Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Neurol ; 13: 1051559, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36452163

RESUMO

Understanding the kinetics and durability of AAV-mediated transgene expression in the brain is essential for conducting basic neuroscience studies as well as for developing gene therapy approaches for CNS diseases. Here, we characterize and compare the temporal profile of transgene expression after bilateral injections into the mouse striatum of rAAV9 encoding GFP under the control of either a ubiquitous promoter (CAG), or the neuron-specific human synapsin (hSyn) and CamKII promoters. GFP protein expression with the CAG promoter was highest at 3 weeks, and then decreased to stable levels at 3 and 6 months. Surprisingly, GFP mRNA levels continued to increase from 3 weeks to 3 months, despite GFP protein expression decreasing during this time. GFP protein expression with hSyn increased more slowly, reaching a maximum at 3 months, which was equivalent to protein expression levels from CAG at that time point. Importantly, transgene expression driven by the hSyn promoter at 6 months was not silenced as previously reported, and GFP mRNA was continuing to rise even at the final 6-month time point. Thus, hSyn as a promoter for transgene expression demonstrates long-term durability but may require more time after vector administration to achieve steady-state levels. Because CAG had the highest GFP protein expression in our comparison, which was at 3 weeks post administration, the early kinetics of transgene expression from CAG was examined (1, 2, 5, and 10 days after injection). This analysis showed that GFP protein expression and GFP mRNA increased during the first 3 weeks after administration. Interestingly, vector DNA rapidly decreased 10-fold over the first 3 weeks following injection as it assembled into stable circular episomes and concatemers. Surprisingly, the processing of vector genomes into circular episomes and concatemers was continually dynamic up to 3 months after injection. These results provide novel insight into the dynamic processing of vector genomes and promoter-specific temporal patterns of transgene expression in the brain.

2.
Viruses ; 14(7)2022 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-35891445

RESUMO

La Crosse virus (LACV) is a major cause of pediatric encephalitis and aseptic meningitis in the Midwestern, Mid-Atlantic, and Southern United States, where it is an emerging pathogen. The LACV Gc glycoprotein plays a critical role in the neuropathogenesis of LACV encephalitis as the putative virus attachment protein. Previously, we identified and experimentally confirmed the location of the LACV fusion peptide within Gc and generated a panel of recombinant LACVs (rLACVs) containing mutations in the fusion peptide as well as the wild-type sequence. These rLACVs retained their ability to cause neuronal death in a primary embryonic rat neuronal culture system, despite decreased replication and fusion phenotypes. To test the role of the fusion peptide in vivo, we tested rLACVs in an age-dependent murine model of LACV encephalitis. When inoculated directly into the CNS of young adult mice (P28), the rLACV fusion peptide mutants were as neurovirulent as the rLACV engineered with a wild-type sequence, confirming the results obtained in tissue culture. In contrast, the fusion peptide mutant rLACVs were less neuroinvasive when suckling (P3) or weanling (P21) mice were inoculated peripherally, demonstrating that the LACV fusion peptide is a determinant of neuroinvasion, but not of neurovirulence. In a challenge experiment, we found that peripheral challenge of weanling (P21) mice with fusion peptide mutant rLACVs protected from a subsequent WT-LACV challenge, suggesting that mutations in the fusion peptide are an attractive target for generating live-attenuated virus vaccines. Importantly, the high degree of conservation of the fusion peptide amongst the Bunyavirales and, structurally, other arboviruses suggests that these findings are broadly applicable to viruses that use a class II fusion mechanism and cause neurologic disease.


Assuntos
Encefalite da Califórnia , Vírus La Crosse , Animais , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Peptídeos/genética , Peptídeos/metabolismo , Ratos , Estados Unidos , Proteínas Virais/genética
3.
Virulence ; 12(1): 430-443, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33487119

RESUMO

Venezuelan equine encephalitis virus (VEEV) is an encephalitic alphavirus that can cause debilitating, acute febrile illness and potentially result in encephalitis. Currently, there are no FDA-licensed vaccines or specific therapeutics for VEEV. Previous studies have demonstrated that VEEV infection results in increased blood-brain barrier (BBB) permeability that is mediated by matrix metalloproteinases (MMPs). Furthermore, after subarachnoid hemorrhage in mice, MMP-9 is upregulated in the brain and mediates BBB permeability in a toll-like receptor 4 (TLR4)-dependent manner. Here, we demonstrate that disease in C3H mice during VEEV TC-83 infection is dependent on TLR4 because intranasal infection of C3H/HeN (TLR4 WT ) mice with VEEV TC-83 resulted in mortality as opposed to survival of TLR4-defective C3H/HeJ (TLR4 mut ) mice. In addition, BBB permeability was induced to a lesser extent in TLR4 mut mice compared with TLR4 WT mice during VEEV TC-83 infection as determined by sodium fluorescein and fluorescently-conjugated dextran extravasation. Moreover, MMP-9, MMP-2, ICAM-1, CCL2 and IFN-γ were all induced to significantly lower levels in the brains of infected TLR4 mut mice compared with infected TLR4 WT mice despite the absence of significantly different viral titers or immune cell populations in the brains of infected TLR4 WT and TLR4 mut mice. These data demonstrate the critical role of TLR4 in mediating BBB permeability and disease in C3H mice during VEEV TC-83 infection, which suggests that TLR4 is a potential target for the development of therapeutics for VEEV.


Assuntos
Barreira Hematoencefálica/metabolismo , Vírus da Encefalite Equina Venezuelana/patogenicidade , Receptor 4 Toll-Like/genética , Animais , Encéfalo/virologia , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/virologia , Feminino , Camundongos , Camundongos Endogâmicos C3H , Permeabilidade , Receptor 4 Toll-Like/metabolismo , Replicação Viral
4.
JCI Insight ; 5(19)2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32841215

RESUMO

The emergence of SARS-CoV-2 has created an international health crisis, and small animal models mirroring SARS-CoV-2 human disease are essential for medical countermeasure (MCM) development. Mice are refractory to SARS-CoV-2 infection owing to low-affinity binding to the murine angiotensin-converting enzyme 2 (ACE2) protein. Here, we evaluated the pathogenesis of SARS-CoV-2 in male and female mice expressing the human ACE2 gene under the control of the keratin 18 promoter (K18). In contrast to nontransgenic mice, intranasal exposure of K18-hACE2 animals to 2 different doses of SARS-CoV-2 resulted in acute disease, including weight loss, lung injury, brain infection, and lethality. Vasculitis was the most prominent finding in the lungs of infected mice. Transcriptomic analysis from lungs of infected animals showed increases in transcripts involved in lung injury and inflammatory cytokines. In the low-dose challenge groups, there was a survival advantage in the female mice, with 60% surviving infection, whereas all male mice succumbed to disease. Male mice that succumbed to disease had higher levels of inflammatory transcripts compared with female mice. To our knowledge, this is the first highly lethal murine infection model for SARS-CoV-2 and should be valuable for the study of SARS-CoV-2 pathogenesis and for the assessment of MCMs.


Assuntos
Causas de Morte , Infecções por Coronavirus/patologia , Progressão da Doença , Peptidil Dipeptidase A/genética , Pneumonia Viral/patologia , Síndrome Respiratória Aguda Grave/patologia , Enzima de Conversão de Angiotensina 2 , Animais , COVID-19 , Infecções por Coronavirus/fisiopatologia , Modelos Animais de Doenças , Feminino , Humanos , Pulmão/patologia , Masculino , Camundongos , Camundongos Transgênicos , Pandemias , Pneumonia Viral/fisiopatologia , Síndrome Respiratória Aguda Grave/fisiopatologia , Índice de Gravidade de Doença , Taxa de Sobrevida , Replicação Viral/genética
5.
Nat Med ; 26(2): 228-235, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32015557

RESUMO

Zika virus (ZIKV) has caused significant disease, with widespread cases of neurological pathology and congenital neurologic defects. Rapid vaccine development has led to a number of candidates capable of eliciting potent ZIKV-neutralizing antibodies (reviewed in refs. 1-3). Despite advances in vaccine development, it remains unclear how ZIKV vaccination affects immune responses in humans with prior flavivirus immunity. Here we show that a single-dose immunization of ZIKV purified inactivated vaccine (ZPIV)4-7 in a dengue virus (DENV)-experienced human elicited potent cross-neutralizing antibodies to both ZIKV and DENV. Using a unique ZIKV virion-based sorting strategy, we isolated and characterized multiple antibodies, including one termed MZ4, which targets a novel site of vulnerability centered on the Envelope (E) domain I/III linker region and protects mice from viremia and viral dissemination following ZIKV or DENV-2 challenge. These data demonstrate that Zika vaccination in a DENV-experienced individual can boost pre-existing flavivirus immunity and elicit protective responses against both ZIKV and DENV. ZPIV vaccination in Puerto Rican individuals with prior flavivirus experience yielded similar cross-neutralizing potency after a single vaccination, highlighting the potential benefit of ZIKV vaccination in flavivirus-endemic areas.


Assuntos
Dengue/imunologia , Doadores de Tecidos , Vacinas Virais/uso terapêutico , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Chlorocebus aethiops , Reações Cruzadas , Vírus da Dengue , Mapeamento de Epitopos , Feminino , Flavivirus/metabolismo , Humanos , Imunoglobulina G/química , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ligação Proteica , Domínios Proteicos , Vacinação , Vacinas de Produtos Inativados/uso terapêutico , Células Vero , Viremia , Zika virus
6.
Viruses ; 11(11)2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31766138

RESUMO

Traditional pathogenesis studies of alphaviruses involves monitoring survival, viremia, and pathogen dissemination via serial necropsies; however, molecular imaging shifts this paradigm and provides a dynamic assessment of pathogen infection. Positron emission tomography (PET) with PET tracers targeted to study neuroinflammation (N,N-diethyl-2-[4-phenyl]-5,7-dimethylpyrazolo[1,5-a]pyrimidine-3-acetamide, [18F]DPA-714), apoptosis (caspase-3 substrate, [18F]CP-18), hypoxia (fluormisonidazole, [18F]FMISO), blood-brain barrier (BBB) integrity ([18F]albumin), and metabolism (fluorodeoxyglucose, [18F]FDG) was performed on C3H/HeN mice infected intranasally with 7000 plaque-forming units (PFU) of Venezuelan equine encephalitis virus (VEEV) TC-83. The main findings are as follows: (1) whole-brain [18F]DPA-714 and [18F]CP-18 uptake increased three-fold demonstrating, neuroinflammation and apoptosis, respectively; (2) [18F]albumin uptake increased by 25% across the brain demonstrating an altered BBB; (3) [18F]FMISO uptake increased by 50% across the whole brain indicating hypoxic regions; (4) whole-brain [18F]FDG uptake was unaffected; (5) [18F]DPA-714 uptake in (a) cortex, thalamus, striatum, hypothalamus, and hippocampus increased through day seven and decreased by day 10 post exposure, (b) olfactory bulb increased at day three, peaked day seven, and decreased day 10, and (c) brain stem and cerebellum increased through day 10. In conclusion, intranasal exposure of C3H/HeN mice to VEEV TC-83 results in both time-dependent and regional increases in brain inflammation, apoptosis, and hypoxia, as well as modest decreases in BBB integrity; however, it has no effect on brain glucose metabolism.


Assuntos
Apoptose , Barreira Hematoencefálica/metabolismo , Vírus da Encefalite Equina Venezuelana , Encefalomielite Equina Venezuelana/diagnóstico , Encefalomielite Equina Venezuelana/metabolismo , Hipóxia/metabolismo , Tomografia por Emissão de Pósitrons , Animais , Biomarcadores , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/fisiologia , Encefalomielite Equina Venezuelana/virologia , Cavalos , Processamento de Imagem Assistida por Computador , Camundongos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/metabolismo
7.
Mol Imaging Biol ; 20(2): 275-283, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28900831

RESUMO

PURPOSE: The association of Zika virus (ZIKV) infection and development of neurological sequelae require a better understanding of the pathogenic mechanisms causing severe disease. The purpose of this study was to evaluate the ability and sensitivity of positron emission tomography (PET) imaging using [18F]DPA-714, a translocator protein (TSPO) 18 kDa radioligand, to detect and quantify neuroinflammation in ZIKV-infected mice. PROCEDURES: We assessed ZIKV-induced pathogenesis in wild-type C57BL/6 mice administered an antibody to inhibit type I interferon (IFN) signaling. [18F]DPA-714 PET imaging was performed on days 3, 6, and 10 post-infection (PI), and tissues were subsequently processed for histological evaluation, quantification of microgliosis, and detection of viral RNA by in situ hybridization (ISH). RESULTS: In susceptible ZIKV-infected mice, viral titers in the brain increased from days 3 to 10 PI. Over this span, these mice showed a two- to sixfold increase in global brain neuroinflammation using [18F]DPA-714 PET imaging despite limited, regional detection of viral RNA. No measurable increase in ionized calcium binding adaptor molecule 1 (Iba-1) expression was noted at day 3 PI; however, there was a modest increase at day 6 PI and an approximately significant fourfold increase in Iba-1 expression at day 10 PI in the susceptible ZIKV-infected group relative to controls. CONCLUSIONS: The results of the current study demonstrate that global neuroinflammation plays a significant role in the progression of ZIKV infection and that [18F]DPA-714 PET imaging is a sensitive tool relative to histology for the detection of neuroinflammation. [18F]DPA-714 PET imaging may be useful in dynamically characterizing the pathology associated with neurotropic viruses and the evaluation of therapeutics being developed for treatment of infectious diseases.


Assuntos
Encéfalo/patologia , Radioisótopos de Flúor/química , Inflamação/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Pirazóis/química , Pirimidinas/química , Infecção por Zika virus/diagnóstico por imagem , Infecção por Zika virus/virologia , Zika virus/fisiologia , Animais , Encéfalo/virologia , Feminino , Gliose/sangue , Gliose/patologia , Inflamação/sangue , Inflamação/patologia , Camundongos Endogâmicos C57BL , RNA Viral/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Infecção por Zika virus/sangue
8.
Am J Trop Med Hyg ; 98(2): 432-444, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29280428

RESUMO

Zika virus (ZIKV) is a mosquito-borne member of the genus Flavivirus that has emerged since 2007 to cause outbreaks in Africa, Asia, Oceania, and most recently, in the Americas. Here, we used an isolate history as well as genetic and phylogenetic analyses to characterize three low-passage isolates representing African (ArD 41525) and Asian (CPC-0740, SV0127-14) lineages to investigate the potential phenotypic differences in vitro and in vivo. The African isolate displayed a large plaque phenotype (∼3-4 mm) on Vero and HEK-293 cells, whereas the Asian isolates either exhibited a small plaque phenotype (∼1-2 mm) or did not produce any plaques. In multistep replication kinetics in nine different vertebrate and insect cell lines, the African isolate consistently displayed faster replication kinetics and yielded ∼10- to 10,000-fold higher peak virus titers (infectious or RNA copies) compared with the Asian isolates. Oral exposure of Aedes aegypti mosquitoes with the African isolate yielded higher infection and dissemination rates compared with the Asian isolates. Infection of Ifnar1-/- mice with the African isolate produced a uniformly fatal disease, whereas infection with the Asian isolates produced either a delay in time-to-death or a significantly lower mortality rate. Last, the African isolate was > 10,000-fold more virulent than the Asian isolates in an interferon type I antibody blockade mouse model. These data demonstrate substantial phenotypic differences between low-passage African and Asian isolates both in vitro and in vivo and warrant further investigation. They also highlight the need for basic characterization of ZIKV isolates, as the utilization of the uncharacterized isolates could have consequences for animal model and therapeutic/vaccine development.


Assuntos
Variação Biológica da População/genética , Zika virus/isolamento & purificação , Aedes/virologia , África , América , Animais , Ásia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos/virologia , Camundongos Endogâmicos C57BL/virologia , Mosquitos Vetores/virologia , Reação em Cadeia da Polimerase em Tempo Real/métodos , Zika virus/genética , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/genética
9.
PLoS Negl Trop Dis ; 11(1): e0005296, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28068342

RESUMO

Animal models are needed to better understand the pathogenic mechanisms of Zika virus (ZIKV) and to evaluate candidate medical countermeasures. Adult mice infected with ZIKV develop a transient viremia, but do not demonstrate signs of morbidity or mortality. Mice deficient in type I or a combination of type I and type II interferon (IFN) responses are highly susceptible to ZIKV infection; however, the absence of a competent immune system limits their usefulness for studying medical countermeasures. Here we employ a murine model for ZIKV using wild-type C57BL/6 mice treated with an antibody to disrupt type I IFN signaling to study ZIKV pathogenesis. We observed 40% mortality in antibody treated mice exposed to ZIKV subcutaneously whereas mice exposed by intraperitoneal inoculation were highly susceptible incurring 100% mortality. Mice infected by both exposure routes experienced weight loss, high viremia, and severe neuropathologic changes. The most significant histopathological findings occurred in the central nervous system where lesions represent an acute to subacute encephalitis/encephalomyelitis that is characterized by neuronal death, astrogliosis, microgliosis, scattered necrotic cellular debris, and inflammatory cell infiltrates. This model of ZIKV pathogenesis will be valuable for evaluating medical countermeasures and the pathogenic mechanisms of ZIKV because it allows immune responses to be elicited in immunologically competent mice with IFN I blockade only induced at the time of infection.


Assuntos
Sistema Nervoso Central/virologia , Interferon Tipo I/imunologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Feminino , Humanos , Interferon Tipo I/genética , Camundongos , Camundongos Endogâmicos C57BL , Infecção por Zika virus/genética , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
10.
J Virol ; 86(15): 7988-8001, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623766

RESUMO

La Crosse virus (LACV) is a leading cause of pediatric encephalitis and aseptic meningitis in the midwestern and southern United States, where it is considered an emerging human pathogen. No specific therapies or vaccines are available for LACV or any other orthobunyaviruses. Inhibition of LACV entry into cells is a potential target for therapeutic intervention, but this approach is limited by our current knowledge of the entry process. Here, we determined that clathrin-mediated endocytosis is the primary mechanism of orthobunyavirus entry and identified key cellular factors in this process. First, we demonstrated that LACV colocalized with clathrin shortly after infection in HeLa cells; we then confirmed the functional requirement of dynamin- and clathrin-mediated endocytosis for orthobunyavirus entry using several independent assays and, importantly, extended these findings to primary neuronal cultures. We also determined that macropinocytosis and caveolar endocytosis, both established routes of virus entry, are not critical for cellular entry of LACV. Moreover, we demonstrated that LACV infection is dependent on Rab5, which plays an important regulatory role in early endosomes, but not on Rab7, which is associated with late endosomes. These findings provide the first description of bunyavirus entry into cells of the central nervous system, where infection can cause severe neurological disease, and will aid in the design and development of antivirals and therapeutics that may be useful in the treatment of LACV and, more broadly, arboviral infections of the central nervous system.


Assuntos
Clatrina/metabolismo , Encefalite da Califórnia/metabolismo , Endocitose , Endossomos/metabolismo , Vírus La Crosse/metabolismo , Internalização do Vírus , Animais , Chlorocebus aethiops , Clatrina/genética , Cricetinae , Encefalite da Califórnia/tratamento farmacológico , Encefalite da Califórnia/genética , Endossomos/genética , Endossomos/virologia , Células HeLa , Humanos , Vírus La Crosse/genética , Células Vero , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
11.
Viruses ; 3(6): 629-58, 2011 06.
Artigo em Inglês | MEDLINE | ID: mdl-21994750

RESUMO

A myriad of factors favor the emergence and re-emergence of arthropod-borne viruses (arboviruses), including migration, climate change, intensified livestock production, an increasing volume of international trade and transportation, and changes to ecosystems (e.g., deforestation and loss of biodiversity). Consequently, arboviruses are distributed worldwide and represent over 30% of all emerging infectious diseases identified in the past decade. Although some arboviral infections go undetected or are associated with mild, flu-like symptoms, many are important human and veterinary pathogens causing serious illnesses such as arthritis, gastroenteritis, encephalitis and hemorrhagic fever and devastating economic loss as a consequence of lost productivity and high mortality rates among livestock. One of the most consistent molecular features of emerging arboviruses, in addition to their near exclusive use of RNA genomes, is the inclusion of viral, non-structural proteins that act as interferon antagonists. In this review, we describe these interferon antagonists and common strategies that arboviruses use to counter the host innate immune response. In addition, we discuss the complex interplay between host factors and viral determinants that are associated with virus emergence and re-emergence, and identify potential targets for vaccine and anti-viral therapies.


Assuntos
Infecções por Arbovirus/imunologia , Arbovírus/imunologia , Arbovírus/patogenicidade , Interferons/antagonistas & inibidores , Interferons/imunologia , Proteínas não Estruturais Virais/imunologia , Animais , Infecções por Arbovirus/virologia , Arbovírus/genética , Humanos , Proteínas não Estruturais Virais/genética
12.
J Neuroimmune Pharmacol ; 5(3): 428-42, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20652430

RESUMO

Arthropod-borne viruses (arboviruses) are of paramount concern as a group of pathogens at the forefront of emerging and re-emerging diseases. Although some arboviral infections are asymptomatic or present with a mild influenza-like illness, many are important human and veterinary pathogens causing serious illness ranging from rash and arthritis to encephalitis and hemorrhagic fever. Here, we discuss arboviruses from diverse families (Flaviviruses, Alphaviruses, and the Bunyaviridae) that are causative agents of encephalitis in humans. An understanding of the natural history of these infections as well as shared mechanisms of neuroinvasion and neurovirulence is critical to control the spread of these viruses and for the development of effective vaccines and treatment modalities.


Assuntos
Arbovírus/patogenicidade , Encefalite por Arbovirus/virologia , Alphavirus/patogenicidade , Infecções por Alphavirus/transmissão , Infecções por Alphavirus/virologia , Bunyaviridae/patogenicidade , Infecções por Bunyaviridae/transmissão , Infecções por Bunyaviridae/virologia , Vírus Chikungunya/patogenicidade , Doenças Transmissíveis Emergentes/transmissão , Doenças Transmissíveis Emergentes/virologia , Encefalite por Arbovirus/transmissão , Encefalite da Califórnia/transmissão , Encefalite da Califórnia/virologia , Flavivirus/patogenicidade , Infecções por Flavivirus/transmissão , Infecções por Flavivirus/virologia , Humanos , Vírus La Crosse/patogenicidade , Febre do Vale de Rift/transmissão , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/patogenicidade , Febre do Nilo Ocidental/transmissão , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/patogenicidade
13.
Virology ; 404(2): 139-47, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20553924

RESUMO

La Crosse virus is a leading cause of pediatric encephalitis in the Midwestern United States and an emerging pathogen in the American South. The LACV glycoprotein Gc plays a critical role in entry as the virus attachment protein. A 22 amino acid hydrophobic region within Gc (1066-1087) was recently identified as the LACV fusion peptide. To further define the role of Gc (1066-1087) in virus entry, fusion, and neuropathogenesis, a panel of recombinant LACV (rLACV) fusion peptide mutant viruses was generated. Replication of mutant rLACVs was significantly reduced. In addition, the fusion peptide mutants demonstrated decreased fusion phenotypes relative to LACV-WT. Interestingly, these viruses maintained their ability to cause neuronal loss in culture, suggesting that the fusion peptide of LACV Gc is a determinant of properties associated with neuroinvasion (growth to high titer in muscle cells and a robust fusion phenotype), but not necessarily of neurovirulence.


Assuntos
Vírus La Crosse/genética , Proteínas Virais de Fusão/genética , Animais , Linhagem Celular , Cricetinae , Fibroblastos/virologia , Vírus La Crosse/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Fenótipo , Proteínas Virais de Fusão/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...