Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Virol J ; 8: 205, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21548931

RESUMO

BACKGROUND: Lassa virus (LASV) infection causes an acute and sometimes fatal hemorrhagic disease in humans and nonhuman primates; however, little is known about the development of Lassa fever. Here, we performed a pilot study to begin to understand the progression of LASV infection in nonhuman primates. METHODS: Six cynomolgus monkeys were experimentally infected with LASV. Tissues from three animals were examined at an early- to mid-stage of disease and compared with tissues from three animals collected at terminal stages of disease. RESULTS: Dendritic cells were identified as a prominent target of LASV infection in a variety of tissues in all animals at day 7 while Kupffer cells, hepatocytes, adrenal cortical cells, and endothelial cells were more frequently infected with LASV in tissues of terminal animals (days 13.5-17). Meningoencephalitis and neuronal necrosis were noteworthy findings in terminal animals. Evidence of coagulopathy was noted; however, the degree of fibrin deposition in tissues was less prominent than has been reported in other viral hemorrhagic fevers. CONCLUSION: The sequence of pathogenic events identified in this study begins to shed light on the development of disease processes during Lassa fever and also may provide new targets for rational prophylactic and chemotherapeutic interventions.


Assuntos
Febre Lassa/veterinária , Vírus Lassa/patogenicidade , Doenças dos Primatas/patologia , Doenças dos Primatas/virologia , Experimentação Animal , Estruturas Animais/patologia , Estruturas Animais/virologia , Animais , Imuno-Histoquímica , Febre Lassa/patologia , Febre Lassa/virologia , Macaca fascicularis , Microscopia
2.
J Virol ; 83(14): 7296-304, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19386702

RESUMO

The filoviruses Marburg virus and Ebola virus cause severe hemorrhagic fever with high mortality in humans and nonhuman primates. Among the most promising filovirus vaccines under development is a system based on recombinant vesicular stomatitis virus (VSV) that expresses a single filovirus glycoprotein (GP) in place of the VSV glycoprotein (G). Here, we performed a proof-of-concept study in order to determine the potential of having one single-injection vaccine capable of protecting nonhuman primates against Sudan ebolavirus (SEBOV), Zaire ebolavirus (ZEBOV), Cote d'Ivoire ebolavirus (CIEBOV), and Marburgvirus (MARV). In this study, 11 cynomolgus monkeys were vaccinated with a blended vaccine consisting of equal parts of the vaccine vectors VSVDeltaG/SEBOVGP, VSVDeltaG/ZEBOVGP, and VSVDeltaG/MARVGP. Four weeks later, three of these animals were challenged with MARV, three with CIEBOV, three with ZEBOV, and two with SEBOV. Three control animals were vaccinated with VSV vectors encoding a nonfilovirus GP and challenged with SEBOV, ZEBOV, and MARV, respectively, and five unvaccinated control animals were challenged with CIEBOV. Importantly, none of the macaques vaccinated with the blended vaccine succumbed to a filovirus challenge. As expected, an experimental control animal vaccinated with VSVDeltaG/ZEBOVGP and challenged with SEBOV succumbed, as did the positive controls challenged with SEBOV, ZEBOV, and MARV, respectively. All five control animals challenged with CIEBOV became severely ill, and three of the animals succumbed on days 12, 12, and 14, respectively. The two animals that survived CIEBOV infection were protected from subsequent challenge with either SEBOV or ZEBOV, suggesting that immunity to CIEBOV may be protective against other species of Ebola virus. In conclusion, we developed an immunization scheme based on a single-injection vaccine that protects nonhuman primates against lethal challenge with representative strains of all human pathogenic filovirus species.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Primatas/imunologia , Vacinas Virais/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Modelos Animais de Doenças , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Humanos , Imunização , Imunoglobulina G/sangue , Macaca fascicularis , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Primatas/virologia , Distribuição Aleatória , Vacinas Virais/imunologia
3.
PLoS Pathog ; 4(11): e1000225, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19043556

RESUMO

Ebola virus (EBOV) is a significant human pathogen that presents a public health concern as an emerging/re-emerging virus and as a potential biological weapon. Substantial progress has been made over the last decade in developing candidate preventive vaccines that can protect nonhuman primates against EBOV. Among these prospects, a vaccine based on recombinant vesicular stomatitis virus (VSV) is particularly robust, as it can also confer protection when administered as a postexposure treatment. A concern that has been raised regarding the replication-competent VSV vectors that express EBOV glycoproteins is how these vectors would be tolerated by individuals with altered or compromised immune systems such as patients infected with HIV. This is especially important as all EBOV outbreaks to date have occurred in areas of Central and Western Africa with high HIV incidence rates in the population. In order to address this concern, we evaluated the safety of the recombinant VSV vector expressing the Zaire ebolavirus glycoprotein (VSVDeltaG/ZEBOVGP) in six rhesus macaques infected with simian-human immunodeficiency virus (SHIV). All six animals showed no evidence of illness associated with the VSVDeltaG/ZEBOVGP vaccine, suggesting that this vaccine may be safe in immunocompromised populations. While one goal of the study was to evaluate the safety of the candidate vaccine platform, it was also of interest to determine if altered immune status would affect vaccine efficacy. The vaccine protected 4 of 6 SHIV-infected macaques from death following ZEBOV challenge. Evaluation of CD4+ T cells in all animals showed that the animals that succumbed to lethal ZEBOV challenge had the lowest CD4+ counts, suggesting that CD4+ T cells may play a role in mediating protection against ZEBOV.


Assuntos
Vacinas contra Ebola/farmacologia , Hospedeiro Imunocomprometido , Estomatite Vesicular , Animais , Linfócitos T CD4-Positivos , Avaliação Pré-Clínica de Medicamentos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Vacinas contra Ebola/administração & dosagem , Vetores Genéticos , Macaca mulatta , Primatas , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Resultado do Tratamento , Proteínas Virais
4.
J Virol ; 82(11): 5664-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18385248

RESUMO

Recombinant vesicular stomatitis virus (VSV) vectors expressing homologous filoviral glycoproteins can completely protect rhesus monkeys against Marburg virus when administered after exposure and can partially protect macaques after challenge with Zaire ebolavirus. Here, we administered a VSV vector expressing the Sudan ebolavirus (SEBOV) glycoprotein to four rhesus macaques shortly after exposure to SEBOV. All four animals survived SEBOV challenge, while a control animal that received a nonspecific vector developed fulminant SEBOV hemorrhagic fever and succumbed. This is the first demonstration of complete postexposure protection against an Ebola virus in nonhuman primates and provides further evidence that postexposure vaccination may have utility in treating exposures to filoviruses.


Assuntos
Ebolavirus/fisiologia , Vetores Genéticos/genética , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/prevenção & controle , Macaca mulatta/virologia , Vesiculovirus/genética , Animais , Doença pelo Vírus Ebola/genética , Macaca mulatta/sangue , Taxa de Sobrevida
5.
PLoS Pathog ; 3(1): e2, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17238284

RESUMO

Ebola viruses are highly lethal human pathogens that have received considerable attention in recent years due to an increasing re-emergence in Central Africa and a potential for use as a biological weapon. There is no vaccine or treatment licensed for human use. In the past, however, important advances have been made in developing preventive vaccines that are protective in animal models. In this regard, we showed that a single injection of a live-attenuated recombinant vesicular stomatitis virus vector expressing the Ebola virus glycoprotein completely protected rodents and nonhuman primates from lethal Ebola challenge. In contrast, progress in developing therapeutic interventions against Ebola virus infections has been much slower and there is clearly an urgent need to develop effective post-exposure strategies to respond to future outbreaks and acts of bioterrorism, as well as to treat laboratory exposures. Here we tested the efficacy of the vesicular stomatitis virus-based Ebola vaccine vector in post-exposure treatment in three relevant animal models. In the guinea pig and mouse models it was possible to protect 50% and 100% of the animals, respectively, following treatment as late as 24 h after lethal challenge. More important, four out of eight rhesus macaques were protected if treated 20 to 30 min following an otherwise uniformly lethal infection. Currently, this approach provides the most effective post-exposure treatment strategy for Ebola infections and is particularly suited for use in accidentally exposed individuals and in the control of secondary transmission during naturally occurring outbreaks or deliberate release.


Assuntos
Vacinas contra Ebola/uso terapêutico , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/terapia , Animais , Surtos de Doenças , Cobaias , Humanos , Macaca mulatta , Camundongos , Dados de Sequência Molecular , Resultado do Tratamento
6.
Lancet ; 367(9520): 1399-404, 2006 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-16650649

RESUMO

BACKGROUND: Effective countermeasures are urgently needed to prevent and treat infections caused by highly pathogenic and biological threat agents such as Marburg virus (MARV). We aimed to test the efficacy of a replication-competent vaccine based on attenuated recombinant vesicular stomatitis virus (rVSV), as a postexposure treatment for MARV haemorrhagic fever. METHODS: We used a rhesus macaque model of MARV haemorrhagic fever that produced 100% lethality. We administered rVSV vectors expressing the MARV Musoke strain glycoprotein to five macaques 20-30 min after a high-dose lethal injection of homologous MARV. Three animals were MARV-positive controls and received non-specific rVSV vectors. We tested for viraemia, undertook analyses for haematology and serum biochemistry, and measured humoral and cellular immune responses. FINDINGS: All five rhesus monkeys that were treated with the rVSV MARV vectors as a postexposure treatment survived a high-dose lethal challenge of MARV for at least 80 days. None of these five animals developed clinical symptoms consistent with MARV haemorrhagic fever. All the control animals developed fulminant disease and succumbed to the MARV challenge by day 12. MARV disease in the controls was indicated by: high titres of MARV (10(3)-10(5) plaque-forming units per mL); development of leucocytosis with concurrent neutrophilia at end-stage disease; and possible damage to the liver, kidney, and pancreas. INTERPRETATION: Postexposure protection against MARV in non-human primates provides a paradigm for the treatment of MARV haemorrhagic fever. Indeed, these data suggest that rVSV-based filoviral vaccines might not only have potential as preventive vaccines, but also could be equally useful for postexposure treatment of filoviral infections.


Assuntos
Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vacinas Virais , Animais , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Macaca mulatta , Doença do Vírus de Marburg/imunologia
7.
PLoS One ; 5(5): e10690, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20502528

RESUMO

Nipah virus (NiV) is an enigmatic emerging pathogen that causes severe and often fatal neurologic and/or respiratory disease in both animals and humans. Amongst people, case fatality rates range between 40 and 75 percent and there are no vaccines or treatments approved for human use. Guinea pigs, hamsters, cats, ferrets, pigs and most recently squirrel monkeys (New World monkey) have been evaluated as animal models of human NiV infection, and with the exception of the ferret, no model recapitulates all aspects of NiV-mediated disease seen in humans. To identify a more viable nonhuman primate (NHP) model, we examined the pathogenesis of NiV in African green monkeys (AGM). Exposure of eight monkeys to NiV produced a severe systemic infection in all eight animals with seven of the animals succumbing to infection. Viral RNA was detected in the plasma of challenged animals and occurred in two of three subjects as a peak between days 7 and 21, providing the first clear demonstration of plasma-associated viremia in NiV experimentally infected animals and suggested a progressive infection that seeded multiple organs simultaneously from the initial site of virus replication. Unlike the cat, hamster and squirrel monkey models of NiV infection, severe respiratory pathology, neurological disease and generalized vasculitis all manifested in NiV-infected AGMs, providing an accurate reflection of what is observed in NiV-infected humans. Our findings demonstrate the first consistent and highly pathogenic NHP model of NiV infection, providing a new and critical platform in the evaluation and licensure of either passive and active immunization or therapeutic strategies for human use.


Assuntos
Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Vírus Nipah/patogenicidade , Primatas/imunologia , Primatas/virologia , Animais , Autorradiografia , Chlorocebus aethiops , Infecções por Henipavirus/sangue , Humanos , Modelos Imunológicos , Vírus Nipah/fisiologia , Especificidade de Órgãos/imunologia , RNA Viral/sangue , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Viral/imunologia , Tropismo Viral/imunologia
8.
Vaccine ; 26(52): 6894-900, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-18930776

RESUMO

Considerable progress has been made over the last decade in developing candidate preventive vaccines that can protect nonhuman primates against Ebola and Marburg viruses. A vaccine based on recombinant vesicular stomatitis virus (VSV) seems to be particularly robust as it can also confer protection when administered as a postexposure treatment. While filoviruses are not thought to be transmitted by aerosol in nature the inhalation route is among the most likely portals of entry in the setting of a bioterrorist event. At present, all candidate filoviral vaccines have been evaluated against parenteral challenges but none have been tested against an aerosol exposure. Here, we evaluated our recombinant VSV-based Zaire ebolavirus (ZEBOV) and Marburg virus (MARV) vaccines against aerosol challenge in cynomolgus macaques. All monkeys vaccinated with a VSV vector expressing the glycoprotein of ZEBOV were completely protected against an aerosol exposure of ZEBOV. Likewise, all monkeys vaccinated with a VSV vector expressing the glycoprotein of MARV were completely protected against an aerosol exposure of MARV. All control animals challenged by the aerosol route with either ZEBOV or MARV succumbed. Interestingly, disease in control animals appeared to progress slower than previously seen in macaques exposed to comparable doses by intramuscular injection.


Assuntos
Vacinas contra Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/veterinária , Macaca fascicularis/imunologia , Doença do Vírus de Marburg/prevenção & controle , Doenças dos Macacos/prevenção & controle , Vírus da Estomatite Vesicular Indiana/imunologia , Vacinas Virais/imunologia , Animais , Formação de Anticorpos/imunologia , Ebolavirus/imunologia , Humanos , Imunidade Celular/imunologia , Masculino , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , RNA Viral/análise , RNA Viral/biossíntese , Vacinas Sintéticas , Viremia/imunologia , Viremia/virologia
9.
J Virol ; 81(6): 2995-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17229700

RESUMO

Enveloped viruses often require cleavage of a surface glycoprotein by a cellular endoprotease such as furin for infectivity and virulence. Previously, we showed that Ebola virus glycoprotein does not require the furin cleavage motif for virus replication in cell culture. Here, we show that there are no appreciable differences in disease progression, hematology, serum biochemistry, virus titers, or lethality in nonhuman primates infected with an Ebola virus lacking the furin recognition sequence compared to those infected with wild-type virus. We conclude that glycoprotein cleavage by subtilisin-like endoproteases is not critical for Ebola virus infectivity and virulence in nonhuman primates.


Assuntos
Ebolavirus/metabolismo , Ebolavirus/fisiologia , Processamento de Proteína Pós-Traducional , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência Consenso , Ebolavirus/química , Ebolavirus/genética , Macaca mulatta , Dados de Sequência Molecular , Mutação , RNA Viral/genética , Virulência
10.
J Infect Dis ; 196 Suppl 2: S323-8, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17940967

RESUMO

Zaire Ebola virus infection in macaques causes a fatal disease with a pathogenesis similar to that in humans. During several independent therapy studies, we noted altered tissue tropism in 6 rhesus macaques that survived longer than those with a typical disease course. The mean time to death for these 6 macaques was 21.7 days, which is significantly longer than the average mean time to death of 8.3 days for 20 untreated historical control animals. In addition to living significantly longer, these 6 animals exhibited a variety of deteriorating clinical signs with pathologic findings that were not seen in the untreated control animals, as well as the presence of viral antigen in the brain, eye, pancreas, thyroid, and lung. We suggest that treatment extended the time course of the disease and permitted the virus to infect tissues not usually affected in the typical model.


Assuntos
Doença pelo Vírus Ebola/patologia , Doenças dos Primatas/virologia , Animais , República Democrática do Congo , Modelos Animais de Doenças , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/transmissão , Hipocampo/patologia , Hipocampo/virologia , Macaca mulatta
11.
J Infect Dis ; 196 Suppl 2: S372-81, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17940973

RESUMO

BACKGROUND: The procoagulant tissue factor (TF) is thought to play a role in the coagulation disorders that characterize filoviral infections. In this study, we evaluated the pathogenesis of lethal infection with the Angola strain of Marburg virus (MARV-Ang) in rhesus macaques and tested the efficacy of recombinant nematode anticoagulant protein c2 (rNAPc2), an inhibitor of TF/factor VIIa, as a potential treatment. METHODS: Twelve rhesus macaques were challenged with a high dose (1000 pfu) of MARV-Ang. Six macaques were treated with rNAPc2, and 6 macaques served as control animals. RESULTS: All 6 control animals succumbed to MARV-Ang challenge by day 8 (mean, 7.3 days), whereas 5 of 6 rNAPc2-treated animals died on day 9 and 1 rNAPc2-treated animal survived. The disease course for MARV-Ang infection appeared to progress more rapidly in rhesus macaques than has been previously reported for other strains of MARV. In contrast to Ebola virus (EBOV) infection in macaques, up-regulation of TF was not as striking, and deposition of fibrin was a less prominent pathologic feature of disease in these animals. CONCLUSIONS: These data show that the pathogenicity of MARV-Ang infection appears to be consistent with the apparent increased human virulence attributed to this strain. The apparent reduced efficacy of rNAPc2 against MARV-Ang infection, compared with its efficacy against EBOV infection, appears to be associated with differences in TF induction and fibrin deposition.


Assuntos
Proteínas de Helminto/uso terapêutico , Doença do Vírus de Marburg/tratamento farmacológico , Doença do Vírus de Marburg/epidemiologia , Marburgvirus/patogenicidade , Angola/epidemiologia , Animais , Macaca mulatta , Doenças dos Primatas/tratamento farmacológico , Doenças dos Primatas/epidemiologia , Proteínas Recombinantes/uso terapêutico
12.
J Infect Dis ; 196 Suppl 2: S390-9, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17940975

RESUMO

BACKGROUND: Infection of primates with Zaire ebolavirus (ZEBOV) leads to hypotension, coagulation disorders, and an impaired immune response and, in many ways, resembles severe sepsis. Rapid decreases in plasma levels of protein C are a prominent feature of severe sepsis and ZEBOV hemorrhagic fever (ZHF). Currently, recombinant human activated protein C (rhAPC [Xigris; Eli Lilly]) is licensed for treating human patients with severe sepsis who are at high risk of death. The aim of this study was to test the efficacy of rhAPC as a potential treatment for ZHF. METHODS: Fourteen rhesus macaques were challenged with a uniformly lethal dose of ZEBOV; 11 of these monkeys were treated by intravenous infusion with rhAPC beginning 30-60 min after challenge and continuing for 7 days. Three control monkeys received sterile saline in parallel. RESULTS: All 3 control monkeys died on day 8, whereas 2 of the 11 rhAPC-treated monkeys survived. The mean time to death for the rhAPC-treated monkeys that did not survive ZEBOV challenge was 12.6 days. The difference in survival was significant when the rhAPC-treated monkeys were compared with historical controls. CONCLUSIONS: The experimental findings provide evidence that ZHF and severe sepsis share underlying mechanisms and may respond to the same therapies.


Assuntos
Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/tratamento farmacológico , Proteína C/uso terapêutico , Animais , Cateterismo Venoso Central , Modelos Animais de Doenças , Macaca mulatta , Oligopeptídeos/uso terapêutico , Primatas , Proteína C/administração & dosagem , Proteína C/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico
13.
Genome Biol ; 8(8): R174, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17725815

RESUMO

BACKGROUND: Infection with Ebola virus (EBOV) causes a fulminant and often fatal hemorrhagic fever. In order to improve our understanding of EBOV pathogenesis and EBOV-host interactions, we examined the molecular features of EBOV infection in vivo. RESULTS: Using high-density cDNA microarrays, we analyzed genome-wide host expression patterns in sequential blood samples from nonhuman primates infected with EBOV. The temporal program of gene expression was strikingly similar between animals. Of particular interest were features of the data that reflect the interferon response, cytokine signaling, and apoptosis. Transcript levels for tumor necrosis factor-alpha converting enzyme (TACE)/alpha-disintegrin and metalloproteinase (ADAM)-17 increased during days 4 to 6 after infection. In addition, the serum concentration of cleaved Ebola glycoprotein (GP2 delta) was elevated in late-stage EBOV infected animals. Of note, we were able to detect changes in gene expression of more than 300 genes before symptoms appeared. CONCLUSION: These results provide the first genome-wide ex vivo analysis of the host response to systemic filovirus infection and disease. These data may elucidate mechanisms of viral pathogenesis and host defense, and may suggest targets for diagnostic and therapeutic development.


Assuntos
Perfilação da Expressão Gênica , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/genética , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Apoptose/genética , Fibrina/metabolismo , Imunidade Inata/genética , Interferons/metabolismo , Leucócitos Mononucleares/metabolismo , Macaca fascicularis , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas do Envelope Viral/sangue , Proteínas do Envelope Viral/metabolismo
14.
J Virol ; 80(19): 9659-66, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973570

RESUMO

Marburg virus (MARV) has been associated with sporadic episodes of hemorrhagic fever, including a recent highly publicized outbreak in Angola that produced severe disease and significant mortality in infected patients. MARV is also considered to have potential as a biological weapon. Recently, we reported the development of a promising attenuated, replication-competent vaccine against MARV based on recombinant vesicular stomatitis virus (VSV) expressing the glycoprotein of the Musoke strain of MARV (VSVDeltaG/MARVGP-Musoke). We used this vaccine to demonstrate complete protection of cynomolgus monkeys against a homologous MARV challenge. While these results are highly encouraging, an effective vaccine would need to confer protection against all relevant strains of MARV. Here, we evaluated the protective efficacy of the VSVDeltaG/MARVGP-Musoke vaccine against two heterologous MARV strains, the seemingly more pathogenic Angola strain and the more distantly related Ravn strain. In this study, seven cynomolgus monkeys were vaccinated with the VSVDeltaG/MARVGP-Musoke vector. Three of these animals were challenged with the Angola strain, three with the Ravn strain, and a single animal with the Musoke strain of MARV. Two animals served as controls and were each injected with a nonspecific VSV vector; these controls were challenged with the Angola and Ravn strains, respectively. Both controls succumbed to challenge by day 8. However, none of the specifically vaccinated animals showed any evidence of illness either from the vaccination or from the MARV challenges and all of these animals survived. These data suggest that the VSVDeltaG/MARVGP-Musoke vaccine should be sufficient to protect against all known MARV strains.


Assuntos
Doença do Vírus de Marburg/imunologia , Marburgvirus/classificação , Marburgvirus/imunologia , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos/imunologia , Reações Cruzadas/imunologia , Macaca fascicularis , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/metabolismo , Marburgvirus/genética , Marburgvirus/metabolismo , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Viremia/sangue , Viremia/imunologia , Viremia/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA