Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Viruses ; 13(4)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33807214

RESUMO

Mastomys natalensis are a ubiquitous and often dominant rodent across sub-Saharan Africa. Importantly, they are a natural reservoir for microbial pathogens including Lassa virus (LASV), the etiological agent of Lassa fever in humans. Lassa-infected rodents have been documented across West Africa and coincide with regions where annual outbreaks occur. Zoonotic transmission to humans most often occurs directly from infected rodents. Little is known about LASV infection kinetics and transmissibility in M.natalensis, primarily due to available animals. Here, we describe the establishment of a laboratory breeding colony of genetically confirmed M.natalensis from wild-captured rodents. This colony will provide a convenient source of animals to study LASV and other emerging pathogens that utilize M. natalensis in their enzootic lifecycles.


Assuntos
Animais Selvagens/genética , Murinae/genética , Seleção Artificial , África Ocidental , Animais , Animais Selvagens/virologia , Feminino , Febre Lassa/transmissão , Vírus Lassa/patogenicidade , Masculino , Modelos Animais , Murinae/fisiologia , Murinae/virologia
2.
NPJ Vaccines ; 6(1): 32, 2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33654106

RESUMO

Lassa virus (LASV) infects hundreds of thousands of individuals each year, highlighting the need for the accelerated development of preventive, diagnostic, and therapeutic interventions. To date, no vaccine has been licensed for LASV. ChAdOx1-Lassa-GPC is a chimpanzee adenovirus-vectored vaccine encoding the Josiah strain LASV glycoprotein precursor (GPC) gene. In the following study, we show that ChAdOx1-Lassa-GPC is immunogenic, inducing robust T-cell and antibody responses in mice. Furthermore, a single dose of ChAdOx1-Lassa-GPC fully protects Hartley guinea pigs against morbidity and mortality following lethal challenge with a guinea pig-adapted LASV (strain Josiah). By contrast, control vaccinated animals reached euthanasia criteria 10-12 days after infection. Limited amounts of LASV RNA were detected in the tissues of vaccinated animals. Viable LASV was detected in only one animal receiving a single dose of the vaccine. A prime-boost regimen of ChAdOx1-Lassa-GPC in guinea pigs significantly increased antigen-specific antibody titers and cleared viable LASV from the tissues. These data support further development of ChAdOx1-Lassa-GPC and testing in non-human primate models of infection.

3.
PLoS Negl Trop Dis ; 13(6): e0007462, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31170144

RESUMO

Nipah virus (NiV) is a highly pathogenic re-emerging virus that causes outbreaks in South East Asia. Currently, no approved and licensed vaccine or antivirals exist. Here, we investigated the efficacy of ChAdOx1 NiVB, a simian adenovirus-based vaccine encoding NiV glycoprotein (G) Bangladesh, in Syrian hamsters. Prime-only as well as prime-boost vaccination resulted in uniform protection against a lethal challenge with NiV Bangladesh: all animals survived challenge and we were unable to find infectious virus either in oral swabs, lung or brain tissue. Furthermore, no pathological lung damage was observed. A single-dose of ChAdOx1 NiVB also prevented disease and lethality from heterologous challenge with NiV Malaysia. While we were unable to detect infectious virus in swabs or tissue of animals challenged with the heterologous strain, a very limited amount of viral RNA could be found in lung tissue by in situ hybridization. A single dose of ChAdOx1 NiVB also provided partial protection against Hendra virus and passive transfer of antibodies elicited by ChAdOx1 NiVB vaccination partially protected Syrian hamsters against NiV Bangladesh. From these data, we conclude that ChAdOx1 NiVB is a suitable candidate for further NiV vaccine pre-clinical development.


Assuntos
Adenovirus dos Símios/genética , Portadores de Fármacos , Infecções por Henipavirus/prevenção & controle , Vírus Nipah/imunologia , Vacinas Virais/imunologia , Estruturas Animais/virologia , Animais , Modelos Animais de Doenças , Feminino , Infecções por Henipavirus/imunologia , Mesocricetus , Vírus Nipah/genética , Análise de Sobrevida , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
4.
Sci Transl Med ; 7(301): 301ra132, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26290414

RESUMO

First identified in 2012, Middle East respiratory syndrome (MERS) is caused by an emerging human coronavirus, which is distinct from the severe acute respiratory syndrome coronavirus (SARS-CoV), and represents a novel member of the lineage C betacoronoviruses. Since its identification, MERS coronavirus (MERS-CoV) has been linked to more than 1372 infections manifesting with severe morbidity and, often, mortality (about 495 deaths) in the Arabian Peninsula, Europe, and, most recently, the United States. Human-to-human transmission has been documented, with nosocomial transmission appearing to be an important route of infection. The recent increase in cases of MERS in the Middle East coupled with the lack of approved antiviral therapies or vaccines to treat or prevent this infection are causes for concern. We report on the development of a synthetic DNA vaccine against MERS-CoV. An optimized DNA vaccine encoding the MERS spike protein induced potent cellular immunity and antigen-specific neutralizing antibodies in mice, macaques, and camels. Vaccinated rhesus macaques seroconverted rapidly and exhibited high levels of virus-neutralizing activity. Upon MERS viral challenge, all of the monkeys in the control-vaccinated group developed characteristic disease, including pneumonia. Vaccinated macaques were protected and failed to demonstrate any clinical or radiographic signs of pneumonia. These studies demonstrate that a consensus MERS spike protein synthetic DNA vaccine can induce protective responses against viral challenge, indicating that this strategy may have value as a possible vaccine modality against this emerging pathogen.


Assuntos
Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vacinas de DNA/uso terapêutico , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Camelus , Macaca mulatta , Camundongos
5.
Science ; 346(6212): 987-91, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25359852

RESUMO

Existing mouse models of lethal Ebola virus infection do not reproduce hallmark symptoms of Ebola hemorrhagic fever, neither delayed blood coagulation and disseminated intravascular coagulation nor death from shock, thus restricting pathogenesis studies to nonhuman primates. Here we show that mice from the Collaborative Cross panel of recombinant inbred mice exhibit distinct disease phenotypes after mouse-adapted Ebola virus infection. Phenotypes range from complete resistance to lethal disease to severe hemorrhagic fever characterized by prolonged coagulation times and 100% mortality. Inflammatory signaling was associated with vascular permeability and endothelial activation, and resistance to lethal infection arose by induction of lymphocyte differentiation and cellular adhesion, probably mediated by the susceptibility allele Tek. These data indicate that genetic background determines susceptibility to Ebola hemorrhagic fever.


Assuntos
Modelos Animais de Doenças , Predisposição Genética para Doença , Doença pelo Vírus Ebola/genética , Doença pelo Vírus Ebola/imunologia , Interações Hospedeiro-Patógeno/genética , Camundongos , Receptor TIE-2/genética , Alelos , Animais , Coagulação Sanguínea/genética , Permeabilidade Capilar/genética , Endotélio Vascular/fisiopatologia , Doença pelo Vírus Ebola/sangue , Fígado/irrigação sanguínea , Fígado/metabolismo , Fígado/patologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/genética
6.
PLoS Pathog ; 10(8): e1004250, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25144235

RESUMO

The availability of a robust disease model is essential for the development of countermeasures for Middle East respiratory syndrome coronavirus (MERS-CoV). While a rhesus macaque model of MERS-CoV has been established, the lack of uniform, severe disease in this model complicates the analysis of countermeasure studies. Modeling of the interaction between the MERS-CoV spike glycoprotein and its receptor dipeptidyl peptidase 4 predicted comparable interaction energies in common marmosets and humans. The suitability of the marmoset as a MERS-CoV model was tested by inoculation via combined intratracheal, intranasal, oral and ocular routes. Most of the marmosets developed a progressive severe pneumonia leading to euthanasia of some animals. Extensive lesions were evident in the lungs of all animals necropsied at different time points post inoculation. Some animals were also viremic; high viral loads were detected in the lungs of all infected animals, and total RNAseq demonstrated the induction of immune and inflammatory pathways. This is the first description of a severe, partially lethal, disease model of MERS-CoV, and as such will have a major impact on the ability to assess the efficacy of vaccines and treatment strategies as well as allowing more detailed pathogenesis studies.


Assuntos
Infecções por Coronavirus/patologia , Modelos Animais de Doenças , Pneumonia Viral/patologia , Animais , Callithrix , Infecções por Coronavirus/virologia , Imuno-Histoquímica , Masculino , Coronavírus da Síndrome Respiratória do Oriente Médio , Pneumonia Viral/virologia , Reação em Cadeia da Polimerase em Tempo Real
7.
Virology ; 449: 109-19, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24418544

RESUMO

The intracellular transport of Mason-Pfizer monkey virus (M-PMV) assembled capsids from the pericentriolar region to the plasma membrane (PM) requires trafficking of envelope glycoprotein (Env) to the assembly site via the recycling endosome. However, it is unclear if Env-containing vesicles play a direct role in trafficking capsids to the PM. Using live cell microscopy, we demonstrate, for the first time, anterograde co-transport of Gag and Env. Nocodazole disruption of microtubules had differential effects on Gag and Env trafficking, with pulse-chase assays showing a delayed release of Env-deficient virions. Particle tracking demonstrated an initial loss of linear movement of GFP-tagged capsids and mCherry-tagged Env, followed by renewed movement of Gag but not Env at 4h post-treatment. Thus, while delayed capsid trafficking can occur in the absence of microtubules, efficient anterograde transport of capsids appears to be mediated by microtubule-associated Env-containing vesicles.


Assuntos
Produtos do Gene env/metabolismo , Produtos do Gene gag/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Microtúbulos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Animais , Membrana Celular/virologia , Chlorocebus aethiops , Produtos do Gene env/genética , Produtos do Gene gag/genética , Macaca mulatta , Vírus dos Macacos de Mason-Pfizer/genética , Microtúbulos/metabolismo , Transporte Proteico , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo
8.
Virulence ; 4(8): 707-15, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24104465

RESUMO

Staphylococcus aureus community-acquired pneumonia is often associated with influenza or an influenza-like syndrome. Morbidity and mortality due to methicillin-resistant S. aureus (MRSA) or influenza and pneumonia, which includes bacterial co-infection, are among the top causes of death by infectious diseases in the United States. We developed a non-lethal influenza A virus (IAV) (H3N2)/S. aureus co-infection model in cynomolgus macaques (Macaca fascicularis) to test the hypothesis that seasonal IAV infection predisposes non-human primates to severe S. aureus pneumonia. Infection and disease progression were monitored by clinical assessment of animal health; analysis of blood chemistry, nasal swabs, and X-rays; and gross pathology and histopathology of lungs from infected animals. Seasonal IAV infection in healthy cynomolgus macaques caused mild pneumonia, but unexpectedly, did not predispose these animals to subsequent severe infection with the community-associated MRSA clone USA300. We conclude that in our co-infection model, seasonal IAV infection alone is not sufficient to promote severe S. aureus pneumonia in otherwise healthy non-human primates. The implication of these findings is that comorbidity factors in addition to IAV infection are required to predispose individuals to secondary S. aureus pneumonia.


Assuntos
Coinfecção/microbiologia , Coinfecção/virologia , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Interações Microbianas , Infecções por Orthomyxoviridae/complicações , Pneumonia Estafilocócica/complicações , Staphylococcus aureus/crescimento & desenvolvimento , Animais , Coinfecção/patologia , Modelos Animais de Doenças , Feminino , Humanos , Pulmão/patologia , Macaca fascicularis , Masculino , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/patologia
9.
J Infect Dis ; 207(2): 306-18, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23045629

RESUMO

Ebola hemorrhagic fever (EHF) is a severe viral infection for which no effective treatment or vaccine is currently available. While the nonhuman primate (NHP) model is used for final evaluation of experimental vaccines and therapeutic efficacy, rodent models have been widely used in ebolavirus research because of their convenience. However, the validity of rodent models has been questioned given their low predictive value for efficacy testing of vaccines and therapeutics, a result of the inconsistent manifestation of coagulopathy seen in EHF. Here, we describe a lethal Syrian hamster model of EHF using mouse-adapted Ebola virus. Infected hamsters displayed most clinical hallmarks of EHF, including severe coagulopathy and uncontrolled host immune responses. Thus, the hamster seems to be superior to the existing rodent models, offering a better tool for understanding the critical processes in pathogenesis and providing a new model for evaluating prophylactic and postexposure interventions prior to testing in NHPs.


Assuntos
Modelos Animais de Doenças , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/fisiopatologia , Mesocricetus , Animais , Coagulação Sanguínea , Chlorocebus aethiops , Cricetinae , Coagulação Intravascular Disseminada , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/mortalidade , Doença pelo Vírus Ebola/virologia , Humanos , Masculino , Camundongos , Células Vero
10.
Sci Transl Med ; 4(146): 146ra107, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22875827

RESUMO

In the 1990s, Hendra virus and Nipah virus (NiV), two closely related and previously unrecognized paramyxoviruses that cause severe disease and death in humans and a variety of animals, were discovered in Australia and Malaysia, respectively. Outbreaks of disease have occurred nearly every year since NiV was first discovered, with case fatality ranging from 10 to 100%. In the African green monkey (AGM), NiV causes a severe lethal respiratory and/or neurological disease that essentially mirrors fatal human disease. Thus, the AGM represents a reliable disease model for vaccine and therapeutic efficacy testing. We show that vaccination of AGMs with a recombinant subunit vaccine based on the henipavirus attachment G glycoprotein affords complete protection against subsequent NiV infection with no evidence of clinical disease, virus replication, or pathology observed in any challenged subjects. Success of the recombinant subunit vaccine in nonhuman primates provides crucial data in supporting its further preclinical development for potential human use.


Assuntos
Antígenos Virais/imunologia , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Glicoproteínas/imunologia , Vírus Hendra/imunologia , Vírus Nipah/imunologia , Animais , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/prevenção & controle , Vírus Nipah/patogenicidade
11.
PLoS Pathog ; 7(12): e1002426, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22194683

RESUMO

Hantavirus pulmonary syndrome (HPS), also referred to as hantavirus cardiopulmonary syndrome (HCPS), is a rare but frequently fatal disease caused by New World hantaviruses. In humans HPS is associated with severe pulmonary edema and cardiogenic shock; however, the pathogenesis of this disease remains unclear largely due to a lack of suitable animal models for the study of disease progression. In this study we monitored clinical, virological, pathophysiological parameters and host immunological responses to decipher pathological factors and events in the lethal Syrian hamster model of HPS following intranasal inoculation of Andes virus. Transcriptional profiling of the host gene responses demonstrated a suppression of innate immune responses in most organs analyzed during the early stage of infection, except for in the lung which had low level activation of several pro-inflammatory genes. During this phase Andes virus established a systemic infection in hamsters, with viral antigen readily detectable in the endothelium of the majority of tissues analyzed by 7-8 days post-inoculation. Despite wide-spread infection, histological analysis confirmed pathological abnormalities were almost exclusively found in the lungs. Immediately preceding clinical signs of disease, intense activation of pro-inflammatory and Th1/Th2 responses were observed in the lungs as well as the heart, but not in peripheral organs, suggesting that localized immune-modulations by infection is paramount to pathogenesis. Throughout the course of infection a strong suppression of regulatory T-cell responses was noted and is hypothesized to be the basis of the aberrant immune activations. The unique and comprehensive monitoring of host immune responses to hantavirus infection increases our understanding of the immuno-pathogenesis of HPS and will facilitate the development of treatment strategies targeting deleterious host immunological responses.


Assuntos
Síndrome Pulmonar por Hantavirus/imunologia , Síndrome Pulmonar por Hantavirus/virologia , Administração Intranasal , Animais , Cricetinae , Modelos Animais de Doenças , Feminino , Orthohantavírus/isolamento & purificação , Orthohantavírus/patogenicidade , Síndrome Pulmonar por Hantavirus/patologia , Interações Hospedeiro-Patógeno , Pulmão/patologia , Pulmão/virologia , Mesocricetus , Linfócitos T Reguladores/imunologia
12.
PLoS Pathog ; 7(9): e1002275, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21980292

RESUMO

Transmissible spongiform encephalopathies (TSE) or prion diseases are neurodegenerative disorders associated with conversion of normal host prion protein (PrP) to a misfolded, protease-resistant form (PrPres). Genetic variations of prion protein in humans and animals can alter susceptibility to both familial and infectious prion diseases. The N171S PrP polymorphism is found mainly in humans of African descent, but its low incidence has precluded study of its possible influence on prion disease. Similar to previous experiments of others, for laboratory studies we created a transgenic model expressing the mouse PrP homolog, PrP-170S, of human PrP-171S. Since PrP polymorphisms can vary in their effects on different TSE diseases, we tested these mice with four different strains of mouse-adapted scrapie. Whereas 22L and ME7 scrapie strains induced typical clinical disease, neuropathology and accumulation of PrPres in all transgenic mice at 99-128 average days post-inoculation, strains RML and 79A produced clinical disease and PrPres formation in only a small subset of mice at very late times. When mice expressing both PrP-170S and PrP-170N were inoculated with RML scrapie, dominant-negative inhibition of disease did not occur, possibly because interaction of strain RML with PrP-170S was minimal. Surprisingly, in vitro PrP conversion using protein misfolding cyclic amplification (PMCA), did not reproduce the in vivo findings, suggesting that the resistance noted in live mice might be due to factors or conditions not present in vitro. These findings suggest that in vivo conversion of PrP-170S by RML and 79A scrapie strains was slow and inefficient. PrP-170S mice may be an example of the conformational selection model where the structure of some prion strains does not favor interactions with PrP molecules expressing certain polymorphisms.


Assuntos
Polimorfismo de Nucleotídeo Único , Príons , Dobramento de Proteína , Scrapie , Animais , Humanos , Camundongos , Camundongos Transgênicos , Príons/genética , Príons/metabolismo , Scrapie/genética , Scrapie/metabolismo , Especificidade da Espécie
13.
J Infect Dis ; 204 Suppl 3: S1090-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21987746

RESUMO

BACKGROUND: Outbreaks of filoviral hemorrhagic fever occur sporadically and unpredictably across wide regions in central Africa and overlap with the occurrence of other infectious diseases of public health importance. METHODS: As a proof of concept we developed a bivalent recombinant vaccine based on vesicular stomatitis virus (VSV) expressing the Zaire ebolavirus (ZEBOV) and Andes virus (ANDV) glycoproteins (VSVΔG/Dual) and evaluated its protective efficacy in the common lethal Syrian hamster model. Hamsters were vaccinated with VSVΔG/Dual and were lethally challenged with ZEBOV or ANDV. Time to immunity and postexposure treatment were evaluated by immunizing hamsters at different times prior to and post ZEBOV challenge. RESULTS: A single immunization with VSVΔG/Dual conferred complete and sterile protection against lethal ZEBOV and ANDV challenge. Complete protection was achieved with an immunization as close as 3 days prior to ZEBOV challenge, and 40% of the animals were even protected when treated with VSVΔG/Dual one day postchallenge. In comparison to the monovalent VSV vaccine, the bivalent vaccine has slightly reduced postexposure efficacy most likely due to its restricted lymphoid organ replication. CONCLUSIONS: Bivalent VSV vectors are a feasible approach to vaccination against multiple pathogens.


Assuntos
Vacinas contra Ebola/normas , Doença pelo Vírus Ebola/prevenção & controle , Vesiculovirus/genética , Animais , Chlorocebus aethiops , Cricetinae , Modelos Animais de Doenças , Células HEK293 , Humanos , Mesocricetus , Profilaxia Pós-Exposição , Vacinas Sintéticas/normas , Células Vero , Replicação Viral
14.
J Infect Dis ; 204 Suppl 3: S991-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21987781

RESUMO

To gain further insight into the interdependent pathogenic processes in Ebola hemorrhagic fever (EHF), we have examined the dynamics of host responses in individual rhesus macaques infected with Zaire ebolavirus over the entire disease course. Examination of coagulation parameters revealed that decreased coagulation inhibitor activity triggered severe coagulopathy as indicated by prolonged coagulation times and decreased fibrinogen levels. This has been proposed as one of the significant mechanisms underlying disseminated intravascular coagulation in EHF patients. Furthermore, monitoring of expression levels for cytokines/chemokines suggested a mixed anti-inflammatory response syndrome (MARS), which indicates that a catastrophic uncontrolled immunological status contributes to the development of fatal hemorrhagic fever. These results highlight the pathological analogies between EHF and severe sepsis and not only contribute to our understanding of the pathogenic process, but will also help to establish novel postexposure treatment modalities.


Assuntos
Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/imunologia , Animais , Coagulação Sanguínea , Quimiocinas/metabolismo , Chlorocebus aethiops , Citocinas/metabolismo , Hemorragia , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/patologia , Interações Hospedeiro-Patógeno , Inflamação , Macaca mulatta , Masculino , Fatores de Tempo , Células Vero , Viremia , Tempo de Coagulação do Sangue Total
15.
PLoS One ; 6(4): e18558, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21533129

RESUMO

The severe acute respiratory syndrome (SARS) epidemic was characterized by increased pathogenicity in the elderly due to an early exacerbated innate host response. SARS-CoV is a zoonotic pathogen that entered the human population through an intermediate host like the palm civet. To prevent future introductions of zoonotic SARS-CoV strains and subsequent transmission into the human population, heterologous disease models are needed to test the efficacy of vaccines and therapeutics against both late human and zoonotic isolates. Here we show that both human and zoonotic SARS-CoV strains can infect cynomolgus macaques and resulted in radiological as well as histopathological changes similar to those seen in mild human cases. Viral replication was higher in animals infected with a late human phase isolate compared to a zoonotic isolate. While there were significant differences in the number of host genes differentially regulated during the host responses between the three SARS-CoV strains, the top pathways and functions were similar and only apparent early during infection with the majority of genes associated with interferon signaling pathways. This study characterizes critical disease models in the evaluation and licensure of therapeutic strategies against SARS-CoV for human use.


Assuntos
Síndrome Respiratória Aguda Grave/patologia , Zoonoses , Animais , Citocinas/biossíntese , Humanos , Imuno-Histoquímica , Macaca fascicularis , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Síndrome Respiratória Aguda Grave/transmissão , Replicação Viral
16.
J Virol ; 85(3): 1214-23, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084481

RESUMO

The first influenza pandemic of the new millennium was caused by a newly emerged swine-origin influenza virus (SOIV) (H1N1). This new virus is characterized by a previously unknown constellation of gene segments derived from North American and Eurasian swine lineages and the absence of common markers predictive of human adaptation. Overall, human infections appeared to be mild, but an alarming number of young individuals presented with symptoms atypical for seasonal influenza. The new SOIV also showed a sustained human-to-human transmissibility and higher reproduction ratio than common seasonal viruses, altogether indicating a higher pathogenic potential for this newly emerged virus. To study the virulence of the SOIV, we used a recently established cynomolgus macaque model and compared parameters of clinical disease, virology, host responses, and pathology/histopathology with a current seasonal H1N1 virus. We here show that infection of macaques with two genetically similar but clinically distinct SOIV isolates from the early stage of the pandemic (A/Mexico/4108/2009 and A/Mexico/InDRE4487/2009) resulted in upper and lower respiratory tract infections and clinical disease ranging from mild to severe pneumonia that was clearly advanced over the mild infection caused by A/Kawasaki/UTK-4/2009, a current seasonal strain. Unexpectedly, we observed heterogeneity among the two SOIV isolates in virus replication, host transcriptional and cytokine responses, and disease progression, demonstrating a higher pathogenic potential for A/Mexico/InDRE4487/2009. Differences in virulence may explain more severe disease, as was seen with certain individuals infected with the emerged pandemic influenza virus. Thus, the nonhuman primate model closely mimics influenza in humans.


Assuntos
Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vírus da Influenza A Subtipo H1N1/patogenicidade , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Doenças dos Primatas/patologia , Doenças dos Primatas/virologia , Animais , Pré-Escolar , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Perfilação da Expressão Gênica , Variação Genética , Humanos , Influenza Humana/virologia , Macaca , Masculino , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Índice de Gravidade de Doença , Virulência
17.
PLoS Pathog ; 6(3): e1000800, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20221436

RESUMO

Prion diseases are fatal neurodegenerative diseases of humans and animals characterized by gray matter spongiosis and accumulation of aggregated, misfolded, protease-resistant prion protein (PrPres). PrPres can be deposited in brain in an amyloid-form and/or non-amyloid form, and is derived from host-encoded protease-sensitive PrP (PrPsen), a protein normally anchored to the plasma membrane by glycosylphosphatidylinositol (GPI). Previously, using heterozygous transgenic mice expressing only anchorless PrP, we found that PrP anchoring to the cell membrane was required for typical clinical scrapie. However, in the present experiments, using homozygous transgenic mice expressing two-fold more anchorless PrP, scrapie infection induced a new fatal disease with unique clinical signs and altered neuropathology, compared to non-transgenic mice expressing only anchored PrP. Brain tissue of transgenic mice had high amounts of infectivity, and histopathology showed dense amyloid PrPres plaque deposits without gray matter spongiosis. In contrast, infected non-transgenic mice had diffuse non-amyloid PrPres deposits with significant gray matter spongiosis. Brain graft studies suggested that anchored PrPsen expression was required for gray matter spongiosis during prion infection. Furthermore, electron and light microscopic studies in infected transgenic mice demonstrated several pathogenic processes not seen in typical prion disease, including cerebral amyloid angiopathy and ultrastructural alterations in perivascular neuropil. These findings were similar to certain human familial prion diseases as well as to non-prion human neurodegenerative diseases, such as Alzheimer's disease.


Assuntos
Amiloidose/patologia , Doenças Priônicas/patologia , Príons/genética , Príons/metabolismo , Scrapie/patologia , Animais , Membrana Basal/patologia , Membrana Basal/ultraestrutura , Transplante de Tecido Encefálico , Membrana Celular/patologia , Membrana Celular/ultraestrutura , Cerebelo/patologia , Córtex Cerebral/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Imunoeletrônica , Neuritos/patologia , Neuritos/ultraestrutura , Neuroglia/patologia , Neuroglia/ultraestrutura , Neurônios/patologia , Neurônios/ultraestrutura , Doenças Priônicas/transmissão , Príons/química , Dobramento de Proteína , Scrapie/transmissão
18.
J Virol ; 83(24): 13037-41, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19812147

RESUMO

Murine norovirus (MNV) is a highly infectious but generally nonpathogenic agent that is commonly found in research mouse colonies in both North America and Europe. In the present study, the effects of acute and chronic infections with MNV on immune responses and recovery from concurrent Friend virus (FV) infections were investigated. No significant differences in T-cell or NK-cell responses, FV-neutralizing antibody responses, or long-term recovery from FV infection were observed. We conclude that concurrent MNV infections had no major impacts on FV infections.


Assuntos
Infecções por Caliciviridae/imunologia , Leucemia Experimental/imunologia , Norovirus , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Doença Aguda , Animais , Anticorpos Antivirais/sangue , Doença Crônica , Vírus da Leucemia Murina de Friend , Camundongos
19.
J Virol ; 82(17): 8906-10, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18579597

RESUMO

Changes in the envelope proteins of retroviruses can alter the ability of these viruses to infect the central nervous system (CNS) and induce neurological disease. In the present study, nine envelope residues were found to influence neurovirulence of the Friend murine polytropic retrovirus Fr98. When projected on a three-dimensional model, these residues were clustered in two spatially separated groups, one in variable region B of the receptor binding site and the other on the opposite side of the envelope. Further studies indicated a role for these residues in virus replication in the CNS, although the residues did not affect viral entry.


Assuntos
Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/patologia , Infecções por Retroviridae/patologia , Infecções Tumorais por Vírus/patologia , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Vírus da Leucemia Murina/isolamento & purificação , Camundongos , Camundongos Endogâmicos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Receptores Virais/metabolismo , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/isolamento & purificação , Virulência/genética
20.
J Neuroimmunol ; 196(1-2): 16-26, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18396336

RESUMO

Prion diseases are neurodegenerative infections with gliosis and vacuolation. The mechanisms of degeneration remain unclear, but chemokines may be important. In current experiments CCR1 knock-out (KO) mice succumbed more rapidly to scrapie infection than WT controls. Infected KO mice had upregulation of CCL3, a CCR1 ligand, and CCR5, a receptor with specificity for CCL3. Both infected KO and WT mice had upregulation of CCR5-mediated signaling involving activation of Erk1/2 in astrocytes; however, activation was earlier in KO mice suggesting a role in pathogenesis. In both mouse strains activation of the Erk1/2 pathway may lead to astrocyte dysfunction resulting in neurodegeneration.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas PrPSc/imunologia , Doenças Priônicas/enzimologia , Doenças Priônicas/genética , Receptores CCR1/deficiência , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Ativação Enzimática/fisiologia , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos , Proteínas PrPSc/metabolismo , Doenças Priônicas/induzido quimicamente , Doenças Priônicas/patologia , Receptores CCR5/genética , Receptores CCR5/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA