBV epidemiology and aid the design of effective vaccination strategies. METHODS: MEDLINE, Embase, and Web of Science were searched on 6th March 2017 for observational studies of risk factors for EBV infection. Studies were excluded if they were published before 2008 to ensure relevance to the modern day, given the importance of influencing future vaccination policies. There were no language restrictions. After title, abstract and full text screening, followed by checking the reference lists of included studies to identify further studies, data were extracted into standardised spreadsheets and quality assessed. A narrative synthesis was undertaken. RESULTS: Seventy-seven papers met our inclusion criteria, including data from 31 countries. There was consistent evidence that EBV seroprevalence was associated with age, increasing throughout childhood and adolescence and remaining constant thereafter. EBV was generally acquired at younger ages in Asia than Europe/North America. There was also compelling evidence for an association between cytomegalovirus infection and EBV. Additional factors associated with EBV seroprevalence, albeit with less consistent evidence, included ethnicity, socioeconomic status, other chronic viral infections, and genetic variants of HLA and immune response genes. CONCLUSIONS: Our study is the first systematic review to draw together the global literature on the risk factors for EBV infection and includes an evaluation of the quality of the published evidence. Across the literature, the factors examined are diverse. In Asia, early vaccination of infants would be required to prevent EBV infection. In contrast, in Western countries a vaccine could be deployed later, particularly if it has only a short duration of protection and the intention was to protect against infectious mononucleosis. There is a lack of high-quality data on the prevalence and age of EBV infection outside of Europe, North America and South-East Asia, which are essential for informing effective vaccination policies in these settings.


Subject(s)
Epstein-Barr Virus Infections/prevention & control , Herpesvirus 4, Human/immunology , Herpesvirus Vaccines/immunology , Infectious Mononucleosis/prevention & control , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/genetics , Humans , Infectious Mononucleosis/immunology , Infectious Mononucleosis/virology , Policy , Risk Factors , Vaccines
7.
Res Vet Sci ; 130: 184-192, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32199177

ABSTRACT

Cyprinid herpesvirus 3 (CyHV-3) is the etiological agent of koi herpersvirus disease (KHVD), which causes serious economic losses in global common carp and ornamental koi carp production of larvae as well as adult type fish. To control KHVD, vaccines against CyHV-3 utilizing different immunization routes have been developed, among them, oral vaccination is the most desirable method to prevent fish diseases occurring at the early larval stage. Here, we developed an oral subunit vaccine through the Saccharomyces cerevisiae cell surface display of CyHV-3 envelope protein pORF65, then, the recombinant yeast fed to Artemia which served as bio-encapsulation vector by subsequently feeding the common carp (Cyprinus carpio var. Jian) larvae. The fluorescent observation showed that the Artemia and S. cerevisiae could deliver intact antigen to the hindgut of carp larvae suggesting the possibility of the vector for oral immunization. On this basis, after three immunizations at a week interval, the oral vaccine induced high level of specific anti-pORF65 antibody. Meanwhile, a significant difference of immune-related genes expression occurred including cxca, IL-1ß, IFN-a1, lysozyme, IgM and CD8α between vaccined group and blank control group. In addition, 30% of relative percent survival of carp larvae after immunization was obtained post the animal infection assay, offered an certain immune protection. Our results indicated that the oral pORF65 subunit vaccine bioencapsulated in Artemia induced the activation of immune response and high level of antibodies, which could be served as an oral vaccine candidate for the prevention of CyHV-3 infection.


Subject(s)
Antigens, Viral/immunology , Fish Diseases/immunology , Herpesviridae Infections/veterinary , Herpesviridae/immunology , Herpesvirus Vaccines/administration & dosage , Vaccination/veterinary , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Artemia/chemistry , Herpesviridae Infections/immunology , Immunity, Innate/drug effects , Protective Agents/administration & dosage , Saccharomyces cerevisiae/chemistry , Vaccines, Synthetic/administration & dosage , Viral Envelope Proteins/immunology
8.
Appl Microbiol Biotechnol ; 104(7): 3011-3023, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32002602

ABSTRACT

Bovine herpesvirus type 1 (BoHV-1) causes considerable economic losses to the cow industry. Vaccination remains an effective strategy to control the diseases associated with BoHV-1. However, live vaccines present safety concerns, especially in pregnant cows; thus, nonreplicating vaccines have been developed to control the disease. The envelope glycoproteins of BoHV-1 induce a protective immune response. In this work, selected epitopes on glycoproteins gD, gC, and gB were constructed in triplicate with linker peptides. Vaccination of rabbits demonstrated that P2-gD/gC/gB with AAYAAY induced higher specific antibodies than that with GGGGS linker. P2-gD/gC/gB with AAYAAY linker was fused with bovine interleukin-6 (BoIL-6) or rabbit IL-6 (RaIL-6) and bacterially expressed. Rabbits were intramuscularly immunized with 100 µg of P2-gD/gC/gB-BoIL-6, P2-gD/gC/gB-RaIL-6, P2-gD/gC/gB, P2-gD/gC/gB plus BoIL-6, P2-(gD-a)3-BoIL-6, or P2-(gD-a)3 emulsified with ISA 206 adjuvant thrice at 3-week intervals. P2-gD/gC/gB-BoIL-6 generated a higher titer of BoHV-1-specific antibodies, neutralizing antibodies, interferon (IFN)-γ, and IL-4 compared with P2-gD/gC/gB plus BoIL-6, P2-gD/gC/gB-RaIL-6, or other formulation. P2-gD/gC/gB-BoIL-6 triggered similar levels of antibodies and significantly higher titer of IFN-γ and IL-4 compared with inactivated bovine viral diarrhea (BVD)-infectious bovine rhinotracheitis (IBR) vaccine. Rabbits vaccinated with P2-gD/gC/gB-BoIL-6 dramatically reduced viral shedding and tissue lesions in lungs and trachea after viral challenge and reactivation compared with those with P2-gD/gC/gB plus BoIL-6 or P2-gD/gC/gB-RaIL-6. P2-gD/gC/gB-BoIL-6 provided protective effects against viral shedding and tissue pathogenesis similar to those of the inactivated vaccine. The data confirmed the safety and immunogenicity of multiple-epitope recombinant protein and a potential vaccine candidate to control the disease, especially for pregnant cattle.


Subject(s)
Herpesviridae Infections/prevention & control , Herpesvirus 1, Bovine/immunology , Herpesvirus Vaccines/immunology , Vaccination/methods , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cattle , Cytokines/blood , Epitopes , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesvirus Vaccines/administration & dosage , Interleukin-6/genetics , Interleukin-6/immunology , Rabbits , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Virus Activation/drug effects , Virus Shedding/drug effects
9.
Fish Shellfish Immunol ; 98: 342-353, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31978531

ABSTRACT

Cyprinid Herpesvirus 3 (CyHV-3), also known as Koi Herpesvirus (KHV), causes Koi Herpesvirus Disease (KHVD) which leads to serious economic losses worldwide. To exploit DNA/subunit vaccine candidates, CyHV-3 ORF131 gene and cDNA was cloned and analyzed in the present study. Major B cell epitopes of deduced CyHV-3 pORF131 was also predicted. Then the complete CDS of CyHV-3 ORF131 was inserted into pEGFP-N1 vector and a modified pYD1/EBY100 system to construct the DNA and subunit vaccine, respectively. Subsequently, carp were immunized with homologous and heterologous prime-boost regimens relying on the constructed DNA and oral subunit vaccines. Then the protective immunity generated from different vaccines and regimens as well as the capacity of yeast (Saccharomyces cerevisiae) as an oral vaccine vehicle was evaluated. Our study confirmed that CyHV-3 ORF131 gene consisted of 2 introns and 3 exons encoding a 428 amino acids peptide. Further analysis indicated that four fragments of CyHV-3 pORF131 contained the major B cell epitopes (Cys20~Val140, Ser169~Tyr245, Thr258~Pro390, Phe414~Gln428), which could be linked and expressed in E. coli (BL21) as a truncated pORF131. The expression of full-length CyHV-3 pORF131 by pEGFP-N1 and yeast surface display was verified by In vitro assays before vaccination. Immunization of carp with CyHV-3 ORF131 DNA and subunit vaccines could evoke the activation of immune-related genes such as CXCa, CXCR1, IL-1ß, TNF-α, INF-a1, Mx-1, IgM, IgT1 and production of specific serum IgM measured by ELISA. RPS (relative percent of survival) ranging from 53.33% to 66.67% was acquired post challenge test. Moreover, flow cytometry analysis illustrated the delivery of surface-displayed CyHV-3 pORF131 to midgut after oral gavage. Thus, our findings suggest that CyHV-3 ORF131 can serve as DNA/subunit vaccines candidate and the yeast as an ideal oral vaccine vehicle.


Subject(s)
Carps , Fish Diseases/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae/immunology , Herpesvirus Vaccines/immunology , Open Reading Frames/immunology , Vaccination/veterinary , Administration, Oral , Animals , Antibodies, Viral/blood , Carps/immunology , Carps/virology , Cell Surface Display Techniques , Epitopes, B-Lymphocyte , Escherichia coli/genetics , Fish Diseases/virology , Gene Expression Regulation/immunology , Herpesviridae Infections/prevention & control , Herpesvirus Vaccines/administration & dosage , Immunization Schedule , Open Reading Frames/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Survival Analysis , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
10.
Fish Shellfish Immunol ; 98: 810-818, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31743761

ABSTRACT

Koi herpesvirus (KHV) also named Cyprinid Herpesvirus 3 (CyHV-3) is one of the most threatening pathogens affecting common carp production as well as the valued ornamental koi carp. The current commercial vaccines available are costly and potentially cause severe stress caused by live virus. KHV ORF149 gene has been proved encoding one of the main immunogenic proteins for KHV. In this study, we coupled a plasmid expression vector for ORF149 to single walled carbon nanotubes (SWCNTs) for an anti-KHV vaccine. The vaccine conferred an 81.9% protection against intraperitoneal challenge with KHV. Importantly, SWCNTs as a promising vehicle can enhanced the protective effects 33.9% over that of the naked DNA vaccine at the same dose. The protection was longer and serum antibody production, enzyme activities and immune-related gene expression were all induced in fish vaccinated with the nanotube-DNA vaccine compared with the DNA alone. Thereby, this study demonstrates that the ORF149 DNA vaccine loaded onto SWCNTs as a novel vaccine might provide an effective method of coping with KHV disease using intra-muscular vaccination.


Subject(s)
Carps , Fish Diseases/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae/immunology , Herpesvirus Vaccines/administration & dosage , Nanotubes, Carbon , Animals , Herpesviridae Infections/prevention & control , Injections, Intramuscular/veterinary , Vaccines, DNA/administration & dosage
11.
Vaccine ; 37(51): 7455-7462, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31590936

ABSTRACT

Bovine respiratory disease (BRD) remains a major health problem despite extensive use of vaccines during the post-weaning period. Apparent vaccine failure is attributed, in part, to primary vaccination during the period of greatest risk for BRD, providing inadequate time for onset of protective immunity. The current study investigated whether intranasal (IN) vaccination of 3-6 week old calves with a modified-live viral (MLV) vaccine induced sufficient immune memory to prevent respiratory disease and accelerate onset of protective immunity 5 months later. Vaccine groups included naïve controls, a single IN vaccination at 3-6 weeks of age, primary IN vaccination at 6 months, and either an IN or subcutaneous (SC) booster vaccination at 6 months (n = 10/group). All calves were challenged with BHV-1 four days after vaccination at 6 months of age. Primary IN vaccination at 6 months did not significantly reduce clinical disease but significantly (P < 0.01) reduced virus shedding. A single IN vaccination at 3-6 weeks of age significantly (P < 0.05) reduced weight loss but did not reduce fever or virus shedding. Both IN and SC booster vaccinations, significantly (P < 0.01) reduced clinical disease but virus shedding was significantly (P < 0.001) reduced only by IN booster vaccination. Reduction in virus shedding was significantly (P < 0.01) greater following booster versus primary IN vaccination at 6 months. All vaccination regimes significantly (P < 0.01) reduced secondary bacterial pneumonia and altered interferon responses relative to naïve controls. Only IN booster vaccination significantly (P < 0.05) increased BHV-1 specific IgA in nasal secretions. These results confirm primary MLV IN vaccination at 3 to 6 weeks of age, when virus neutralizing maternal antibody was present, induced immune memory with a 5 month duration. This immune memory supported rapid onset of protective immunity four days after an IN booster vaccination.


Subject(s)
Herpesvirus 1, Bovine/immunology , Herpesvirus Vaccines/administration & dosage , Immunization, Secondary/methods , Immunologic Memory/drug effects , Infectious Bovine Rhinotracheitis/prevention & control , Pneumonia, Bacterial/prevention & control , Administration, Intranasal , Animals , Animals, Newborn , Antibodies, Viral/blood , Cattle , Colostrum/chemistry , Colostrum/immunology , Female , Herpesvirus 1, Bovine/drug effects , Herpesvirus 1, Bovine/pathogenicity , Immunity, Mucosal/drug effects , Immunoglobulin A/blood , Infectious Bovine Rhinotracheitis/immunology , Infectious Bovine Rhinotracheitis/mortality , Infectious Bovine Rhinotracheitis/virology , Male , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/mortality , Pregnancy , Survival Analysis , Vaccination/methods , Vaccines, Attenuated , Viral Load/drug effects , Virus Shedding/drug effects , Weight Loss/drug effects
12.
Virology ; 531: 219-232, 2019 05.
Article in English | MEDLINE | ID: mdl-30928700

ABSTRACT

Equid herpesvirus-1 (EHV-1) outbreaks continue despite widely used vaccination. We demonstrated previously that an ORF1/ORF71 gene deletion mutant of the EHV-1 strain Ab4 (Ab4ΔORF1/71) is less virulent than its parent Ab4 virus. Here, we describe the Ab4 challenge infection evaluating protection induced by the Ab4ΔORF1/71 vaccine candidate. Susceptible control horses developed respiratory disease, fever, nasal shedding, and viremia. Full protection after challenge infection was observed in 5/5 previously Ab4 infected horses and 3/5 Ab4ΔORF1/71 horses. Two Ab4ΔORF1/71 horses developed short-lasting viremia and/or virus shedding. Protective immunity in the respiratory tract was characterized by pre-existing EHV-1-specific IgG4/7 antibodies, the absence of IFN-α secretion and rapidly increasing IgG4/7 upon challenge infection. Pre-existing systemic EHV-1-specific IgG4/7 highly correlated with protection. T-cell immunity was overall low. In conclusion, protective immunity against EHV-1 infection including prevention of viremia was associated with robust systemic and intranasal IgG4/7 antibodies suggesting immediate virus neutralization at the local site.


Subject(s)
Antibodies, Viral/immunology , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/immunology , Herpesvirus Vaccines/administration & dosage , Horse Diseases/prevention & control , Immunoglobulin G/immunology , Viremia/veterinary , Administration, Intranasal , Animals , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus 1, Equid/drug effects , Herpesvirus 1, Equid/genetics , Herpesvirus 1, Equid/physiology , Herpesvirus Vaccines/immunology , Horse Diseases/immunology , Horse Diseases/virology , Horses , Male , Nasal Mucosa/immunology , Nasal Mucosa/virology , Vaccination , Viremia/immunology , Viremia/prevention & control , Viremia/virology , Virus Shedding
13.
Vet Microbiol ; 222: 18-24, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30080668

ABSTRACT

Immune responses were compared after intranasal (IN) and intramuscular (IM) vaccination of horses with a modified live equine herpesvirus type-1 (EHV-1) vaccine, and the protective effect after EHV-1 challenge was evaluated. IN- and IM-vaccinated groups (n = 5 each) showed significant rises in serum virus-neutralizing titers with increased levels of IgGa and IgGb antibodies after the first vaccination (P < 0.05). In nasal secretions, the IN group had significantly increased levels of IgA antibodies after vaccination (P < 0.05), whereas the response of the IM group was dominated by IgGa and IgGb subclasses. After challenge infection, the numbers of pyretic horses from 1 to 4 days post-inoculation were 3/5 in the placebo (PBO) group (n = 5), 0/5 in the IN group, and 1/5 in the IM group. The IN and IM groups had significantly lower levels of virus shedding than the PBO group (P < 0.05). There were no significant between-group differences in the numbers of viremic horses each day. Notably, two horses in the IM group had no virus shedding or viremia, whereas all horses in the other group did. Both IN and IM vaccination induced systemic humoral immunity and mucosal immunity, suppressing virus replication in the nasal mucosa, and partially protected horses from pyrexia, especially early in infection. This study showed a mucosal antibody response was induced, not only by IN vaccination but also by IM vaccination.


Subject(s)
Administration, Intranasal/methods , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/immunology , Herpesvirus Vaccines/immunology , Injections, Intramuscular/methods , Vaccination/veterinary , Animals , Antibodies, Viral/blood , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus Vaccines/administration & dosage , Horse Diseases/immunology , Horse Diseases/prevention & control , Horse Diseases/virology , Horses , Immunity, Mucosal , Immunoglobulin A/blood , Immunoglobulin G/blood , Neutralization Tests , Vaccination/methods , Vaccines, Attenuated/administration & dosage , Virus Shedding/immunology
14.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29950407

ABSTRACT

We previously isolated a herpes simplex virus 1 (HSV-1) mutant, KOS-NA, that carries two nonsynonymous mutations in UL39, resulting in L393P and R950H amino acid substitutions in infected cell protein 6 (ICP6). Our published data studying KOS-NA pathogenesis strongly suggest that one of these ICP6 substitutions expressed from KOS-NA, R950H, severely impaired acute viral replication in the eyes and trigeminal ganglia of mice after inoculation onto the cornea and consequently impaired establishment and reactivation from latency. Because of its significant neuroattenuation, we tested KOS-NA as a potential prophylactic vaccine against HSV-1 in a mouse model of corneal infection. KOS-NA stimulated stronger antibody and T cell responses than a replication-competent ICP0-null mutant and a replication-incompetent ICP8-null mutant optimized for immunogenicity. Immunizations with the ICP0-, ICP8-, and KOS-NA viruses all reduced replication of wild-type HSV-1 challenge virus in the corneal epithelium to similar extents. Low immunizing doses of KOS-NA and the ICP8- virus, but not the ICP0- virus, protected mice against eyelid disease (blepharitis). Notably, only KOS-NA protected almost completely against corneal disease (keratitis) and greatly reduced latent infection by challenge virus. Thus, vaccination of mice with KOS-NA prior to corneal challenge provides significant protection against HSV-1-mediated disease of the eye, even at a very low immunizing dose. These results suggest that KOS-NA may be the foundation of an effective prophylactic vaccine to prevent or limit HSV-1 ocular diseases.IMPORTANCE HSV-1 is a ubiquitous human pathogen that infects the majority of the world's population. Although most infections are asymptomatic, HSV-1 establishes lifelong latency in infected sensory neurons, from which it can reactivate to cause deadly encephalitis or potentially blinding eye disease. No clinically effective vaccine is available. In this study, we tested the protective potential of a neuroattenuated HSV-1 mutant (KOS-NA) as a vaccine in mice. We compared the effects of immunization with KOS-NA to those of two other attenuated viruses, a replication-competent (ICP0-) virus and a replication-incompetent (ICP8-) virus. Our data show that KOS-NA proved superior to the ICP0- and ICP8-null mutants in protecting mice from corneal disease and latent infection. With its significant neuroattenuation, severe impairment in establishing latency, and excellent protective effect, KOS-NA represents a significant discovery in the field of HSV-1 vaccine development.


Subject(s)
Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/immunology , Keratitis, Herpetic/prevention & control , Viral Proteins/genetics , Animals , Chlorocebus aethiops , Disease Models, Animal , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , Mice , Mutation , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vero Cells , Viral Proteins/immunology , Virus Latency , Virus Replication
15.
Adv Exp Med Biol ; 1045: 123-142, 2018.
Article in English | MEDLINE | ID: mdl-29896666

ABSTRACT

Varicella-zoster virus (VZV) is the first and only human herpesvirus for which a licensed live attenuated vaccine, vOka, has been developed. vOka has highly safe and effective profiles; however, worldwide herd immunity against VZV has not yet been established and it is far from eradication. Despite the successful reduction in the burden of VZV-related illness by the introduction of the vaccine, some concerns about vOka critically prevent worldwide acceptance and establishment of herd immunity, and difficulties in addressing these criticisms often relate to its ill-defined mechanism of attenuation. Advances in scientific technologies have been applied in the VZV research field and have contributed toward uncovering the mechanism of vOka attenuation as well as VZV biology at the molecular level. A subunit vaccine targeting single VZV glycoprotein, rationally designed based on the virological and immunological research, has great potential to improve the strategy for eradication of VZV infection in combination with vOka.


Subject(s)
Herpesvirus 3, Human/immunology , Herpesvirus Vaccines/immunology , Varicella Zoster Virus Infection/prevention & control , Animals , Drug Design , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/physiology , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/genetics , Humans , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Varicella Zoster Virus Infection/immunology , Varicella Zoster Virus Infection/virology
16.
Adv Exp Med Biol ; 1045: 477-493, 2018.
Article in English | MEDLINE | ID: mdl-29896681

ABSTRACT

Epstein-Barr virus (EBV) is the primary cause of infectious mononucleosis and is associated with several malignancies, including nasopharyngeal carcinoma, gastric carcinoma, Hodgkin lymphoma, Burkitt lymphoma, and lymphomas in immunocompromised persons, as well as multiple sclerosis. A vaccine is currently unavailable. While monomeric EBV gp350 was shown in a phase 2 trial to reduce the incidence of infectious mononucleosis, but not the rate of EBV infection, newer formulations of gp350 including multimeric forms, viruslike particles, and nanoparticles may be more effective. A vaccine that also includes additional viral glycoproteins, lytic proteins, or latency proteins might improve the effectiveness of an EBV gp350 vaccine. Clinical trials to determine if an EBV vaccine can reduce the rate of infectious mononucleosis or posttransplant lymphoproliferative disease should be performed. The former is important since infectious mononucleosis can be associated with debilitating fatigue as well as other complications, and EBV infectious mononucleosis is associated with increased rates of Hodgkin lymphoma and multiple sclerosis. A vaccine to reduce EBV posttransplant lymphoproliferative disease would be an important proof of principle to prevent an EBV-associated malignancy. Trials of an EBV vaccine to reduce the incidence of Hodgkin lymphoma, multiple sclerosis, or Burkitt lymphoma would be difficult but feasible.


Subject(s)
Epstein-Barr Virus Infections/prevention & control , Herpesvirus 4, Human/immunology , Herpesvirus Vaccines/immunology , Animals , Clinical Trials, Phase II as Topic , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/genetics , Humans , Infectious Mononucleosis/immunology , Infectious Mononucleosis/prevention & control , Infectious Mononucleosis/virology
17.
J Virol ; 92(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29899091

ABSTRACT

Replication-competent controlled virus vectors were derived from the virulent herpes simplex virus 1 (HSV-1) wild-type strain 17syn+ by placing one or two replication-essential genes under the stringent control of a gene switch that is coactivated by heat and an antiprogestin. Upon activation of the gene switch, the vectors replicate in infected cells with an efficacy that approaches that of the wild-type virus from which they were derived. Essentially no replication occurs in the absence of activation. When administered to mice, localized application of a transient heat treatment in the presence of systemic antiprogestin results in efficient but limited virus replication at the site of administration. The immunogenicity of these viral vectors was tested in a mouse footpad lethal challenge model. Unactivated viral vectors-which may be regarded as equivalents of inactivated vaccines-induced detectable protection against lethality caused by wild-type virus challenge. Single activation of the viral vectors at the site of administration (rear footpads) greatly enhanced protective immune responses, and a second immunization resulted in complete protection. Once activated, vectors also induced far better neutralizing antibody and HSV-1-specific cellular immune responses than unactivated vectors. To find out whether the immunogenicity of a heterologous antigen was also enhanced in the context of efficient transient vector replication, a virus vector constitutively expressing an equine influenza virus hemagglutinin was constructed. Immunization of mice with this recombinant induced detectable antibody-mediated neutralization of equine influenza virus, as well as a hemagglutinin-specific cellular immune response. Single activation of viral replication resulted in a severalfold enhancement of these immune responses.IMPORTANCE We hypothesized that vigorous replication of a pathogen may be critical for eliciting the most potent and balanced immune response against it. Hence, attenuation/inactivation (as in conventional vaccines) should be avoided. Instead, the necessary safety should be provided by placing replication of the pathogen under stringent control and by activating time-limited replication of the pathogen strictly in an administration region in which pathology cannot develop. Immunization will then occur in the context of highly efficient pathogen replication and uncompromised safety. We found that localized activation in mice of efficient but limited replication of a replication-competent controlled herpesvirus vector resulted in a greatly enhanced immune response to the virus or an expressed heterologous antigen. This finding supports the above-mentioned hypothesis and suggests that the vectors may be promising novel agents worth exploring for the prevention/mitigation of infectious diseases for which efficient vaccination is lacking, in particular in immunocompromised patients.


Subject(s)
Drug Carriers , Genetic Vectors , Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/immunology , Hot Temperature , Influenza Vaccines/immunology , Virus Replication , Animals , Antibodies, Viral/blood , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/genetics , Immunity, Cellular , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
Vaccine ; 36(20): 2842-2849, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29655629

ABSTRACT

Herpes simplex virus is a common causative agent of oral and genital diseases. Novel vaccines and therapeutics are needed to combat herpes infections especially after the failure of subunit vaccines in human clinical trials. We have shown that the live-attenuated HSV-1 VC2 vaccine strain is unable to establish latency in vaccinated animals and produces a robust immune response capable of completely protecting mice against lethal vaginal HSV-1 or HSV-2 infections. The guinea pig represents the best small animal model of genital HSV-2 disease. Reported here, twenty-one female Hartley guinea pigs received intramuscular injection with either the VC2 vaccine, or equal volume of conditioned tissue culture media. Animals received 2 booster vaccinations at 21 day intervals following the initial vaccination. After vaccination, animals were challenged with the highly virulent HSV-2 (G) strain. Histologically, VC2 vaccinated animals had little to no apparent inflammation/disease following challenge. Unvaccinated animals developed moderate to severe erosive and ulcerative vaginitis. Quantitative reverse-transcriptase PCR analysis in VC2 vaccinated and challenged animals identified transcriptional signatures of Th17 and regulatory Tr1 cells associated with the inflammatory response primed by VC2 vaccination. Treatment of cultured human vaginal epithelial cells (VK2 cells) with a combination of IL-17A and IL-22 resulted in the significant induction of beta-defensin 3 expression. Further, treatment of VK2 cells with IL-17A, IL-22, IL-36 or beta-defensin 3 resulted in diminished HSV-2 replication. Overall, these results suggest that intramuscular vaccination with the live-attenuated vaccine VC2 primes a mucosal immune response predisposing the adaptive expression of transcripts associated with a Th17 response to challenge and these responses contribute to antiviral immunity.


Subject(s)
Herpes Genitalis/prevention & control , Herpesvirus 2, Human/immunology , Herpesvirus Vaccines/immunology , Injections, Intramuscular , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Vagina/immunology , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Gene Expression Profiling , Guinea Pigs , Herpes Genitalis/immunology , Herpesvirus Vaccines/administration & dosage , Histocytochemistry , Humans , Immunization Schedule , Mice , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vagina/pathology , beta-Defensins/analysis
19.
Vaccine ; 36(18): 2394-2402, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29602705

ABSTRACT

Herpes simplex virus is a prevalent pathogen of humans of various age groups. The fact that no prophylactic or therapeutic vaccine is currently available suggests a significant need to further investigate the immune mechanisms induced by the virus and various vaccine candidates. We previously generated an HSV-1 mutant strain, M3, with partial deletions in ul7, ul41 and LAT that produced an attenuated phenotype in mice. In the present study, we performed a comparative analysis to characterize the immune responses induced by M3 versus wild-type HSV-1 in a mouse model. Infection with wild-type HSV-1 triggered an inflammatory-dominated response and adaptive immunity suppression and was accompanied by severe pathological damage. In contrast, infection with M3 induced a systematic immune response involving full activation of both innate and adaptive immunity and was accompanied by no obvious pathological changes. Furthermore, the immune response induced by M3 protected mice from lethal challenge with wild-type strains of HSV-1 and restrained virus proliferation and impaired latency. These data are useful for further HSV-1 vaccine development using a mutant strain construction strategy.


Subject(s)
Adaptive Immunity , Gene Expression Profiling , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Herpesvirus Vaccines/immunology , Immunity, Innate , Animals , Disease Models, Animal , Female , Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/administration & dosage , Immune Evasion , Mice, Inbred BALB C , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL