Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Trop Med Infect Dis ; 2(3)2017 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-30270894

RESUMO

Lagos bat virus (LBV) is a phylogroup II lyssavirus exclusively found in Africa. Previous studies indicated that this virus is lethal to mice after intracranial and intramuscular inoculation. The antigenic composition of LBV differs substantially from that of rabies virus (RABV) and current rabies vaccines do not provide cross protection against phylogroup II lyssaviruses. To investigate the potential role of the LBV matrix protein (M) and glycoprotein (G) in pathogenesis, reverse genetics technology was used to construct recombinant viruses. The genes encoding the glycoprotein, or the matrix and glycoprotein of the attenuated RABV strain SPBN, were replaced with those of LBV resulting in SPBN-LBVG and SPBN-LBVM-LBVG, respectively. To evaluate the immunogenicity of the LBV G, the recombinant RABV SPBNGAS-LBVG-GAS was constructed with the LBV G inserted between two mutated RABV G genes (termed GAS). All the recombinant viruses were lethal to mice after intracranial (i.c.) inoculation although the pathogenicity of SPBNGAS-LBVG-GAS was lower compared to the other recombinant viruses. Following intramuscular (i.m.) inoculation, only SPBN-LBVM-LBVG was lethal to mice, indicating that both the M and G of LBV play a role in the pathogenesis. Most interestingly, serum collected from mice that were inoculated i.m. with SPBNGAS-LBVG-GAS neutralized phylogroup I and II lyssaviruses including RABV, Duvenhage virus (DUVV), LBV, and Mokola virus (MOKV), indicating that this recombinant virus has potential to be developed as a pan-lyssavirus vaccine.

2.
Trop Med Infect Dis ; 2(4)2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-30270916

RESUMO

Rabies is an acute, progressive, incurable viral encephalitis found throughout the world. Despite being one of the oldest recognized pathogens, its impact remains substantial in public health, veterinary medicine, and conservation biology.[...].

3.
PLoS One ; 9(4): e87180, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24763072

RESUMO

Central nervous system (CNS) metabolic profiles were examined from rabies virus (RABV)-infected mice that were either mock-treated or received post-exposure treatment (PET) with a single dose of the live recombinant RABV vaccine TriGAS. CNS tissue harvested from mock-treated mice at middle and late stage infection revealed numerous changes in energy metabolites, neurotransmitters and stress hormones that correlated with replication levels of viral RNA. Although the large majority of these metabolic changes were completely absent in the brains of TriGAS-treated mice most likely due to the strong reduction in virus spread, TriGAS treatment resulted in the up-regulation of the expression of carnitine and several acylcarnitines, suggesting that these compounds are neuroprotective. The most striking change seen in mock-treated RABV-infected mice was a dramatic increase in brain and serum corticosterone levels, with the later becoming elevated before clinical signs or loss of body weight occurred. We speculate that the rise in corticosterone is part of a strategy of RABV to block the induction of immune responses that would otherwise interfere with its spread. In support of this concept, we show that pharmacological intervention to inhibit corticosterone biosynthesis, in the absence of vaccine treatment, significantly reduces the pathogenicity of RABV. Our results suggest that widespread metabolic changes, including hypothalamic-pituitary-adrenal axis activation, contribute to the pathogenesis of RABV and that preventing these alterations early in infection with PET or pharmacological blockade helps protect brain homeostasis, thereby reducing disease mortality.


Assuntos
Encéfalo/metabolismo , Vírus da Raiva/imunologia , Raiva/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Imunidade Adaptativa , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Encéfalo/virologia , Carnitina/análogos & derivados , Carnitina/metabolismo , Corticosterona/sangue , Progressão da Doença , Metabolismo Energético , Feminino , Expressão Gênica , Interações Hospedeiro-Patógeno , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/virologia , Camundongos , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/virologia , Piridinas/farmacologia , Piridinas/uso terapêutico , Raiva/tratamento farmacológico , Raiva/imunologia , Carga Viral , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vacinas Virais/uso terapêutico
4.
J Virol ; 87(3): 1834-41, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23192867

RESUMO

A single intramuscular application of the live but not UV-inactivated recombinant rabies virus (RABV) variant TriGAS in mice induces the robust and sustained production of RABV-neutralizing antibodies that correlate with long-term protection against challenge with an otherwise lethal dose of the wild-type RABV. To obtain insight into the mechanism by which live TriGAS induces long-lasting protective immunity, quantitative PCR (qPCR) analysis of muscle tissue, draining lymph nodes, spleen, spinal cord, and brain at different times after TriGAS inoculation revealed the presence of significant copy numbers of RABV-specific RNA in muscle, lymph node, and to a lesser extent, spleen for several days postinfection. Notably, no significant amounts of RABV RNA were detected in brain or spinal cord at any time after TriGAS inoculation. Differential qPCR analysis revealed that the RABV-specific RNA detected in muscle is predominantly genomic RNA, whereas RABV RNA detected in draining lymph nodes is predominantly mRNA. Comparison of genomic RNA and mRNA obtained from isolated lymph node cells showed the highest mRNA-to-genomic-RNA ratios in B cells and dendritic cells (DCs), suggesting that these cells represent the major cell population that is infected in the lymph node. Since RABV RNA declined to undetectable levels by 14 days postinoculation of TriGAS, we speculate that a transient infection of DCs with TriGAS may be highly immunostimulatory through mechanisms that enhance antigen presentation. Our results support the superior efficacy and safety of TriGAS and advocate for its utility as a vaccine.


Assuntos
Linfonodos/virologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Animais , Linfócitos B/virologia , Encéfalo/patologia , Encéfalo/virologia , Células Dendríticas/virologia , Feminino , Injeções Intramusculares , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Músculos/patologia , Músculos/virologia , RNA Viral/análise , RNA Viral/genética , Raiva/virologia , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/patogenicidade , Reação em Cadeia da Polimerase em Tempo Real , Medula Espinal/patologia , Medula Espinal/virologia , Baço/patologia , Baço/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
5.
J Virol ; 86(6): 3200-10, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22238315

RESUMO

Postexposure treatment (PET) of wild-type rabies virus (RV)-infected mice with a live-attenuated triple-glycoprotein RV variant (TriGAS) promotes survival but does not prevent the pathogenic RV from invading and replicating in the brain. Successful PET is associated with the induction of a robust virus-neutralizing antibody response and clearance of the wild-type RV from brain tissues. Comparison of the transcriptomes of normal mouse brain with those of wild-type-RV-infected mice that had received either mock or TriGAS PET treatment revealed that many of the host genes activated in the mock-treated mice represent type I interferon (IFN) response genes. This indicates that RV infection induces an early type I IFN response that is unable to control the infection. In contrast, most of the activated genes in the brain of the RV-infected, TriGAS-treated mouse play a role in adaptive immunity, including the regulation of T cell activation, T cell differentiation, and the regulation of lymphocyte and mononuclear cell proliferation. These findings were confirmed by quantitative PCR (qPCR) array studies, which showed that 3 genes in particular, encoding chemokine ligand 3 (Ccl3), natural killer cell activator 2 (interleukin 12B [IL-12B]), and granzyme A (GzmA), were activated earlier and to a greater extent in the brains of RV-infected mice treated with TriGAS than in the brains of mock-treated mice. The activation of these genes, known to play key roles in the regulation of lymphocyte and mononuclear cell proliferation, is likely an important part of the mechanism by which TriGAS mediates its PET activity.


Assuntos
Imunidade Adaptativa , Sistema Nervoso Central/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/genética , Raiva/prevenção & controle , Regulação para Cima , Animais , Anticorpos Antivirais/imunologia , Sistema Nervoso Central/virologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Profilaxia Pós-Exposição , Raiva/tratamento farmacológico , Raiva/imunologia , Vacina Antirrábica/uso terapêutico , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico
6.
Adv Virus Res ; 79: 115-26, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21601045

RESUMO

The host response to infection generally begins with interactions between pathogen-associated molecular patterns common to a variety of infectious agents and reciprocal pattern-recognition receptors (PRRs) expressed by cells of the innate immune system. The innate responses triggered by these interactions contribute to the early, innate control of infection as well as the induction of pathogen-specific adaptive immunity. The outcome of infection with wild-type rabies virus is particularly dependent upon the rapid induction of innate and adaptive immune mechanisms that can prevent the virus from reaching central nervous system (CNS) tissues, where it can evade immune clearance. However, laboratory strains that reach the CNS can be cleared, and this has evidently occurred in individuals with rabies. Therefore, PRRs may be active in the periphery and the CNS during rabies virus infection, possibly depending upon the nature of the infecting virus. To investigate these possibilities, we first examined the outcome of infection with attenuated rabies virus in mice lacking MyD88, an adaptor protein that is used to activate the transcription factor NF-κB by a number of PRRs including all of the Toll-like receptors (TLRs) except for TLR3. Finding that attenuated rabies virus mediates lethal disease in the absence of MyD88, we then examined the effects of the deletion of receptors using MyD88 including TLRs 2, 4, 7, and 9 as well as IL-1-receptor 1, and IFN-αßR on infection. Only mice lacking TLR7 exhibited a phenotype, with mortality intermediate between MyD88(-/-) and control mice with deficits in both the development of peripheral immunity and rabies virus clearance from the CNS.


Assuntos
Vírus da Raiva/imunologia , Raiva/imunologia , Receptores Toll-Like/imunologia , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Vírus da Raiva/patogenicidade , Virulência
7.
PLoS Negl Trop Dis ; 3(11): e542, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19888334

RESUMO

As the demand for rabies post-exposure prophylaxis (PEP) treatments has increased exponentially in recent years, the limited supply of human and equine rabies immunoglobulin (HRIG and ERIG) has failed to provide the required passive immune component in PEP in countries where canine rabies is endemic. Replacement of HRIG and ERIG with a potentially cheaper and efficacious alternative biological for treatment of rabies in humans, therefore, remains a high priority. In this study, we set out to assess a mouse monoclonal antibody (MoMAb) cocktail with the ultimate goal to develop a product at the lowest possible cost that can be used in developing countries as a replacement for RIG in PEP. Five MoMAbs, E559.9.14, 1112-1, 62-71-3, M727-5-1, and M777-16-3, were selected from available panels based on stringent criteria, such as biological activity, neutralizing potency, binding specificity, spectrum of neutralization of lyssaviruses, and history of each hybridoma. Four of these MoMAbs recognize epitopes in antigenic site II and one recognizes an epitope in antigenic site III on the rabies virus (RABV) glycoprotein, as determined by nucleotide sequence analysis of the glycoprotein gene of unique MoMAb neutralization-escape mutants. The MoMAbs were produced under Good Laboratory Practice (GLP) conditions. Unique combinations (cocktails) were prepared, using different concentrations of the MoMAbs that were capable of targeting non-overlapping epitopes of antigenic sites II and III. Blind in vitro efficacy studies showed the MoMab cocktails neutralized a broad spectrum of lyssaviruses except for lyssaviruses belonging to phylogroups II and III. In vivo, MoMAb cocktails resulted in protection as a component of PEP that was comparable to HRIG. In conclusion, all three novel combinations of MoMAbs were shown to have equal efficacy to HRIG and therefore could be considered a potentially less expensive alternative biological agent for use in PEP and prevention of rabies in humans.


Assuntos
Anticorpos Monoclonais/farmacologia , Profilaxia Pós-Exposição/métodos , Raiva/prevenção & controle , Animais , Anticorpos Monoclonais/economia , Anticorpos Monoclonais/imunologia , Linhagem Celular , Cricetinae , Humanos , Camundongos , Testes de Neutralização , Profilaxia Pós-Exposição/economia , Raiva/tratamento farmacológico , Raiva/imunologia , Vírus da Raiva/efeitos dos fármacos , Vírus da Raiva/enzimologia , Vírus da Raiva/imunologia
8.
Proc Natl Acad Sci U S A ; 106(27): 11300-5, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19581599

RESUMO

Rabies remains an important public health problem with more than 95% of all human rabies cases caused by exposure to rabid dogs in areas where effective, inexpensive vaccines are unavailable. Because of their ability to induce strong innate and adaptive immune responses capable of clearing the infection from the CNS after a single immunization, live-attenuated rabies virus (RV) vaccines could be particularly useful not only for the global eradication of canine rabies but also for late-stage rabies postexposure prophylaxis of humans. To overcome concerns regarding the safety of live-attenuated RV vaccines, we developed the highly attenuated triple RV G variant, SPBAANGAS-GAS-GAS. In contrast to most attenuated recombinant RVs generated thus far, SPBAANGAS-GAS-GAS is completely nonpathogenic after intracranial infection of mice that are either developmentally immunocompromised (e.g., 5-day-old mice) or have inherited deficits in immune function (e.g., antibody production or type I IFN signaling), as well as normal adult animals. In addition, SPBAANGAS-GAS-GAS induces immune mechanisms capable of containing a CNS infection with pathogenic RV, thereby preventing lethal rabies encephalopathy. The lack of pathogenicity together with excellent immunogenicity and the capacity to deliver immune effectors to CNS tissues makes SPBAANGAS-GAS-GAS a promising vaccine candidate for both the preexposure and postexposure prophylaxis of rabies.


Assuntos
Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Raiva/prevenção & controle , Envelhecimento/imunologia , Animais , Animais Lactentes , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/virologia , Hospedeiro Imunocomprometido , Camundongos , Permeabilidade , Vírus da Raiva/patogenicidade , Análise de Sobrevida , Resultado do Tratamento , Vacinação , Vacinas Atenuadas
9.
PLoS Pathog ; 5(6): e1000485, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19543379

RESUMO

The majority of rabies virus (RV) infections are caused by bites or scratches from rabid carnivores or bats. Usually, RV utilizes the retrograde transport within the neuronal network to spread from the infection site to the central nervous system (CNS) where it replicates in neuronal somata and infects other neurons via trans-synaptic spread. We speculate that in addition to the neuronal transport of the virus, hematogenous spread from the site of infection directly to the brain after accidental spill over into the vascular system might represent an alternative way for RV to invade the CNS. So far, it is unknown whether hematogenous spread has any relevance in RV pathogenesis. To determine whether certain RV variants might have the capacity to invade the CNS from the periphery via hematogenous spread, we infected mice either intramuscularly (i.m.) or intravenously (i.v.) with the dog-associated RV DOG4 or the silver-haired bat-associated RV SB. In addition to monitoring the progression of clinical signs of rabies we used immunohistochemistry and quantitative reverse transcription polymerase chain reaction (qRT-PCR) to follow the spread of the virus from the infection site to the brain. In contrast to i.m. infection where both variants caused a lethal encephalopathy, only i.v. infection with SB resulted in the development of a lethal infection. While qRT-PCR did not reveal major differences in virus loads in spinal cord or brain at different times after i.m. or i.v. infection of SB, immunohistochemical analysis showed that only i.v. administered SB directly infected the forebrain. The earliest affected regions were those hypothalamic nuclei, which are connected by neurosecretory fibers to the circumventricular organs neurohypophysis and median eminence. Our data suggest that hematogenous spread of SB can lead to a fatal encephalopathy through direct retrograde invasion of the CNS at the neurovascular interface of the hypothalamus-hypophysis system. This alternative mode of virus spread has implications for the post exposure prophylaxis of rabies, particularly with silver-haired bat-associated RV.


Assuntos
Encefalopatias/virologia , Quirópteros/virologia , Vírus da Raiva/fisiologia , Raiva/transmissão , Análise de Variância , Animais , Antígenos Virais/análise , Encéfalo/virologia , Cães , Imuno-Histoquímica , Injeções Intramusculares , Injeções Intravenosas , Eminência Mediana/virologia , Camundongos , Fibras Nervosas/virologia , Neuro-Hipófise/virologia , RNA Viral/análise , RNA Viral/sangue , Raiva/virologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Medula Espinal/virologia , Distribuição Tecidual , Carga Viral
10.
Vaccine ; 26(50): 6405-14, 2008 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-18804506

RESUMO

The type of immune response induced by a vaccine is a critical factor that determines its effectiveness in preventing infection or disease. Inactivated and live rabies virus (RV) vaccine strains elicit an IgG1-biased and IgG1/IgG2a-balanced antibody response, respectively. However, IgG2a antibodies are potent inducers of anti-viral effector functions, and therefore, a viral vaccine vector that can elicit an IgG2a-biased antibody response may be more effective against RV infection. Here we describe the humoral immune response of a live replication-deficient phosphoprotein (P)-deleted RV vector (SPBN-DeltaP), or a recombinant P-deleted virus that expresses two copies of the RV glycoprotein (G) gene (SPBN-DeltaP-RVG), and compare it to a UV-inactivated RV. Mice inoculated with UV-inactivated RV induced predominantly an IgG1-specific antibody response, while live recombinant SPBN-DeltaP exhibited a mixed IgG1/IgG2a antibody response, which is consistent with the isotype profiles from the replication-competent parental viruses. Survivorship in mice after pathogenic RV challenge indicates a 10-fold higher efficiency of live SPBN-DeltaP compared to UV-inactivated SPBN-DeltaP. In addition, SPBN-DeltaP-RVG induced a more rapid and robust IgG2a response that protected mice more effectively than SPBN-DeltaP. Of note, 10(3)ffu of SPBN-DeltaP-RVG-induced anti-RV antibodies that were 100% protective in mice against pathogenic RV challenge. The increased immune response was directed not only against RV G but also against the ribonucleoprotein (RNP), indicating that the expression of two RV G genes from SPBN-DeltaP-RVG enhances the immune response to other RV antigens as well. In addition, Rag2 mice inoculated intramuscularly with 10(5)ffu/mouse of SPBN-DeltaP showed no clinical signs of rabies, and no viral RNA was detected in the spinal cord or brain of inoculated mice. Therefore, the safety of the P-deleted vectors along with the onset and magnitude of the IgG2a-induced immune response by SPBN-DeltaP-RVG indicate that this vector holds great promise as either a therapeutic or preventative vaccine against RV or other infectious diseases.


Assuntos
Anticorpos Antivirais/sangue , Dosagem de Genes , Vetores Genéticos , Vacina Antirrábica/imunologia , Raiva/prevenção & controle , Proteínas do Envelope Viral/imunologia , Viroses/prevenção & controle , Animais , Cricetinae , Feminino , Deleção de Genes , Camundongos , Camundongos Endogâmicos BALB C , Fosfoproteínas/genética , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/genética , Vírus da Raiva/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Viroses/imunologia , Viroses/virologia
11.
Vaccine ; 26(3): 419-26, 2008 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-18082293

RESUMO

To assess the potential role of dendritic cells (DCs) or monocytes in the development of a protective immune response, we infected human immature DCs or monocytes with a live rabies virus (RV) vaccine strain (SPBNGAS-GAS) and a pathogenic RV (DOG4). Both cell types were infected with SPBNGAS-GAS and DOG4 and both RVs were similarly potent in inducing maturation of immature DCs or monocytes. However, in contrast to DOG4, SPBNGAS-GAS induced very high levels of IFN-alpha1 mRNA in monocytes and DCs. Furthermore, at least 26 other genes related to the NFkappaB signaling pathway were strongly upregulated in SPBNGAS-GAS-infected DCs, but only somewhat increased in DOG4-infected cells. Thus, the extent of upregulation of NFkappaB pathway-related genes in DCs infected with the live RV vaccine strain might explain the strong protective activity of SPBNGAS-GAS.


Assuntos
Diferenciação Celular , Células Dendríticas/citologia , Células Dendríticas/virologia , Monócitos/virologia , NF-kappa B/metabolismo , Vírus da Raiva/patogenicidade , Transdução de Sinais , Regulação para Cima , Humanos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Reação em Cadeia da Polimerase , Proteínas/genética , Proteínas/metabolismo
12.
J Virol ; 82(5): 2330-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18094173

RESUMO

While the glycoprotein (G) of rabies virus (RV) is known to play a predominant role in the pathogenesis of rabies, the function of the RV matrix protein (M) in RV pathogenicity is not completely clear. To further investigate the roles of these proteins in viral pathogenicity, we constructed chimeric recombinant viruses by exchanging the G and M genes of the attenuated SN strain with those of the highly pathogenic SB strain. Infection of mice with these chimeric viruses revealed a significant increase in the pathogenicity of the SN strain bearing the RV G from the pathogenic SB strain. Moreover, the pathogenicity was further increased when both G and M from SB were introduced into SN. Interestingly, the replacement of the G or M gene or both in SN by the corresponding genes of SB was associated with a significant decrease in the rate of viral replication and viral RNA synthesis. In addition, a chimeric SN virus bearing both the M and G genes from SB exhibited more efficient cell-to-cell spread than a chimeric SN virus in which only the G gene was replaced. Together, these data indicate that both G and M play an important role in RV pathogenesis by regulating virus replication and facilitating cell-to-cell spread.


Assuntos
Movimento Celular/fisiologia , Glicoproteínas/fisiologia , Vírus da Raiva/patogenicidade , Proteínas da Matriz Viral/fisiologia , Replicação Viral/fisiologia , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Cricetinae , Primers do DNA , Feminino , Camundongos , Vírus da Raiva/genética , Vírus da Raiva/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Virulência
13.
Future Virol ; 3(5): 481-490, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19578477

RESUMO

Rabies is a zoonotic disease that remains an important public health problem worldwide and causes more than 70,000 human deaths each year. The causative agent of rabies is rabies virus (RV), a negative-stranded RNA virus of the rhabdovirus family. Neuroinvasiveness and neurotropism are the main features that define the pathogenesis of rabies. Although RV pathogenicity is a multigenic trait involving several elements of the RV genome, the RV glycoprotein plays a major role in RV pathogenesis by controlling the rate of virus uptake and trans-synaptic virus spread, and by regulating the rate of virus replication. Pathogenic street RV strains differ significantly from tissue culture-adapted RV strains in their neuroinvasiveness. Whereas street RV strains are highly neuroinvasive, most tissue culture-adapted RV strains have either no or only limited ability to invade the CNS from a peripheral site. The high neuroinvasiveness of pathogenic street RVs is, at least in part, due to their ability to evade immune responses and to conserve the structures of neurons. The finding that tissue culture-adapted RV strains replicate very fast and induce strong innate and adaptive immune responses opens new avenues for therapeutic intervention against rabies.

14.
Vaccine ; 25(42): 7296-300, 2007 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-17826874

RESUMO

Oral vaccination is an important tool currently in use to control the spread of rabies in wildlife populations in various programs around the world. Oral rabies vaccination (ORV) of raccoons represents the largest targeted program to control wildlife rabies in the United States. Currently, the vaccinia-rabies glycoprotein recombinant virus vaccine (V-RG) is the only licensed oral rabies vaccine in the US. In the current study, captive raccoons were used to evaluate two previously described constructs of a rabies virus vaccine developed by reverse genetics (SPBNGAS and SPBNGAS-GAS) for immunogenicity and efficacy compared to the V-RG vaccine. Four of five control animals succumbed to rabies virus after severe challenge, while three of five animals vaccinated orally with SPBNGAS succumbed. No mortality was observed for animals administered SPBNGAS-GAS or the V-RG vaccine. The results of this preliminary study suggest that SPBNGAS-GAS provides comparable efficacy to V-RG. Additional studies will be needed to determine the duration of immunity and optimal dosage of SPBNGAS-GAS and to examine its efficacy in other reservoir species.


Assuntos
Vacina Antirrábica/administração & dosagem , Guaxinins/imunologia , Guaxinins/virologia , Administração Oral , Animais , Anticorpos Antivirais/sangue , Raiva/imunologia , Raiva/prevenção & controle , Raiva/veterinária , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Segurança , Estados Unidos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
15.
J Virol ; 81(13): 7041-7, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17459937

RESUMO

The nonpathogenic phenotype of the live rabies virus (RV) vaccine SPBNGAN is determined by an Arg-->Glu exchange at position 333 in the glycoprotein, designated GAN. We recently showed that after several passages of SPBNGAN in mice, an Asn-->Lys mutation arose at position 194 of GAN, resulting in GAK, which was associated with a reversion to the pathogenic phenotype. Because an RV vaccine candidate containing two GAN genes (SPBNGAN-GAN) exhibits increased immunogenicity in vivo compared to the single-GAN construct, we tested whether the presence of two GAN genes might also enhance the probability of reversion to pathogenicity. Comparison of SPBNGAN-GAN with RVs constructed to contain either both GAN and GAK genes (SPBNGAN-GAK and SPBNGAK-GAN) or two GAK genes (SPBNGAK-GAK) showed that while SPBNGAK-GAK was pathogenic, SPBNGAN-GAN and SPBNGAN-GAK were completely nonpathogenic and SPBNGAK-GAN showed strongly reduced pathogenicity. Analysis of genomic RV RNA in mouse brain tissue revealed significantly lower virus loads in SPBNGAN-GAK- and SPBNGAK-GAN-infected brains than those detected in SPBNGAK-GAK-infected brains, indicating the dominance of the nonpathogenic phenotype determined by GAN over the GAK-associated pathogenic phenotype. Virus production and viral RNA synthesis were markedly higher in SPBNGAN-, SPBNGAK-GAN-, and SPBNGAN-GAK-infected neuroblastoma cells than in the SPBNGAK- and SPBNGAK-GAK-infected counterparts, suggesting control of GAN dominance at the level of viral RNA synthesis. These data point to the lower risk of reversion to pathogenicity of a recombinant RV carrying two identical GAN genes compared to that of an RV carrying only a single GAN gene.


Assuntos
Genes Dominantes , Genes Virais , Glicoproteínas/metabolismo , Vacina Antirrábica/metabolismo , Vírus da Raiva/metabolismo , Raiva/metabolismo , Proteínas Virais/metabolismo , Substituição de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Linhagem Celular , Glicoproteínas/genética , Masculino , Camundongos , Mutação de Sentido Incorreto , RNA Viral/biossíntese , RNA Viral/genética , Raiva/genética , Vacina Antirrábica/genética , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Carga Viral , Proteínas Virais/genética
16.
J Infect Dis ; 195(7): 980-8, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17330788

RESUMO

We analyzed the safety and immunogenicity of attenuated rabies virus vectors expressing simian-human immunodeficiency virus (SHIV)-1(89.6P) Env or simian immunodeficiency virus (SIV)(mac239) Gag in rhesus macaques. Four test macaques were immunized with both vaccine constructs, and 2 control macaques received an empty rabies vector. Seroconversion against rabies virus glycoprotein (G) and SHIV(89.6P) Env was detected after the initial immunization, but no cellular responses against SHIV antigens were observed. HIV/SIV-specific immune responses were not enhanced by boosts with the same vectors. Therefore, we constructed vectors expressing SHIV(89.6P) Env and SIV(mac239) Gag in which the rabies G was replaced with the G protein of vesicular stomatitis virus (VSV). Two years after initial immunization, a boost with the rabies-VSV G vectors resulted in SIV/HIV-specific immune responses. Upon challenge with SHIV(89.6P) test macaques controlled the infection, whereas control macaques had high levels of viremia and a profound loss of CD4(+) T cells, with 1 control macaque dying of an AIDS-like disease.


Assuntos
Produtos do Gene env/imunologia , Produtos do Gene gag/imunologia , Vacinas contra a SAIDS/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vacinas Sintéticas/uso terapêutico , Animais , Primers do DNA , Produtos do Gene env/genética , Produtos do Gene gag/genética , Vetores Genéticos , Macaca mulatta , Masculino , RNA Viral/análise , Vírus da Raiva/genética , Vírus da Imunodeficiência Símia/genética
17.
J Wildl Dis ; 42(3): 663-6, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17092899

RESUMO

Oral vaccination of free-ranging wildlife is a promising technique in rabies control. The small Asian mongoose (Herpestes javanicus) is an important reservoir of rabies on several Caribbean islands, but no vaccines have been evaluated for this species. Captive mongooses were used to test the safety and efficacy of the commercially licensed vaccinia-rabies glycoprotein (V-RG) recombinant vaccine and a newly developed genetically engineered oral rabies virus vaccine (SPBNGA-S). In one study using V-RG, no vaccinated animals developed detectable rabies virus-neutralizing antibodies, and all but one died after experimental challenge with rabies virus. In contrast, all animals given SPBNGA-S demonstrated seroconversion within 7 to 14 days after vaccination and survived rabies virus challenge. On the basis of these preliminary results indicating the greater efficacy of SPBNGA-S vs. V-RG vaccine, additional investigations will be necessary to determine the optimal dose and duration of vaccination, as well as incorporation of the SPBNGA-S vaccine into edible bait.


Assuntos
Herpestidae , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/imunologia , Raiva/veterinária , Administração Oral , Animais , Animais Selvagens , Anticorpos Antivirais/sangue , Reservatórios de Doenças/veterinária , Relação Dose-Resposta Imunológica , Feminino , Masculino , Raiva/prevenção & controle , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
18.
Virology ; 356(1-2): 147-54, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16938327

RESUMO

Rabies vaccines based on live attenuated rabies viruses or recombinant pox viruses expressing the rabies virus (RV) glycoprotein (G) hold the greatest promise of safety and efficacy, particularly for oral immunization of wildlife. However, while these vaccines induce protective immunity in foxes, they are less effective in other animals, and safety concerns have been raised for some of these vaccines. Because canine adenovirus 2 (CAV2) is licensed for use as a live vaccine for dogs and has an excellent efficacy and safety record, we used this virus as an expression vector for the RVG. The recombinant CAV2-RV G produces virus titers similar to those produced by wild-type CAV2, indicating that the RVG gene does not affect virus replication. Comparison of RVG expressed by CAV2-RV G with that of vaccinia-RV G recombinant virus (V-RG) revealed similar amounts of RV G on the cell surface. A single intramuscular or intranasal immunization of mice with CAV2-RVG induced protective immunity in a dose-dependent manner, with no clinical signs or discomfort from the virus infection regardless of the route of administration or the amount of virus.


Assuntos
Adenovirus Caninos/genética , Antígenos Virais/imunologia , Glicoproteínas/imunologia , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Recombinação Genética , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Linhagem Celular , Cães , Feminino , Glicoproteínas/genética , Imunização , Camundongos , Testes de Neutralização , Raiva/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/patogenicidade , Proteínas do Envelope Viral/genética
19.
Virology ; 353(2): 344-56, 2006 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16820183

RESUMO

Live viral vectors expressing foreign antigens have shown great promise as vaccines against viral diseases. However, safety concerns remain a major problem regarding the use of even highly attenuated viral vectors. Using the rabies virus (RV) envelope protein as a carrier molecule, we show here that inactivated RV particles can be utilized to present Bacillus anthracis protective antigen (PA) domain-4 in the viral membrane. In addition to the RV glycoprotein (G) transmembrane and cytoplasmic domains, a portion of the RV G ectodomain was required to express the chimeric RV G anthrax PA on the cell surface. The novel antigen was also efficiently incorporated into RV virions. Mice immunized with the inactivated recombinant RV virions exhibited seroconversion against both RV G and anthrax PA, and a second inoculation greatly increased these responses. These data demonstrate that a viral envelope protein can carry a bacterial protein and that a viral carrier can display whole polypeptides compared to the limited epitope presentation of previous viral systems.


Assuntos
Vacinas contra Antraz/administração & dosagem , Antraz/prevenção & controle , Antígenos de Bactérias/genética , Antígenos Virais/genética , Bacillus anthracis/imunologia , Toxinas Bacterianas/genética , Vetores Genéticos/genética , Glicoproteínas/genética , Vacina Antirrábica/genética , Vacinação , Proteínas do Envelope Viral/genética , Animais , Antraz/sangue , Antraz/imunologia , Anticorpos Antibacterianos/sangue , Anticorpos Antivirais/sangue , Especificidade de Anticorpos , Antígenos de Bactérias/imunologia , Antígenos Virais/imunologia , Toxinas Bacterianas/imunologia , Bioterrorismo/prevenção & controle , Vetores Genéticos/imunologia , Glicoproteínas/imunologia , Esquemas de Imunização , Injeções Intramusculares , Contagem de Linfócitos , Linfócitos/citologia , Linfócitos/imunologia , Camundongos , Vacina Antirrábica/imunologia , Recombinação Genética , Células Th2/imunologia , Vacinas Sintéticas/administração & dosagem , Proteínas do Envelope Viral/imunologia
20.
J Infect Dis ; 193(6): 796-801, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16479514

RESUMO

The World Health Organization estimates human mortality from endemic canine rabies to be 55,000 deaths/year. Limited supply hampers the accessibility of appropriate lifesaving treatment, particularly in areas where rabies is endemic. Anti-rabies antibodies are key to protection against lethal rabies. Currently, only human and equine polyclonal anti-rabies immune globulin (HRIG and ERIG) is available. Replacement of HRIG and ERIG with a safer and more widely available product is recommended. We have recently identified a combination of 2 human monoclonal antibodies (MAbs), CR57 and CR4098, that has high potential. We here describe a head-to-head comparison between an CR57/CR4098 MAb cocktail and HRIG. The MAb cocktail neutralized all viruses from a panel of 26 representative street rabies virus isolates. In combination with vaccine, the MAb cocktail protected Syrian hamsters against lethal rabies when administered 24 h after exposure, comparable with the results obtained with HRIG. Furthermore, the MAb cocktail did not interfere with rabies vaccine differently from HRIG. These results demonstrate that the human MAb cocktail of CR57 and CR4098 is a safe and efficacious alternative to RIG in rabies postexposure prophylaxis.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Imunoglobulinas/administração & dosagem , Vírus da Raiva/imunologia , Raiva/terapia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Linhagem Celular , Cricetinae , Humanos , Imunoglobulinas/efeitos adversos , Imunoterapia , Mesocricetus , Raiva/imunologia , Raiva/prevenção & controle , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...