Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30567978

RESUMO

Marburg virus (MARV) is a filovirus related to Ebola virus (EBOV) associated with human hemorrhagic disease. Outbreaks are sporadic and severe, with a reported case mortality rate of upward of 88%. There is currently no antiviral or vaccine available. Given the sporadic nature of outbreaks, vaccines provide the best approach for long-term control of MARV in regions of endemicity. We have developed an inactivated rabies virus-vectored MARV vaccine (FILORAB3) to protect against Marburg virus disease. Immunogenicity studies in our labs have shown that a Th1-biased seroconversion to both rabies virus and MARV glycoproteins (GPs) is beneficial for protection in a preclinical murine model. As such, we adjuvanted FILORAB3 with glucopyranosyl lipid adjuvant (GLA), a Toll-like receptor 4 agonist, in a squalene-in-water emulsion. Across two different BALB/c mouse challenge models, we achieved 92% protection against murine-adapted Marburg virus (ma-MARV). Although our vaccine elicited strong MARV GP antibodies, it did not strongly induce neutralizing antibodies. Through both in vitro and in vivo approaches, we elucidated a critical role for NK cell-dependent antibody-mediated cellular cytotoxicity (ADCC) in vaccine-induced protection. Overall, these findings demonstrate that FILORAB3 is a promising vaccine candidate for Marburg virus disease.IMPORTANCE Marburg virus (MARV) is a virus similar to Ebola virus and also causes a hemorrhagic disease which is highly lethal. In contrast to EBOV, only a few vaccines have been developed against MARV, and researchers do not understand what kind of immune responses are required to protect from MARV. Here we show that antibodies directed against MARV after application of our vaccine protect in an animal system but fail to neutralize the virus in a widely used virus neutralization assay against MARV. This newly discovered activity needs to be considered more when analyzing MARV vaccines or infections.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Glicoproteínas/imunologia , Doença do Vírus de Marburg/imunologia , Marburgvirus/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacina Antirrábica/imunologia , Vacinação/métodos , Células Vero , Vacinas Virais/imunologia
2.
J Infect Dis ; 214(suppl 3): S342-S354, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27456709

RESUMO

The 2013-2016 West African Ebola virus (EBOV) disease outbreak was the largest filovirus outbreak to date. Over 28 000 suspected, probable, or confirmed cases have been reported, with a 53% case-fatality rate. The magnitude and international impact of this EBOV outbreak has highlighted the urgent need for a safe and efficient EBOV vaccine. To this end, we demonstrate the immunogenicity and protective efficacy of FILORAB1, a recombinant, bivalent, inactivated rabies virus-based EBOV vaccine, in rhesus and cynomolgus monkeys. Our results demonstrate that the use of the synthetic Toll-like receptor 4 agonist glucopyranosyl lipid A in stable emulsion (GLA-SE) as an adjuvant increased the efficacy of FILORAB1 to 100% protection against lethal EBOV challenge, with no to mild clinical signs of disease. Furthermore, all vaccinated subjects developed protective anti-rabies virus antibody titers. Taken together, these results support further development of FILORAB1/GLA-SE as an effective preexposure EBOV vaccine.


Assuntos
Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Glucosídeos/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Lipídeo A/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Adjuvantes Imunológicos , Animais , Anticorpos Antivirais/imunologia , Emulsões , Feminino , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Macaca fascicularis , Macaca mulatta , Masculino , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/imunologia , Receptor 4 Toll-Like/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/imunologia
3.
J Gen Virol ; 97(8): 1942-1954, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27166137

RESUMO

We previously demonstrated that small-particle (0.5-3.0 µm) aerosol infection of rhesus monkeys (Macaca mulatta) with cowpox virus (CPXV)-Brighton Red (BR) results in fulminant respiratory tract disease characterized by severe lung parenchymal pathology but only limited systemic virus dissemination and limited classic epidermal pox-like lesion development (Johnson et al., 2015). Based on these results, and to further develop CPXV as an improved model of human smallpox, we evaluated a novel large-particle aerosol (7.0-9.0 µm) exposure of rhesus monkeys to CPXV-BR and monitored for respiratory tract disease by serial computed tomography (CT). As expected, the upper respiratory tract and large airways were the major sites of virus-induced pathology following large-particle aerosol exposure. Large-particle aerosol CPXV exposure of rhesus macaques resulted in severe upper airway and large airway pathology with limited systemic dissemination.


Assuntos
Aerossóis , Vírus da Varíola Bovina/patogenicidade , Varíola Bovina/patologia , Varíola Bovina/virologia , Modelos Animais de Doenças , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Animais , Macaca mulatta , Infecções Respiratórias/diagnóstico por imagem , Tomografia Computadorizada por Raios X
4.
J Infect Dis ; 212 Suppl 2: S414-24, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26063224

RESUMO

We previously described the generation of a novel Ebola virus (EBOV) vaccine based on inactivated rabies virus (RABV) containing EBOV glycoprotein (GP) incorporated in the RABV virion. Our results demonstrated safety, immunogenicity, and protective efficacy in mice and nonhuman primates (NHPs). Protection against viral challenge depended largely on the quality of the humoral immune response against EBOV GP.Here we present the extension and improvement of this vaccine by increasing the amount of GP incorporation into virions via GP codon-optimization as well as the addition of Sudan virus (SUDV) and Marburg virus (MARV) GP containing virions. Immunogenicity studies in mice indicate similar immune responses for both SUDV GP and MARV GP compared to EBOV GP. Immunizing mice with multiple antigens resulted in immune responses similar to immunization with a single antigen. Moreover, immunization of NHP with the new inactivated RABV EBOV vaccine resulted in high titer neutralizing antibody levels and 100% protection against lethal EBOV challenge when applied with adjuvant.Our results indicate that an inactivated polyvalent vaccine against RABV filoviruses is achievable. Finally, the novel vaccines are produced on approved VERO cells and a clinical grade RABV/EBOV vaccine for human trials has been produced.


Assuntos
Filoviridae/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Vacinas de Produtos Inativados/imunologia , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos/métodos , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Glicoproteínas/imunologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Macaca fascicularis , Marburgvirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Raiva/virologia , Sudão , Vacinação/métodos , Células Vero
5.
PLoS Pathog ; 9(5): e1003389, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23737747

RESUMO

We have previously described the generation of a novel Ebola virus (EBOV) vaccine platform based on (a) replication-competent rabies virus (RABV), (b) replication-deficient RABV, or (c) chemically inactivated RABV expressing EBOV glycoprotein (GP). Mouse studies demonstrated safety, immunogenicity, and protective efficacy of these live or inactivated RABV/EBOV vaccines. Here, we evaluated these vaccines in nonhuman primates. Our results indicate that all three vaccines do induce potent immune responses against both RABV and EBOV, while the protection of immunized animals against EBOV was largely dependent on the quality of humoral immune response against EBOV GP. We also determined if the induced antibodies against EBOV GP differ in their target, affinity, or the isotype. Our results show that IgG1-biased humoral responses as well as high levels of GP-specific antibodies were beneficial for the control of EBOV infection after immunization. These results further support the concept that a successful EBOV vaccine needs to induce strong antibodies against EBOV. We also showed that a dual vaccine against RABV and filoviruses is achievable; therefore addressing concerns for the marketability of this urgently needed vaccine.


Assuntos
Anticorpos Antivirais/imunologia , Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Imunoglobulina G/imunologia , Vacina Antirrábica , Vírus da Raiva , Proteínas da Matriz Viral , Animais , Vacinas contra Ebola/genética , Vacinas contra Ebola/imunologia , Vacinas contra Ebola/farmacologia , Ebolavirus/genética , Ebolavirus/imunologia , Feminino , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Macaca mulatta , Masculino , Camundongos , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vacina Antirrábica/farmacologia , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/farmacologia
6.
Neuropharmacology ; 226: 109397, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36623805

RESUMO

We recently reported that the "Dopamine Neuron Challenge Test" (DNC Test), a diagnostic method that measures the levels of dopamine metabolites in cerebrospinal fluid (CSF) and plasma samples after pharmacologically inducing a transient dopamine release, can detect early-stage Parkinson's disease (PD) with high sensitivity and selectivity in mouse models. The use of haloperidol in the original DNC test to challenge dopamine neurons was less than ideal, as it may cause extrapyramidal motor symptoms. Here we report an improved DNC Test, in which the original challenging agents, haloperidol and methylphenidate, are replaced by a single challenging agent, a dopamine autoreceptor preferring antagonist AJ76 or UH232. We show that the improved DNC Test can achieve the same level of sensitivity and selectivity in detecting early PD in a mouse model without causing motor side effects. These findings significantly improve the practicality of using the DNC Test as a screening or diagnostic test for detecting early-stage PD in the high-risk population in humans.


Assuntos
Dopamina , Doença de Parkinson , Animais , Camundongos , Humanos , Dopamina/metabolismo , Doença de Parkinson/diagnóstico , Doença de Parkinson/tratamento farmacológico , Haloperidol/farmacologia , Haloperidol/uso terapêutico , Ácido Homovanílico/metabolismo , Antagonistas de Dopamina/farmacologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo
7.
J Virol ; 85(20): 10605-16, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21849459

RESUMO

The search for a safe and efficacious vaccine for Ebola virus continues, as no current vaccine candidate is nearing licensure. We have developed (i) replication-competent, (ii) replication-deficient, and (iii) chemically inactivated rabies virus (RABV) vaccines expressing Zaire Ebola virus (ZEBOV) glycoprotein (GP) by a reverse genetics system based on the SAD B19 RABV wildlife vaccine. ZEBOV GP is efficiently expressed by these vaccine candidates and is incorporated into virions. The vaccine candidates were avirulent after inoculation of adult mice, and viruses with a deletion in the RABV glycoprotein had greatly reduced neurovirulence after intracerebral inoculation in suckling mice. Immunization with live or inactivated RABV vaccines expressing ZEBOV GP induced humoral immunity against each virus and conferred protection from both lethal RABV and EBOV challenge in mice. The bivalent RABV/ZEBOV vaccines described here have several distinct advantages that may speed the development of inactivated vaccines for use in humans and potentially live or inactivated vaccines for use in nonhuman primates at risk of EBOV infection in endemic areas.


Assuntos
Vacinas contra Ebola/imunologia , Vacina Antirrábica/imunologia , Animais , Anticorpos Antivirais/sangue , Encéfalo/virologia , Modelos Animais de Doenças , Vacinas contra Ebola/administração & dosagem , Vacinas contra Ebola/efeitos adversos , Vacinas contra Ebola/genética , Ebolavirus/genética , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Raiva/prevenção & controle , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/efeitos adversos , Vacina Antirrábica/genética , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Doenças dos Roedores/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/efeitos adversos , Vacinas de Produtos Inativados/genética , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Virulência
8.
Curr Protoc ; 2(11): e587, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36373979

RESUMO

This article describes how to assemble and operate a spectrometer-based fiber photometry system for in vivo simultaneous measurements of multiple fluorescent biosensors in freely moving mice. The first section of the article describes the step-by-step procedure to assemble a basic single-spectrometer fiber photometry system and how to expand it into a dual-spectrometer system that allows for simultaneous recordings from two sites. The second part describes the steps for a typical fiber probe implantation surgery. The last section describes how to acquire and analyze the time-lapsed spectral data. This article is intended for teaching labs how to build their own fiber photometry systems (with a video tutorial) from commercially available parts and perform in vivo recordings in behaving mice. © Published 2022. This article is a U.S. Government work and is in the public domain in the USA. Basic Protocol 1: Assembling a dual-laser, single-spectrometer fiber photometry system Support Protocol: Dual-spectrometer fiber photometry assembly Basic Protocol 2: Optical fiber probe implantation Basic Protocol 3: Data acquisition and analysis.


Assuntos
Fibras Ópticas , Fotometria , Animais , Camundongos , Fotometria/métodos
9.
NPJ Parkinsons Dis ; 7(1): 116, 2021 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-34916526

RESUMO

Diagnosing Parkinson's disease (PD) before the clinical onset proves difficult because the hallmark PD symptoms do not manifest until more than 60% of dopamine neurons in the substantia nigra pars compacta have been lost. Here we show that, by evoking a transient dopamine release and subsequently measuring the levels of dopamine metabolites in the cerebrospinal fluid and plasma, a hypodopaminergic state can be revealed when less than 30% of dopamine neurons are lost in mouse PD models. These findings may lead to sensitive and practical screening and diagnostic tests for detecting early PD in the high-risk population.

10.
J Infect Dis ; 200(8): 1251-60, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19764884

RESUMO

Although current postexposure prophylaxis rabies virus (RV) vaccines are effective, approximately 40,000-70,000 rabies-related deaths are reported annually worldwide. The development of effective formulations requiring only 1-2 applications would significantly reduce mortality. We assessed in mice and nonhuman primates the efficacy of replication-deficient RV vaccine vectors that lack either the matrix (M) or phosphoprotein (P) gene. A single dose of M gene-deficient RV induced a more rapid and efficient anti-RV response than did P gene-deficient RV immunization. Furthermore, the M gene-deleted RV vaccine induced 4-fold higher virus-neutralizing antibody (VNA) levels in rhesus macaques than did a commercial vaccine within 10 days after inoculation, and at 180 days after immunization rhesus macaques remained healthy and had higher-avidity antibodies, higher VNA titers, and a more potent antibody response typical of a type 1 T helper response than did animals immunized with a commercial vaccine. The data presented in this article suggest that the M gene-deleted RV vaccine is safe and effective and holds the potential of replacing current pre- and postexposure RV vaccines.


Assuntos
Anticorpos Antivirais/sangue , Vacina Antirrábica/imunologia , Vírus da Raiva/fisiologia , Raiva/prevenção & controle , Vacinas Atenuadas/imunologia , Animais , Afinidade de Anticorpos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Deleção de Genes , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Raiva/imunologia , Vacina Antirrábica/efeitos adversos , Vacinas Atenuadas/efeitos adversos , Replicação Viral
11.
J Virol Methods ; 281: 113863, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32371233

RESUMO

Recombinant adeno-associated viruses (rAAVs) are robust and versatile tools for in vivo gene delivery. Natural and designer capsid variations in rAAVs allow for targeted gene delivery to specific cell types. Low immunogenicity and lack of pathogenesis also add to the popularity of this virus as an innocuous gene delivery vector for gene therapy. rAAVs are routinely used to express recombinases, sensors, detectors, CRISPR-Cas9 components, or to simply overexpress a gene of interest for functional studies. High production demand has given rise to multiple platforms for the production and purification of rAAVs. However, most platforms rely heavily on large amounts of starting material and multiple purification steps to produce highly purified viral particles. Often, researchers require several small-scale purified rAAVs. Here, we describe a simple and efficient technique for purification of recombinant rAAVs from small amounts of starting material in a two-step purification method. In this method, rAAVs are released into the packaging cell medium using high salt concentration, pelleted by ultracentrifugation to remove soluble impurities. Then, the resuspended pellet is purified using a protein spin-concentrator. In this protocol, we modify the conventional rAAV purification methods to eliminate the need for fraction collection and the labor-intensive steps for evaluating the titer and purity of individual fractions. The resulting rAAV preparations are comparable in titer and purity to commercially available samples. This simplified process can be used to generate highly purified rAAV particles on a small scale, thereby saving resources, generating less waste, and reducing a laboratory's environmental footprint.


Assuntos
Dependovirus/isolamento & purificação , Virologia/métodos , Animais , Vetores Genéticos , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ultracentrifugação
12.
J Virol ; 82(19): 9730-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18667490

RESUMO

Late (L) domains containing the highly conserved sequence PPXY were first described for retroviruses, and later research confirmed their conservation and importance for efficient budding of several negative-stranded RNA viruses. Rabies virus (RV), a member of the Rhabdoviridae family, contains the sequence PPEY (amino acids 35 to 38) within the N terminus of the matrix (M) protein, but the functions of this potential L-domain in the viral life cycle, viral pathogenicity, and immunogenicity have not been established. Here we constructed a series of recombinant RVs containing mutations within the PPEY motif and analyzed their effects on viral replication and RV pathogenicity. Our results indicate that the first proline at position 35 is the most important for viral replication, whereas P36 and Y38 have a lesser but still noticeable impact. The reduction in viral replication was most likely due to inhibition of virion release, because initially no major impact on RV RNA synthesis was observed. In addition, results from electron microscopy demonstrated that the M4A mutant virus (PPEY-->SAEA) displayed a more cell-associated phenotype than that of wild-type RV. Furthermore, all mutations within the PPEY motif resulted in reduced spread of the recombinant RVs as indicated by a reduction in focus size. Importantly, recombinant PPEY L-domain mutants were highly attenuated in mice yet still elicited potent antibody responses against RV G protein that were as high as those observed after infection with wild-type virus. Our data indicate that the RV PPEY motif has L-domain activity essential for efficient virus production and pathogenicity but is not essential for immunogenicity and thus can be targeted to increase the safety of rabies vaccine vectors.


Assuntos
Vírus da Raiva/metabolismo , Vírion/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Primers do DNA/química , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , RNA/química , Vírus da Raiva/genética , Homologia de Sequência de Aminoácidos , Proteínas da Matriz Viral/química
13.
Neuron ; 98(4): 707-717.e4, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29731250

RESUMO

To achieve simultaneous measurement of multiple cellular events in molecularly defined groups of neurons in vivo, we designed a spectrometer-based fiber photometry system that allows for spectral unmixing of multiple fluorescence signals recorded from deep brain structures in behaving animals. Using green and red Ca2+ indicators differentially expressed in striatal direct- and indirect-pathway neurons, we were able to simultaneously monitor the neural activity in these two pathways in freely moving animals. We found that the activities were highly synchronized between the direct and indirect pathways within one hemisphere and were desynchronized between the two hemispheres. We further analyzed the relationship between the movement patterns and the magnitude of activation in direct- and indirect-pathway neurons and found that the striatal direct and indirect pathways coordinately control the dynamics and fate of movement. VIDEO ABSTRACT.


Assuntos
Corpo Estriado/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Espectrometria de Fluorescência/métodos , Animais , Encéfalo/diagnóstico por imagem , Corpo Estriado/diagnóstico por imagem , Proteínas de Fluorescência Verde , Microscopia Intravital , Proteínas Luminescentes , Camundongos , Vias Neurais/diagnóstico por imagem , Imagem Óptica , Fotometria , Proteína Vermelha Fluorescente
14.
Expert Rev Vaccines ; 14(7): 949-62, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25864502

RESUMO

The recent emergence of Middle East respiratory syndrome (MERS) highlights the need to engineer new methods for expediting vaccine development against emerging diseases. However, several obstacles prevent pursuit of a licensable MERS vaccine. First, the lack of a suitable animal model for MERS complicates the in vivo testing of candidate vaccines. Second, due to the low number of MERS cases, pharmaceutical companies have little incentive to pursue MERS vaccine production as the costs of clinical trials are high. In addition, the timeline from bench research to approved vaccine use is 10 years or longer. Using novel methods and cost-saving strategies, genetically engineered vaccines can be produced quickly and cost-effectively. Along with progress in MERS animal model development, these obstacles can be circumvented or at least mitigated.


Assuntos
Infecções por Coronavirus/prevenção & controle , Vacinas Virais/imunologia , Vacinas Virais/isolamento & purificação , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Descoberta de Drogas/economia , Descoberta de Drogas/tendências , Avaliação Pré-Clínica de Medicamentos/economia , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos
15.
Virus Res ; 197: 54-8, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25481284

RESUMO

Using a recombinant rabies (RABV) vaccine platform, we have developed several safe and effective vaccines. Most recently, we have developed a RABV-based ebolavirus (EBOV) vaccine that is efficacious in nonhuman primates. One safety feature of this vaccine is the utilization of a live but replication-deficient RABV construct. In this construct, the RABV glycoprotein (G) has been deleted from the genome, requiring G trans complementation in order for new infectious viruses to be released from the initial infected cell. Here we analyze this safety feature of the bivalent RABV-based EBOV vaccine comprised of the G-deleted RABV backbone expressing EBOV glycoprotein (GP). We found that, while the level of RABV genome in infected cells is equivalent regardless of G supplementation, the production of infectious virus is indeed restricted by the lack of G, and most importantly, that the presence of EBOV GP does not substitute for G. These findings further support the safety profile of this replication-deficient RABV-EBOV bivalent vaccine.


Assuntos
Antígenos Virais/biossíntese , Vacinas contra Ebola/imunologia , Expressão Gênica , Glicoproteínas/biossíntese , Vacina Antirrábica/imunologia , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/imunologia , Animais , Antígenos Virais/genética , Vacinas contra Ebola/genética , Deleção de Genes , Teste de Complementação Genética , Glicoproteínas/genética , Doença pelo Vírus Ebola/prevenção & controle , Primatas , Raiva/prevenção & controle , Vacina Antirrábica/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/genética
16.
Vaccine ; 31(49): 5897-902, 2013 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-24120673

RESUMO

The safety and availability of the human polyclonal sera that is currently utilized for post-exposure treatment (PET) of rabies virus (RABV) infection remain a concern. Recombinant monoclonal antibodies have been postulated as suitable alternatives by WHO. To this extent, CL184, the RABV human antibody combination comprising monoclonal antibodies (mAbs) CR57 and CR4098, has been developed and has delivered promising clinical data to support its use for RABV PET. For this fully human IgG1 cocktail, mAbs CR57 and CR4098 are produced in the PER.C6 human cell line and combined in equal amounts in the final product. During preclinical evaluation, CR57 was shown to bind to antigenic site I whereas CR4098 neutralization was influenced by a mutation of position 336 (N336) located within antigenic site III. Here, alanine scanning was used to analyze the influence of mutations within the potential binding site for CR4098, antigenic site III, in order to evaluate the possibility of mutated rabies viruses escaping neutralization. For this approach, twenty flanking amino acids (10 upstream and 10 downstream) of the RABV glycoprotein (G) asparagine (N336) were exchanged to alanine (or serine, if already alanine) by site-directed mutagenesis. Analysis of G expression revealed four of the twenty mutant Gs to be non-functional, as shown by their lack of cell surface expression, which is a requirement for the production of infectious RABV. Therefore, these mutants were excluded from further study. The remaining sixteen mutants were introduced in an infectious clone of RABV, and recombinant RABVs (rRABVs) were recovered and utilized for in vitro neutralization assays. All of the viruses were effectively neutralized by CR4098 as well as by CR57, indicating that single amino acid exchanges in this region does not affect the broad neutralizing capability of the CL184 mAb combination.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos Virais/genética , Glicoproteínas/genética , Profilaxia Pós-Exposição , Raiva/terapia , Alanina/genética , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Sítios de Ligação de Anticorpos , Linhagem Celular , Cricetinae , Glicoproteínas/imunologia , Humanos , Mutagênese Sítio-Dirigida , Testes de Neutralização , Vacina Antirrábica/uso terapêutico , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
17.
Virology ; 434(1): 18-26, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-22889613

RESUMO

We are developing inactivated and live-attenuated rabies virus (RABV) vaccines expressing Ebola virus (EBOV) glycoprotein for use in humans and endangered wildlife, respectively. Here, we further characterize the pathogenesis of the live-attenuated RABV/EBOV vaccine candidates in mice in an effort to define their growth properties and potential for safety. RABV vaccines expressing GP (RV-GP) or a replication-deficient derivative with a deletion of the RABV G gene (RVΔG-GP) are both avirulent after intracerebral inoculation of adult mice. Furthermore, RVΔG-GP is completely avirulent upon intracerebral inoculation of suckling mice unlike parental RABV vaccine or RV-GP. Analysis of RVΔG-GP in the brain by quantitative PCR, determination of virus titer, and immunohistochemistry indicated greatly restricted virus replication. In summary, our findings indicate that RV-GP retains the attenuation phenotype of the live-attenuated RABV vaccine, and RVΔG-GP would appear to be an even safer alternative for use in wildlife or consideration for human use.


Assuntos
Vacinas contra Ebola/efeitos adversos , Vacinas contra Ebola/imunologia , Vacina Antirrábica/efeitos adversos , Vacina Antirrábica/imunologia , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Encéfalo/virologia , Modelos Animais de Doenças , Vacinas contra Ebola/genética , Feminino , Deleção de Genes , Genes Virais , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Vacina Antirrábica/genética , Reação em Cadeia da Polimerase em Tempo Real , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Carga Viral , Virulência
18.
Vaccine ; 30(43): 6136-41, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22884661

RESUMO

We have previously developed (a) replication-competent, (b) replication-deficient, and (c) chemically inactivated rabies virus (RABV) vaccines expressing Ebola virus (EBOV) glycoprotein (GP) that induce humoral immunity against each virus and confer protection from both lethal RABV and mouse-adapted EBOV challenge in mice. Here, we expand our investigation of the immunogenic properties of these bivalent vaccines in mice. Both live and killed vaccines induced primary EBOV GP-specific T-cells and a robust recall response as measured by interferon-γ ELISPOT assay. In addition to cellular immunity, an effective filovirus vaccine will likely require a multivalent humoral immune response against multiple virus species. As a proof-of-principle experiment, we demonstrated that inactivated RV-GP could be formulated with another inactivated RABV vaccine expressing the nontoxic fragment of botulinum neurotoxin A heavy chain (HC50) without a reduction in immunity to each component. Finally, we demonstrated that humoral immunity to GP could be induced by immunization of mice with inactivated RV-GP in the presence of pre-existing immunity to RABV. The ability of these novel vaccines to induce strong humoral and cellular immunity indicates that they should be further evaluated in additional animal models of infection.


Assuntos
Especificidade de Anticorpos , Vacinas contra Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Glicoproteínas de Membrana/imunologia , Vacina Antirrábica/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Ebolavirus/imunologia , Imunidade Celular , Imunidade Humoral , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T/imunologia , Vacinas de Produtos Inativados/imunologia
19.
Nat Rev Microbiol ; 8(1): 51-61, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19946287

RESUMO

Rabies virus, the prototypical neurotropic virus, causes one of the most lethal zoonotic diseases. According to official estimates, over 55,000 people die of the disease annually, but this is probably a severe underestimation. A combination of virulence factors enables the virus to enter neurons at peripheral sites and travel through the spinal cord to the brain of the infected host, where it often induces aggression that facilitates the transfer of the virus to a new host. This Review summarizes the current knowledge of the replication cycle of rabies virus and virus- host cell interactions, both of which are fundamental elements in our quest to understand the life cycle of rabies virus and the pathogenesis of rabies.


Assuntos
Encéfalo/virologia , Sistema Nervoso Periférico/virologia , Vírus da Raiva/fisiologia , Medula Espinal/virologia , Animais , Humanos , Modelos Biológicos , Vírus da Raiva/patogenicidade , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia
20.
Vaccine ; 28(2): 299-308, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19879223

RESUMO

Highly attenuated rabies virus (RV) vaccine vectors were evaluated for their ability to protect against highly pathogenic SIV(mac251) challenge. Mamu-A*01 negative rhesus macaques were immunized in groups of four with either: RV expressing SIV(mac239)-GagPol, a combination of RV expressing SIV(mac239)-Env and RV expressing SIV(mac239)-GagPol, or with empty RV vectors. Eight weeks later animals received a booster immunization with a heterologous RV expressing the same antigens. At 12 weeks post-boost, all animals were challenged intravenously with 100 TCID(50) of pathogenic SIV(mac251-CX). Immunized macaques in both vaccine groups had 1.3-1.6-log-fold decrease in viral set point compared to control animals. The GagPol/Env immunized animals also had a significantly lower peak viral load. When compared to control animals following challenge, vaccinated macaques had a more rapid induction of SIV(mac251) neutralizing antibodies and of CD8(+) T cell responses to various SIV epitopes. Moreover, vaccinated macaques better maintained peripheral memory CD4(+) T cells and were able to mount a poly-functional CD8(+) T cell response in the mucosa. These findings indicate promise for RV-based vectors and have important implications for the development of an efficacious HIV vaccine.


Assuntos
Anticorpos Neutralizantes/imunologia , Linfócitos T CD8-Positivos/imunologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Vírus da Raiva/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Animais , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Vírus da Imunodeficiência Símia/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA