Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Avian Dis ; 63(2): 351-358, 2019 06 01.
Article En | MEDLINE | ID: mdl-31251537

Infectious laryngotracheitis (ILT) is a highly contagious respiratory disease of chickens that produces significant economic losses to the poultry industry. The disease is caused by Gallid alpha herpesvirus-1 (GaHV-1), commonly known as the infectious laryngotracheitis virus (ILTV). Vaccination remains necessary for the control of the disease. Due to the inherent virulence of live attenuated vaccines, in particular that of the chicken embryo origin (CEO) vaccines, the use of ILT viral vector recombinant vaccines has significantly expanded worldwide as a safer vaccination strategy. However, the protective efficacy of recombinant ILT vaccines can be compromised by the use of fractional doses and improper handling and administration of the vaccine. The objective of this study was twofold: 1) to evaluate the protection efficacy induced by a commercial recombinant HVT-LT (rHVT-LT) vaccine when administered in ovo to broilers at three standardized doses (6000 plaque-forming units [PFU], 3000 PFU, and 1000 PFU), and 2) to assess the potential of rHVT-LT-vaccinated chickens to spread virus to contact chickens after challenge. Independently of the vaccine dose, vaccinated chickens showed reduction in clinical signs, maintained body weight gain after challenge, and lessened the challenge virus replication in the trachea at a rate of 52%-65%. However, in spite of this reduction, transmission of challenge virus from rHVT-LT-vaccinated (6000/Ch, 3000/Ch) to contact-naive chickens was evident. This study is the first to support that rHVT-LT vaccination did not prevent spread of challenge virus to contact birds.


Eficacia de la protección de una vacuna con un herpesvirus de los pavos (HVT) recombinante contra el virus de la laringotraqueitis infecciosa (ILTV) administrada in ovo en pollos de engorde en tres dosis estandarizadas. La laringotraqueítis infecciosa (ILT, por sus siglas en inglés) es una enfermedad respiratoria altamente contagiosa de los pollos que produce importantes pérdidas económicas para la industria avícola. La enfermedad es causada por el alfa herpesvirus-1 del pollo (GaHV-1), conocido comúnmente como el virus de la laringotraqueitis infecciosa (ILTV). La vacunación sigue siendo necesaria para el control de la enfermedad. Debido a la virulencia inherente de las vacunas atenuadas vivas, en particular la de las vacunas con origen embrion de pollo (CEO), el uso de vacunas contra la laringotraqueítis con vectores virales recombinantes se ha extendido significativamente en todo el mundo como una estrategia de vacunación más segura. Sin embargo, la eficacia protectora de las vacunas recombinantes contra la laringotraqueítis puede verse comprometida por el uso de dosis fraccionarias y por el manejo y administración inadecuados de la vacuna. El objetivo de este estudio fue doble: 1) evaluar la eficacia de la protección inducida por una vacuna comercial recombinante HVT-LT (rHVT-LT) cuando se administró in ovo en pollos de engorde en tres dosis estandarizadas (6000 unidades formadoras de placa [PFU], 3000 PFU y 1000 PFU), y 2) para evaluar el potencial de los pollos vacunados con rHVT-LT para propagar el virus a los pollos en contacto después del desafío. Independientemente de la dosis de la vacuna, los pollos vacunados mostraron una reducción en los signos clínicos, mantuvieron el aumento de peso corporal después del desafío y disminuyeron la replicación del virus de desafío en la tráquea a una tasa de 52% -65%. Sin embargo, a pesar de esta reducción, la transmisión del virus de desafío de los pollos vacunados con rHVT-LT con 6000 unidades formadoras de placa y desafiados o con 3000 unidades formadoras de placa y también desafiados a los pollos susceptibles en contacto fue evidente. Este estudio es el primero en demostrar que la vacunación con rHVT-LT no impidió la propagación del virus de desafío a las aves en contacto.


Chickens , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/immunology , Herpesvirus Vaccines/pharmacology , Poultry Diseases/prevention & control , Animals , Dose-Response Relationship, Immunologic , Herpesviridae Infections/prevention & control , Ovum/immunology , Vaccines, Synthetic/pharmacology
2.
Front Immunol ; 10: 500, 2019.
Article En | MEDLINE | ID: mdl-30949169

Herpes simplex virus type-2 (HSV-2) is a common cause of genital infections throughout the world. Currently no prophylactic vaccine or therapeutic cure exists against the virus that establishes a latent infection for the life of the host. Intravaginal microbivac is a developing out-of-the-box strategy that combines instant microbicidal effects with future vaccine-like benefits. We have recently shown that our uniquely designed zinc oxide tetrapod nanoparticles (ZOTEN) show strong microbivac efficacy against HSV-2 infection in a murine model of genital infection. In our attempts to further understand the antiviral and immune bolstering effects of ZOTEN microbivac and to develop ZOTEN as a platform for future live virus vaccines, we tested a ZOTEN/HSV-2 cocktail and found that prior incubation of HSV-2 with ZOTEN inhibits the ability of the virus to infect vaginal tissue in female Balb/c mice and blocks virus shedding as judged by plaque assays. Quite interestingly, the ZOTEN-neutralized virions elicit a local immune response that is highly comparable with the HSV-2 infection alone with reduced inflammation and clinical manifestations of disease. Information provided by our study will pave the way for the further development of ZOTEN as a microbivac and a future platform for live virus vaccines.


Administration, Intravaginal , Herpes Genitalis , Herpesvirus 2, Human/immunology , Herpesvirus Vaccines , Nanoparticles/chemistry , Animals , Female , Herpes Genitalis/immunology , Herpes Genitalis/pathology , Herpes Genitalis/prevention & control , Herpesvirus Vaccines/chemistry , Herpesvirus Vaccines/pharmacology , Mice , Zinc Oxide/chemistry
3.
J Virol ; 93(9)2019 05 01.
Article En | MEDLINE | ID: mdl-30787156

Reactivation of herpes simplex virus 2 (HSV-2) from latency causes viral shedding that develops into recurrent genital lesions. The immune mechanisms of protection against recurrent genital herpes remain to be fully elucidated. In this preclinical study, we investigated the protective therapeutic efficacy, in the guinea pig model of recurrent genital herpes, of subunit vaccine candidates that were based on eight recombinantly expressed HSV-2 envelope and tegument proteins. These viral protein antigens (Ags) were rationally selected for their ability to recall strong CD4+ and CD8+ T-cell responses from naturally "protected" asymptomatic individuals, who, despite being infected, never develop any recurrent herpetic disease. Out of the eight HSV-2 proteins, the envelope glycoprotein D (gD), the tegument protein VP22 (encoded by the UL49 gene), and ribonucleotide reductase subunit 2 protein (RR2; encoded by the UL40 gene) produced significant protection against recurrent genital herpes. The RR2 protein, delivered either intramuscularly or intravaginally with CpG and alum adjuvants, (i) boosted the highest neutralizing antibodies, which appear to cross-react with both gB and gD, and (ii) enhanced the numbers of functional gamma interferon (IFN-γ)-producing CRTAM+ CFSE+ CD4+ and CRTAM+ CFSE+ CD8+ TRM cells, which express low levels of PD-1 and TIM-3 exhaustion markers and were localized to healed sites of the vaginal mucocutaneous (VM) tissues. The strong B- and T-cell immunogenicity of the RR2 protein was associated with a significant decrease in virus shedding and a reduction in both the severity and frequency of recurrent genital herpes lesions. In vivo depletion of either CD4+ or CD8+ T cells significantly abrogated the protection. Taken together, these preclinical results provide new insights into the immune mechanisms of protection against recurrent genital herpes and promote the tegument RR2 protein as a viable candidate Ag to be incorporated in future genital herpes therapeutic mucosal vaccines.IMPORTANCE Recurrent genital herpes is one of the most common sexually transmitted diseases, with a global prevalence of HSV-2 infection predicted to be over 536 million worldwide. Despite the availability of many intervention strategies, such as sexual behavior education, barrier methods, and the costly antiviral drug treatments, eliminating or at least reducing recurrent genital herpes remains a challenge. Currently, no FDA-approved therapeutic vaccines are available. In this preclinical study, we investigated the immunogenicity and protective efficacy, in the guinea pig model of recurrent genital herpes, of subunit vaccine candidates that were based on eight recombinantly expressed herpes envelope and tegument proteins. We discovered that similar to the dl5-29 vaccine, based on a replication-defective HSV-2 mutant virus, which has been recently tested in clinical trials, the RR2 protein-based subunit vaccine elicited a significant reduction in virus shedding and a decrease in both the severity and frequency of recurrent genital herpes sores. This protection correlated with an increase in numbers of functional tissue-resident IFN-γ+ CRTAM+ CFSE+ CD4+ and IFN-γ+ CRTAM+ CFSE+ CD8+ TRM cells that infiltrate healed sites of the vaginal tissues. Our study sheds new light on the role of TRM cells in protection against recurrent genital herpes and promotes the RR2-based subunit therapeutic vaccine to be tested in the clinic.


Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Herpes Genitalis/prevention & control , Herpesvirus 2, Human/immunology , Herpesvirus Vaccines/pharmacology , Immunization, Secondary , Ribonucleotide Reductases/pharmacology , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Female , Guinea Pigs , Herpes Genitalis/immunology , Herpes Genitalis/pathology , Herpesvirus Vaccines/immunology , Humans , Immunity, Mucosal/drug effects , Male , Middle Aged , Ribonucleotide Reductases/immunology
4.
Protein Expr Purif ; 130: 21-27, 2017 02.
Article En | MEDLINE | ID: mdl-27693624

Bovine herpesvirus (BoHV) glycoprotein E (gE) is a non-essential envelope glycoprotein and the deletion of gE has been used to develop BoHV-1 and BoHV-5 differential vaccine strains. The DIVA (Differentiation of Infected from Vaccinated Animals) strategy, using marker vaccines based on gE-negative BoHV strains, allows the identification of vaccinated or infected animals in immunoassays designed to detect anti-gE antibodies. In this study a codon optimized synthetic sequence of gE containing highly conserved regions from BoHV-1 and BoHV-5 was expressed in Pichia pastoris. Following expression, the recombinant gE (rgE) was secreted and purified from the culture medium. The rgE was identified by Western blotting (WB) using sera from cattle naturally infected with BoHV-1 and/or BoHV-5, or sera from bovines experimentally infected with wild-type BoHV-5. Sera collected from cattle vaccinated with a BoHV-5 gI/gE/US9¯ marker vaccine failed to recognise rgE. Expression of rgE, based on a sequence containing highly conserved regions from BoHV-1 and BoHV-5, in P. pastoris enabled the production of large quantities of rgE suitable for use in immunoassays for the differentiation vaccinated or infected cattle.


Gene Expression , Herpesvirus 1, Bovine/genetics , Herpesvirus 5, Bovine , Infectious Bovine Rhinotracheitis , Pichia/metabolism , Viral Envelope Proteins , Viral Proteins , Animals , Cattle , Herpesvirus 1, Bovine/metabolism , Herpesvirus Vaccines/pharmacology , Infectious Bovine Rhinotracheitis/blood , Infectious Bovine Rhinotracheitis/diagnosis , Pichia/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/isolation & purification , Viral Envelope Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/isolation & purification , Viral Proteins/metabolism
5.
Pediatr Int ; 58(6): 534-6, 2016 Jun.
Article En | MEDLINE | ID: mdl-27322864

Herpes zoster (HZ) is an emerging concern for public health officials. The aim of this study was to determine whether universal Varicella immunization implemented in 2004 had an impact on HZ hospitalization in immunocompetent children in Greece. All HZ hospitalizations were recorded during the period 1999-2011. The overall attributable hospitalization rate was 13.89 cases/1000 hospital admissions (95%CI: 11.69-16.38 cases/1000 hospital admissions). HZ hospitalization rate remained unchanged during the study period. These data provide a basis for monitoring HZ hospitalization rate among children following universal toddler immunization.


Herpes Zoster/therapy , Herpesvirus 3, Human/immunology , Herpesvirus Vaccines/pharmacology , Hospitalization/trends , Immunization/methods , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Greece/epidemiology , Herpes Zoster/epidemiology , Humans , Incidence , Infant , Male , Retrospective Studies , Time Factors
6.
Vet J ; 201(2): 202-6, 2014 Aug.
Article En | MEDLINE | ID: mdl-24873978

The administration of intranasal (IN) or subcutaneous (SC) vaccines containing modified live feline herpesvirus 1 (FHV-1) offers some level of protection against FHV-1 challenge, but relative efficacy is <100%. In this study, clinical signs and viral shedding in kittens were compared among three groups: (1) kittens vaccinated concurrently with IN and SC vaccines containing FHV-1 (Group 1, n = 8); (2) kittens vaccinated with a SC FHV-1 vaccine alone (Group 2, n = 8), and (3) unvaccinated control kittens (Group 3, n = 8). All kittens were FHV-1 naïve at enrolment, and challenge with a virulent strain of FHV-1 was performed 1 week after vaccination. Daily clinical signs and pharyngeal FHV-1 shedding were recorded over a 21-day infection period. Overall, kittens in Group 1 had significantly less severe clinical illness than those in Group 2 (P < 0.05). Additionally, significantly less FHV-1 DNA was detected on pharyngeal swabs from kittens in Group 1 compared to those in Group 2 (P < 0.001). Concomitant administration of IN and SC FHV-1 vaccines was superior to administration of the SC FHV-1 vaccine alone in this challenge model of FHV-1 naïve kittens.


Cat Diseases/therapy , Herpesviridae Infections/veterinary , Herpesvirus Vaccines/pharmacology , Varicellovirus/physiology , Administration, Intranasal/veterinary , Animals , Cat Diseases/virology , Cats , Female , Herpesviridae Infections/therapy , Herpesviridae Infections/virology , Herpesvirus Vaccines/administration & dosage , Injections, Subcutaneous/veterinary , Male , Varicellovirus/immunology , Virus Shedding
7.
J Virol ; 83(22): 11734-45, 2009 Nov.
Article En | MEDLINE | ID: mdl-19726497

Some human herpesviruses (HHV) are etiological contributors to a wide range of malignant diseases. These HHV express latent membrane proteins (LMPs), which are type III membrane proteins consistently exposed at the cell surface in these malignancies. These LMPs have relatively large cytoplasmic domains but only short extracellular loops connecting transmembrane segments that are accessible at the surface of infected cells, but they do not elicit antibodies in the course of natural infection and tumorigenesis. We report here that conformational peptides mimicking two adjacent loops of the Epstein-Barr virus (EBV) LMP1 (2LS peptides) induce high-affinity antibodies with remarkable antitumor activities in mice. In active immunization experiments, LMP1-targeting 2LS vaccine conferred tumor protection in BALB/c mice. Moreover, this tumor protection is dependent upon a humoral anti-2LS immune response as demonstrated in DO11.10 (TCR-OVA) mice challenged with LMP1-expressing tumor and in SCID mice xenografted with human EBV-positive lymphoma cells. These data provide a proof of concept for 2LS immunization against short external loops of viral LMPs. This approach might possibly be extended to other infectious agents expressing type III membrane proteins.


Antibodies, Viral/therapeutic use , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Herpesvirus Vaccines/pharmacology , Viral Matrix Proteins/immunology , Animals , Cell Line , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Vaccination
8.
J Reprod Immunol ; 78(1): 49-57, 2008 Jun.
Article En | MEDLINE | ID: mdl-17945349

Toll-like receptors (TLRs) play an important role as pattern-recognition receptors to sense and respond to pathogens. Our laboratory and others have shown recently that activation of TLR/MyD88 signaling through vaginal administration of CpG oligodeoxynucleotides, either singly or in combination with recombinant glycoprotein from herpes simplex virus type 2 (HSV-2), confers immunity against genital herpes infection. In this study, we have investigated the importance of the myeloid differentiation factor 88 (MyD88), a critical adaptor protein shared by all TLRs, in innate and acquired immunity against genital HSV-2 infection in mice. We demonstrate that MyD88 is essential for innate immune resistance against HSV-2. Thus, MyD88 deficient (MyD88(-/-)) mice show more vaginal HSV-2 titers, more rapid disease progression and earlier death compared to C57Bl/6 mice following a vaginal challenge with high (9 x 10(4) PFU) or low (9 x 10(3) PFU) virus dose. In contrast, use of MyD88 appears dispensable for induction of HSV-specific serum IgG antibody as well as local and systemic cell-mediated immune responses elicited by vaginal immunization with live attenuated thymidine kinase-deficient HSV-2 (HSV-2 TK(-)). Importantly, and similar to immunized C57Bl/6 mice, immunized MyD88(-/-) mice were completely protected against subsequent vaginal challenge with a lethal dose of virulent strain of HSV-2. These results provide evidence that the adaptor protein MyD88 is important for innate early control of genital HSV-2 infection, and that use of MyD88 is not required for induction of acquired immunity following vaginal immunization with HSV-2 TK(-).


Herpes Genitalis/immunology , Herpesvirus 2, Human/immunology , Immunity, Innate/physiology , Myeloid Differentiation Factor 88/immunology , Animals , Antibodies, Viral/immunology , Female , Herpesvirus Vaccines/pharmacology , Immunity, Cellular/drug effects , Immunity, Cellular/physiology , Immunity, Innate/drug effects , Immunization , Immunoglobulin G/immunology , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/pharmacology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Vaccines, Attenuated/pharmacology
9.
Vaccine ; 25(46): 7927-30, 2007 Nov 14.
Article En | MEDLINE | ID: mdl-17920733

Caprine herpesvirus 1 provides a unique virus-animal model to investigate potential tools applicable for the therapy and prophylaxis of genital herpesvirus infections of humans. In order to evaluate the efficacy of mucosal immunization in the goat model, an inactivated CpHV-1 vaccine was adjuvated with the enzymatically inactive mutant of the heat-labile enterotoxin of Escherichia coli, LTK63, and used to immunize goats by the vaginal route, by administering two doses at a 3-week interval. The mucosal vaccine was safe, as neither local nor systemic reactions were associated with the vaccine administration. The vaccinated animals displayed high levels of secretory IgA and were significantly protected after challenge with the virulent CpHV-1 strain, with marked decrease in virus shedding, while the unvaccinated goats were not. These findings suggest that mucosal immunization is potentially exploitable in the control of genital infection by herpesviruses.


Adjuvants, Immunologic/pharmacology , Bacterial Toxins/pharmacology , Enterotoxins/pharmacology , Escherichia coli Proteins/pharmacology , Goat Diseases/prevention & control , Herpes Genitalis/prevention & control , Herpesvirus Vaccines/pharmacology , Immunity, Mucosal/drug effects , Mutation , Varicellovirus/immunology , Adjuvants, Immunologic/genetics , Animals , Antibodies, Viral/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Disease Models, Animal , Enterotoxins/genetics , Enterotoxins/immunology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Goat Diseases/genetics , Goat Diseases/immunology , Goats , Herpes Genitalis/genetics , Herpes Genitalis/immunology , Herpes Genitalis/veterinary , Herpesvirus Vaccines/immunology , Humans , Immunoglobulin A/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/pharmacology , Virus Shedding/drug effects , Virus Shedding/immunology
10.
Vaccine ; 25(20): 3934-45, 2007 May 16.
Article En | MEDLINE | ID: mdl-17433507

Efficient vaccines against AIDS, Hepatitis C and other persistent virus infections are urgently needed. Vaccine development has been especially hampered by the lack of suitable small animal models to reliably test the protective capacity of candidate vaccines against such chronic viral infections. A natural mouse pathogen such as MHV-68 that persists lifelong after infection, appears to be a particularly promising candidate for a more relevant model system. Here, we investigated infections with recombinant MHV-68 as novel mouse challenge model to test the efficacy of heterologous vaccines based on recombinant modified vaccinia virus Ankara (MVA). To apply ovalbumin (OVA) as a model antigen, we constructed the recombinant virus MHV-68-OVA by BAC technology and characterized genetic stability and replicative capacity of the virus in vitro and in vivo. We demonstrated the ability of MHV-68-OVA to produce ovalbumin upon tissue culture infection. Moreover, the use of MHV-68-OVA-infected target cells allowed for efficient ex vivo amplification of OVA-specific, MHC class I-restricted CD8 T cells derived from MVA-OVA-vaccinated C57BL/6 mice. Finally, we immunized C57BL/6 mice with MVA-OVA and challenged the animals with MHV-68-OVA testing different time points and routes of infection. Vaccinated mice were infected with MHV-68-OVA but showed reduced viral loads in the acute and latent phase of challenge infection. These data strongly suggest the usefulness of the MHV-68 challenge model for further evaluation of recombinant vaccines against persisting virus infections.


Gammaherpesvirinae/immunology , Herpesviridae Infections/prevention & control , Herpesvirus Vaccines/pharmacology , Ovalbumin/immunology , Vaccinia virus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Chickens , Disease Models, Animal , Gammaherpesvirinae/genetics , Gammaherpesvirinae/growth & development , Genes, MHC Class I/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus Vaccines/genetics , Herpesvirus Vaccines/immunology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology , Vaccinia virus/genetics , Viral Load
11.
Vet Immunol Immunopathol ; 116(1-2): 79-84, 2007 Mar 15.
Article En | MEDLINE | ID: mdl-17275918

Despite recent technological advances in vaccine production, most vaccines depend on the association with adjuvant substances. In this study, propolis, which has been attracting the attention of researchers due to its bioactive properties, was evaluated as an immunological adjuvant. The association of 40mg/dose of an ethanolic extract of green propolis with an inactivated oil vaccine against bovine herpesvirus type 5 (BoHV-5), resulted in a significant increase (P<0.01) in the neutralizing antibody levels, comparing to the bovines that received the same vaccine without propolis. Besides, propolis increased the percentage of animals with high antibody titers (above 32). Phenolic compounds such as artepillin C (3,5-diprenyl-4-hydroxycinnamic acid) and the derivatives of cinnamic acid besides other flavonoid substances were abundant in the propolis extract used, and they could be the main substances with adjuvant action. The effect of the green propolis extract on the humoral immune response can be exploited in the development of new vaccines.


Adjuvants, Immunologic/pharmacology , Cattle Diseases/immunology , Encephalitis, Viral/veterinary , Herpesviridae Infections/veterinary , Herpesvirus 5, Bovine/immunology , Herpesvirus Vaccines/immunology , Meningoencephalitis/veterinary , Propolis/pharmacology , Animals , Antibodies, Viral/blood , Cattle , Cattle Diseases/prevention & control , Cattle Diseases/virology , Encephalitis, Viral/immunology , Encephalitis, Viral/prevention & control , Encephalitis, Viral/virology , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus Vaccines/pharmacology , Immunization/methods , Immunization/veterinary , Male , Meningoencephalitis/immunology , Meningoencephalitis/prevention & control , Meningoencephalitis/virology , Neutralization Tests/veterinary , Propolis/immunology
12.
J Virol ; 77(17): 9463-73, 2003 Sep.
Article En | MEDLINE | ID: mdl-12915561

The molecular characterization of the epitope repertoire on herpes simplex virus (HSV) antigens would greatly expand our knowledge of HSV immunity and improve immune interventions against herpesvirus infections. HSV glycoprotein D (gD) is an immunodominant viral coat protein and is considered an excellent vaccine candidate antigen. By using the TEPITOPE prediction algorithm, we have identified and characterized a total of 12 regions within the HSV type 1 (HSV-1) gD bearing potential CD4(+) T-cell epitopes, each 27 to 34 amino acids in length. Immunogenicity studies of the corresponding medium-sized peptides confirmed all previously known gD epitopes and additionally revealed four new immunodominant regions (gD(49-82), gD(146-179), gD(228-257), and gD(332-358)), each containing naturally processed epitopes. These epitopes elicited potent T-cell responses in mice of diverse major histocompatibility complex backgrounds. Each of the four new immunodominant peptide epitopes generated strong CD4(+) Th1 T cells that were biologically active against HSV-1-infected bone marrow-derived dendritic cells. Importantly, immunization of H-2(d) mice with the four newly identified CD4(+) Th1 peptide epitopes but not with four CD4(+) Th2 peptide epitopes induced a robust protective immunity against lethal ocular HSV-1 challenge. These peptide epitopes may prove to be important components of an effective immunoprophylactic strategy against herpes.


Antigens, Viral/genetics , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Immunodominant Epitopes/genetics , Th1 Cells/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Algorithms , Amino Acid Sequence , Animals , Antigens, Viral/chemistry , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Herpesvirus Vaccines/genetics , Herpesvirus Vaccines/pharmacology , Immunization , Immunodominant Epitopes/chemistry , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data
13.
Curr Eye Res ; 26(3-4): 185-94, 2003.
Article En | MEDLINE | ID: mdl-12815546

PURPOSE: To quantify and characterize immune protection from herpes simplex virus (HSV) latent infection in mice following corneal challenge. METHODS: Mice immunized or mock-immunized and boosted in the flank with an HSV replication-deficient mutant were challenged by corneal inoculation with wild type (wt) or thymidine kinase-negative (TK(-)) HSV. At specified times post challenge, trigeminal ganglia were assayed for in vitro reactivation, latent and acute viral load (using quantitative PCR), acute infection, and cellular infiltration (hematoxylin and eosin stained sections). RESULTS: With wt HSV challenge infection, immunization led to reduced reactivation, significantly less latent and acute viral DNA, and no acute viral replication in ganglia, and rapid infiltration of inflammatory cells. Immunization had little effect on viral load following challenge with replication-conditional TK(-) mutant virus. CONCLUSION: These results indicate that immune protection from latent HSV infection in mouse trigeminal ganglia following ocular infection can act under these experimental conditions to block acute viral replication in ganglia and is directed to antigenic targets within the ganglia.


Herpes Simplex/prevention & control , Herpesvirus 1, Human , Immunization , Trigeminal Ganglion/virology , Virus Latency , Animals , Eye/virology , Female , Herpes Simplex/pathology , Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Neuritis/pathology , Thymidine Kinase/deficiency , Thymidine Kinase/genetics , Time Factors , Trigeminal Ganglion/immunology , Trigeminal Ganglion/pathology , Viral Load , Virus Activation , Virus Replication/drug effects , Virus Replication/genetics
14.
Int Rev Immunol ; 22(1): 43-63, 2003.
Article En | MEDLINE | ID: mdl-12710503

Herpes simplex virus type 2 (HSV-2) is a sexually transmitted pathogen that infects the genital tract. Efforts to develop vaccines to protect women against this and other sexually transmitted pathogens would be facilitated by a better understanding of the immune mechanisms that protect the female reproductive tract against such infections. Such information would be invaluable in developing vaccine strategies to promote the type and magnitude of immune responses in the genital tract that would effectively protect against infection. This review focuses on recent studies using a progestin-treated adult mouse model to explore mucosal immunity to HSV-2 in the vagina. Evidence indicating a major role for both humoral and T cell immunity is presented.


Herpes Genitalis/immunology , Herpesvirus 2, Human/immunology , Vagina/immunology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Female , Herpes Genitalis/prevention & control , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/pharmacology , Humans , Immunity, Cellular , Immunity, Mucosal , Immunoglobulin A, Secretory/metabolism , Immunoglobulin G/blood , Immunoglobulin G/metabolism , Interferon-gamma/metabolism , Lymphocytes/immunology , Mice , Mice, Knockout
15.
Vaccine ; 19(23-24): 3076-90, 2001 Apr 30.
Article En | MEDLINE | ID: mdl-11312002

The epidemiology of herpes zoster and post-herpetic neuralgia (PHN) was quantified from a variety of data sources and the potential cost-effectiveness of vaccination assessed. The annual incidence and severity of zoster increases sharply with age, as measured by physician consultation and hospitalisation rates, average length of stay, average proportion of cases developing PHN and the age-specific case-fatality ratio. Combining these data with information on health related quality of life results in an estimated loss of 20000 quality adjusted life years (QALYs) annually in England and Wales from herpes zoster (17400 due to PHN). The current cost of treating herpes zoster associated disease is estimated to be 47.6m pounds annually. Since both the health and economic burden are high, vaccination of the elderly is expected to be cost-effective under most scenarios, the attractiveness of immunisation increasing with age due to the increased burden of disease in the very elderly.


Herpes Zoster/epidemiology , Herpes Zoster/prevention & control , Herpesvirus Vaccines/economics , Herpesvirus Vaccines/pharmacology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , Cost-Benefit Analysis , England/epidemiology , Herpes Zoster/economics , Herpes Zoster/immunology , Humans , Infant , Infant, Newborn , Middle Aged , Models, Economic , Sensitivity and Specificity , Wales/epidemiology
16.
Vaccine ; 19(11-12): 1538-46, 2001 Jan 08.
Article En | MEDLINE | ID: mdl-11163680

To evaluate the utility of plasmid DNA vaccination against disease caused by herpes simplex virus (HSV), we compared the strength of protection against lethal challenge following natural virus infection with that following vaccination with a plasmid encoding HSV glycoprotein gD (gD-DNA). We further determined the cellular basis of each type of protection using lymphocyte deficient knockout mice. Establishment of immunity to HSV using live virus immunization required CD8+ T cells and B cells, but not CD4+ or gamma/delta+ T cells, and was related to specific antibody levels; surprisingly, CD4 knockout mice had large quantities of IgG anti-HSV serum antibodies. Establishment of immunity to HSV using gD-DNA immunization approached the strength of that generated following sublethal infection, but was dependent on alpha/beta+ CD4+ T cells, CD8+ T cells, B cells, and even partially on gamma/delta+ T cells, and not strictly correlated with antibody levels.


Herpes Simplex/immunology , Herpes Simplex/prevention & control , Herpesvirus 1, Human/immunology , Herpesvirus Vaccines/pharmacology , Vaccines, DNA/pharmacology , Animals , Antibodies, Viral/biosynthesis , B-Lymphocytes/immunology , Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/genetics , Herpesvirus Vaccines/immunology , Immunoglobulin G/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, SCID , T-Lymphocyte Subsets/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
...