Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 529
Filtrar
1.
STAR Protoc ; 4(1): 102033, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36853733

RESUMO

Characterization of double-stranded (ds)RNAs is relevant to the understanding of viral replication and immune sensing. Here, we provide a protocol describing the use of anti-dsRNA antibodies for immunofluorescence and immunoblotting in virus-infected insect cells, which can also be applied to tissues and other organisms. We describe the procedures to prepare insect cells for viral infection, followed by RNA extraction and in vitro production of synthetic dsRNA controls. We then detail the steps for dsRNA detection by immunoblotting and immunofluorescence. For complete details on the use and execution of this protocol, please refer to de Faria et al. (2022).1.


Assuntos
Vírus de Insetos , Insetos , RNA de Cadeia Dupla , Insetos/citologia , Insetos/virologia , Vírus de Insetos/genética , Imunofluorescência , Immunoblotting
2.
Viruses ; 14(2)2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35215821

RESUMO

Insect pollinators provide major pollination services for wild plants and crops. Honeybee viruses can cause serious damage to honeybee colonies. However, viruses of other wild pollinating insects have yet to be fully explored. In the present study, we used RNA sequencing to investigate the viral diversity of 50 species of wild pollinating insects. A total of 3 pathogenic honeybee viruses, 8 previously reported viruses, and 26 novel viruses were identified in sequenced samples. Among these, 7 novel viruses were shown to be closely related to honeybee pathogenic viruses, and 4 were determined to have potential pathogenicity for their hosts. The viruses detected in wild insect pollinators were mainly from the order Picornavirales and the families Orthomyxoviridae, Sinhaliviridae, Rhabdoviridae, and Flaviviridae. Our study expanded the species range of known insect pollinator viruses, contributing to future efforts to protect economic honeybees and wild pollinating insects.


Assuntos
Vírus de Insetos/isolamento & purificação , Insetos/virologia , Viroma , Animais , Abelhas/fisiologia , Abelhas/virologia , Pequim , Biodiversidade , China , Vírus de Insetos/classificação , Vírus de Insetos/genética , Insetos/fisiologia , Filogenia , Polinização
3.
J Gen Virol ; 102(12)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34919512

RESUMO

The family Geminiviridae includes viruses with mono- or bipartite single-stranded, circular DNA genomes of 2.5-5.2 kb. They cause economically important diseases in most tropical and subtropical regions of the world. Geminiviruses infect dicot and monocot plants and are transmitted by insect vectors. DNA satellites are associated with some geminiviruses. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Geminiviridae which is available at ictv.global/report/geminiviridae.


Assuntos
Geminiviridae/classificação , Doenças das Plantas/virologia , Animais , DNA de Cadeia Simples/química , DNA de Cadeia Simples/genética , DNA Viral/química , DNA Viral/genética , Geminiviridae/genética , Geminiviridae/fisiologia , Geminiviridae/ultraestrutura , Ordem dos Genes , Insetos/virologia , Vírion/química , Vírion/genética , Vírion/ultraestrutura , Replicação Viral
4.
Viruses ; 13(11)2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34834988

RESUMO

RNA interference (RNAi)-mediated antiviral immunity is believed to be the primary defense against viral infection in mosquitoes. The production of virus-specific small RNA has been demonstrated in mosquitoes and mosquito-derived cell lines for viruses in all of the major arbovirus families. However, many if not all mosquitoes are infected with a group of viruses known as insect-specific viruses (ISVs), and little is known about the mosquito immune response to this group of viruses. Therefore, in this study, we sequenced small RNA from an Aedes albopictus-derived cell line infected with either Lammi virus (LamV) or Hanko virus (HakV). These viruses belong to two distinct phylogenetic groups of insect-specific flaviviruses (ISFVs). The results revealed that both viruses elicited a strong virus-derived small interfering RNA (vsiRNA) response that increased over time and that targeted the whole viral genome, with a few predominant hotspots observed. Furthermore, only the LamV-infected cells produced virus-derived Piwi-like RNAs (vpiRNAs); however, they were mainly derived from the antisense genome and did not show the typical ping-pong signatures. HakV, which is more distantly related to the dual-host flaviviruses than LamV, may lack certain unknown sequence elements or structures required for vpiRNA production. Our findings increase the understanding of mosquito innate immunity and ISFVs' effects on their host.


Assuntos
Aedes/virologia , Flaviviridae/genética , Flavivirus/genética , Vírus de Insetos/genética , Insetos/virologia , Animais , Linhagem Celular , Flaviviridae/classificação , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Vírus de Insetos/classificação , Mosquitos Vetores/virologia , Filogenia , RNA de Cadeia Dupla , RNA Interferente Pequeno/genética , RNA Viral/genética , Análise de Sequência
5.
Viruses ; 13(11)2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34835110

RESUMO

ss(+)RNA viruses represent the dominant group of plant viruses. They owe their evolutionary superiority to the large number of mutations that occur during replication, courtesy of RNA-dependent RNA polymerase. Natural selection rewards successful viral subtypes, whose effective tuning of the ecosystem regulates the interactions between its participants. Thus, ss(+)RNA viruses act as shuttles for the functionally important genes of the participants in symbiotic relationships within the ecosystem, of which the most common ecological triad is "plant-virus-insect". Due to their short life cycle and large number of offspring, RNA viruses act as skillful tuners of the ecosystem, which benefits both viruses and the system as a whole. A fundamental understanding of this aspect of the role played by viruses in the ecosystem makes it possible to apply this knowledge to the creation of DNA insecticides. In fact, since the genes that viruses are involved in transferring are functionally important for both insects and plants, silencing these genes (for example, in insects) can be used to regulate the pest population. RNA viruses are increasingly treated not as micropathogens but as necessary regulators of ecosystem balance.


Assuntos
Vírus de Insetos , Insetos/virologia , Doenças das Plantas/virologia , Vírus de Plantas , Plantas/virologia , Vírus de RNA , Animais , Evolução Biológica , Genoma Viral , Interações Hospedeiro-Patógeno , Vírus de Insetos/genética , Vírus de Insetos/fisiologia , Vírus de Plantas/genética , Vírus de Plantas/fisiologia , Vírus de RNA/genética , Vírus de RNA/fisiologia , Simbiose
6.
Viruses ; 13(8)2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34452522

RESUMO

Viruses are excellent manipulators of host cellular machinery, behavior, and life cycle, with the host cell cytoskeleton being a primordial viral target. Viruses infecting insects generally enter host cells through clathrin-mediated endocytosis or membrane fusion mechanisms followed by transport of the viral particles to the corresponding replication sites. After viral replication, the viral progeny egresses toward adjacent cells and reaches the different target tissues. Throughout all these steps, actin and tubulin re-arrangements are driven by viruses. The mechanisms used by viruses to manipulate the insect host cytoskeleton are well documented in the case of alphabaculoviruses infecting Lepidoptera hosts and plant viruses infecting Hemiptera vectors, but they are not well studied in case of other insect-virus systems such as arboviruses-mosquito vectors. Here, we summarize the available knowledge on how viruses manipulate the insect host cell cytoskeleton, and we emphasize the primordial role of cytoskeleton components in insect virus motility and the need to expand the study of this interaction.


Assuntos
Vírus de Insetos/fisiologia , Insetos/virologia , Animais , Citoesqueleto/virologia , Interações Hospedeiro-Patógeno , Vírus de Insetos/genética , Insetos/fisiologia
7.
Mol Biotechnol ; 63(11): 1068-1080, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34228257

RESUMO

Rabies is an ancient zoonotic disease that still causes the death of over 59,000 people worldwide each year. The rabies lyssavirus encodes five proteins, including the envelope glycoprotein and the matrix protein. RVGP is the only protein exposed on the surface of viral particle, and it can induce immune response with neutralizing antibody formation. RVM has the ability to assist with production process of virus-like particles. VLPs were produced in recombinant baculovirus system. In this work, two recombinant baculoviruses carrying the RVGP and RVM genes were constructed. From the infection and coinfection assays, we standardized the best multiplicity of infection and the best harvest time. Cell supernatants were collected, concentrated, and purified by sucrose gradient. Each step was used for protein detection through immunoassays. Sucrose gradient analysis enabled to verify the separation of VLPs from rBV. Through the negative contrast technique, we visualized structures resembling rabies VLPs produced in insect cells and rBV in the different fractions of the sucrose gradient. Using ELISA to measure total RVGP, the recovery efficiency of VLPs at each stage of the purification process was verified. Thus, these results encourage further studies to confirm whether rabies VLPs are a promising candidate for a veterinary rabies vaccine.


Assuntos
Baculoviridae/genética , Insetos/metabolismo , Vacina Antirrábica/biossíntese , Vírus da Raiva/metabolismo , Raiva/virologia , Vacinas de Partículas Semelhantes a Vírus/biossíntese , Animais , Baculoviridae/isolamento & purificação , Baculoviridae/metabolismo , Células Cultivadas , Humanos , Insetos/imunologia , Insetos/virologia , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vacina Antirrábica/isolamento & purificação , Vírus da Raiva/imunologia , Vírus da Raiva/isolamento & purificação , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/isolamento & purificação
8.
J Invertebr Pathol ; 184: 107644, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34237297

RESUMO

The complex nature of climate change-mediated multitrophic interaction is an underexplored area, but has the potential to dramatically shift transmission and distribution of many insects and their pathogens, placing some populations closer to the brink of extinction. However, for individual insect-pathogen interactions climate change will have complicated hard-to-anticipate impacts. Thus, both pathogen virulence and insect host immunity are intrinsically linked with generalized stress responses, and in both pathogen and host have extensive trade-offs with nutrition (e.g., host plant quality), growth and reproduction. Potentially alleviating or exasperating these impacts, some pathogens and hosts respond genetically and rapidly to environmental shifts. This review identifies many areas for future research including a particular need to identify how altered global warming interacts with other environmental changes and stressors, and how consistent these impacts are across pathogens and hosts. With that achieved we would be closer to producing an overarching framework to integrate knowledge on all environmental interplay and infectious disease events.


Assuntos
Mudança Climática , Interações Hospedeiro-Patógeno , Insetos , Animais , Interações Hospedeiro-Parasita , Insetos/microbiologia , Insetos/parasitologia , Insetos/fisiologia , Insetos/virologia
9.
Dev Comp Immunol ; 122: 104116, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33991532

RESUMO

Protection against viral infection in hosts concerns diverse cellular and molecular mechanisms, among which RNA interference (RNAi) response is a vital one. Small interfering RNAs (siRNAs), microRNAs (miRNAs) and PIWI interacting RNAs (piRNAs) are primary categories of small RNAs involved in RNAi response, playing significant roles in restraining viral invasion. However, during a long-term coevolution, viruses have gained the ability to evade, avoid, or suppress antiviral immunity to ensure efficient replication and transmission. Baculoviruses are enveloped, insect-pathogenic viruses with double-stranded circular DNA genomes, which encode suppressors of siRNA pathway and miRNAs targeting immune-related genes to mask the antiviral activity of their hosts. This review summarized recent findings for the RNAi-based antiviral immunity in insects as well as the strategies that baculoviruses exploit to break the shield of host siRNA pathway, and hijack cellular miRNAs or encode their own miRNAs that regulate both viral and cellular gene expression to create a favorable environment for viral infection.


Assuntos
Baculoviridae/imunologia , Insetos/imunologia , Insetos/virologia , MicroRNAs/genética , RNA Interferente Pequeno/genética , Animais , Interações entre Hospedeiro e Microrganismos/imunologia , Interferência de RNA , Viroses/imunologia , Viroses/prevenção & controle
10.
Curr Opin Insect Sci ; 44: 64-71, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33866043

RESUMO

Insect parasitoids have evolved symbiotic interactions with several viruses and thousands of parasitoid species have established mutualistic associations with polydnaviruses (PDVs). While PDVs have often been described as virulence factors allowing development of immature parasitoids inside their herbivore hosts, there is increasing awareness that PDVs can affect plant-insect interactions. We review recent literature showing that PDVs alter not only host physiology, but also feeding patterns and composition of herbivore's oral secretions. In turn PDV-induced changes in herbivore phenotype affect plant responses to herbivory with consequences ranging from differential expression of plant defense-related genes to wider ecological effects across multiple trophic levels. In this opinion paper we also highlight important missing gaps to fully understand the role of PDVs and other parasitoid-associated viral symbionts in a plant-insect interaction perspective. Because PDVs negatively impact performance and survival of herbivore pests, we conclude arguing that PDV genomes offer potential opportunities for biological control.


Assuntos
Herbivoria , Interações Hospedeiro-Parasita , Insetos/virologia , Controle Biológico de Vetores , Polydnaviridae/fisiologia , Animais , Insetos/parasitologia , Plantas , Simbiose , Vespas/virologia
11.
Viruses ; 13(5)2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33925296

RESUMO

Flaviviruses implement a broad range of antagonism strategies against the host antiviral response. A pivotal component of the early host response is production and signaling of type I interferon (IFN-I). Ubiquitin, a prevalent cellular protein-modifying molecule, is heavily involved in the cellular regulation of this and other immune response pathways. Viruses use ubiquitin and ubiquitin machinery to antagonize various steps of these pathways through diverse mechanisms. Here, we highlight ways in which flaviviruses use or inhibit ubiquitin to antagonize the antiviral IFN-I response.


Assuntos
Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Flavivirus/fisiologia , Interações Hospedeiro-Patógeno , Interferon Tipo I/metabolismo , Ubiquitina/metabolismo , Animais , Regulação Viral da Expressão Gênica , Humanos , Insetos/metabolismo , Insetos/virologia , Janus Quinases/metabolismo , Ligação Proteica , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Ubiquitinação
12.
Virology ; 559: 30-39, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33812340

RESUMO

Long Pine Key virus (LPKV) and Lammi virus are insect-specific flaviviruses that phylogenetically affiliate with dual-host flaviviruses. The goal of this study was to provide insight into the genetic determinants that condition this host range restriction. Chimeras were initially created by replacing select regions of the Zika virus genome, including the premembrane and envelope protein (prM-E) genes, with the corresponding regions of the LPKV genome. Of the four chimeras produced, one (the prM-E swap) yielded virus that replicated in mosquito cells. Another chimeric virus with a mosquito replication-competent phenotype was created by inserting the prM-E genes of Lammi virus into a Zika virus genetic background. Vertebrate cells did not support the replication of either chimeric virus although trace to modest amounts of viral antigen were produced, consistent with suboptimal viral entry. These data suggest that dual-host affiliated insect-specific flaviviruses cannot replicate in vertebrate cells due to entry and post-translational restrictions.


Assuntos
Insetos/virologia , Processamento de Proteína Pós-Traducional , Proteínas Estruturais Virais/genética , Replicação Viral/genética , Zika virus/genética , Animais , Flavivirus/classificação , Flavivirus/genética , Flavivirus/fisiologia , Proteômica , Zika virus/fisiologia , Infecção por Zika virus
13.
Front Immunol ; 12: 613729, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33708207

RESUMO

Viral infection triggers insect immune response, including RNA interference, apoptosis and autophagy, and profoundly changes the gene expression profiles in infected cells. Although intracellular degradation is crucial for restricting viral infection, intercellular communication is required to mount a robust systemic immune response. This review focuses on recent advances in understanding the intercellular communications in insect antiviral immunity, including protein-based and virus-derived RNA based cell-cell communications, with emphasis on the signaling pathway that induces the production of the potential cytokines. The prospects and challenges of future work are also discussed.


Assuntos
Comunicação Celular , Resistência à Doença/imunologia , Interações Hospedeiro-Patógeno/imunologia , Insetos/imunologia , Insetos/virologia , Doenças dos Animais/genética , Doenças dos Animais/imunologia , Doenças dos Animais/metabolismo , Doenças dos Animais/virologia , Animais , Biomarcadores , Citocinas/metabolismo , Resistência à Doença/genética , Interações Hospedeiro-Patógeno/genética , Imunidade Inata , Insetos/metabolismo
14.
Commun Biol ; 4(1): 345, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33753850

RESUMO

Bunyaviruses have a genome that is divided over multiple segments. Genome segmentation complicates the generation of progeny virus, since each newly formed virus particle should preferably contain a full set of genome segments in order to disseminate efficiently within and between hosts. Here, we combine immunofluorescence and fluorescence in situ hybridization techniques to simultaneously visualize bunyavirus progeny virions and their genomic content at single-molecule resolution in the context of singly infected cells. Using Rift Valley fever virus and Schmallenberg virus as prototype tri-segmented bunyaviruses, we show that bunyavirus genome packaging is influenced by the intracellular viral genome content of individual cells, which results in greatly variable packaging efficiencies within a cell population. We further show that bunyavirus genome packaging is more efficient in insect cells compared to mammalian cells and provide new insights on the possibility that incomplete particles may contribute to bunyavirus spread as well.


Assuntos
Insetos/virologia , Orthobunyavirus/genética , Ribonucleoproteínas/genética , Empacotamento do Genoma Viral , Proteínas Virais/genética , Vírion/metabolismo , Animais , Chlorocebus aethiops , Imunofluorescência , Hibridização in Situ Fluorescente , Orthobunyavirus/metabolismo , Orthobunyavirus/patogenicidade , Ribonucleoproteínas/metabolismo , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/metabolismo , Vírus da Febre do Vale do Rift/patogenicidade , Células Vero , Proteínas Virais/metabolismo , Vírion/genética
15.
Microbiome ; 9(1): 18, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33478588

RESUMO

BACKGROUND: As the largest group of mammalian species, which are also widely distributed all over the world, rodents are the natural reservoirs for many diverse zoonotic viruses. A comprehensive understanding of the core virome of diverse rodents should therefore assist in efforts to reduce the risk of future emergence or re-emergence of rodent-borne zoonotic pathogens. RESULTS: This study aimed to describe the viral range that could be detected in the lungs of rodents from Mainland Southeast Asia. Lung samples were collected from 3284 rodents and insectivores of the orders Rodentia, Scandentia, and Eulipotyphla in eighteen provinces of Thailand, Lao PDR, and Cambodia throughout 2006-2018. Meta-transcriptomic analysis was used to outline the unique spectral characteristics of the mammalian viruses within these lungs and the ecological and genetic imprints of the novel viruses. Many mammalian- or arthropod-related viruses from distinct evolutionary lineages were reported for the first time in these species, and viruses related to known pathogens were characterized for their genomic and evolutionary characteristics, host species, and locations. CONCLUSIONS: These results expand our understanding of the core viromes of rodents and insectivores from Mainland Southeast Asia and suggest that a high diversity of viruses remains to be found in rodent species of this area. These findings, combined with our previous virome data from China, increase our knowledge of the viral community in wildlife and arthropod vectors in emerging disease hotspots of East and Southeast Asia. Video abstract.


Assuntos
Pulmão/virologia , RNA Viral/análise , Roedores/virologia , Viroma/genética , Animais , Sudeste Asiático , Insetos/virologia
16.
Annu Rev Entomol ; 66: 61-79, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33417818

RESUMO

As an overarching immune mechanism, RNA interference (RNAi) displays pathogen specificity and memory via different pathways. The small interfering RNA (siRNA) pathway is the primary antiviral defense mechanism against RNA viruses of insects and plays a lesser role in defense against DNA viruses. Reflecting the pivotal role of the siRNA pathway in virus selection, different virus families have independently evolved unique strategies to counter this host response, including protein-mediated, decoy RNA-based, and microRNA-based strategies. In this review, we outline the interplay between insect viruses and the different pathways of the RNAi antiviral response; describe practical application of these interactions for improved expression systems and for pest and disease management; and highlight research avenues for advancement of the field.


Assuntos
Interações Hospedeiro-Patógeno , Vírus de Insetos/fisiologia , Insetos/virologia , Interferência de RNA , Animais , Insetos/genética , Insetos/imunologia
18.
Viruses ; 14(1)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-35062206

RESUMO

Insects are crucial for ecosystem functions and services and directly influence human well-being and health [...].


Assuntos
Evolução Molecular , Vírus de Insetos/genética , Vírus de Insetos/fisiologia , Insetos/virologia , Animais , Vírus de Insetos/isolamento & purificação
19.
Virology ; 552: 73-82, 2021 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-33075709

RESUMO

Zika virus (ZIKV) is a mosquito-borne flavivirus that replicates in both vertebrate and insect cells, whereas insect-specific flaviviruses (ISF) replicate only in insect cells. We sought to convert ZIKV, from a dual-tropic flavivirus, into an insect-specific virus for the eventual development of a safe ZIKV vaccine. Reverse genetics was used to introduce specific mutations into the furin cleavage motif within the ZIKV pre-membrane protein (prM). Mutant clones were selected, which replicated well in C6/36 insect cells but exhibited reduced replication in non-human primate (Vero) cells. Further characterization of the furin cleavage site mutants indicated they replicated poorly in both human (HeLa, U251), and baby hamster kidney (BHK-21) cells. One clone with the induced mutation in the prM protein and at positions 291and 452 within the NS3 protein was totally and stably replication-defective in vertebrate cells (VSRD-ZIKV). Preliminary studies in ZIKV sensitive, immunodeficient mice demonstrated that VSRD-ZIKV-infected mice survived and were virus-negative. Our study indicates that a reverse genetic approach targeting the furin cleavage site in prM can be used to select an insect-specific ZIKV with the potential utility as a vaccine strain.


Assuntos
Insetos/virologia , Proteínas de Membrana/metabolismo , Vertebrados/virologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Infecção por Zika virus/virologia , Zika virus/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Furina/metabolismo , Células HeLa , Especificidade de Hospedeiro , Humanos , Isoquinolinas , Camundongos , Mutação , Genética Reversa/métodos , Células Vero , Vertebrados/imunologia , Proteínas Virais/metabolismo , Infecção por Zika virus/imunologia
20.
Virus Res ; 292: 198228, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33188797

RESUMO

Given the dual life cycle of arboviruses in insect and animal hosts and the importance of serum factors as a first line antiviral defense, we have examined the outcome of interactions between the arbovirus La Crosse Virus (LACV) and human serum. To mimic the life cycle between species, we used LACV derived from insect (I-LACV) and human keratinocyte (HaCaT) cells. Incubation of I-LACV with normal human serum did not result in neutralization, but instead stabilized I-LACV virions and enhanced the amount of infectious virus. Enhanced infectivity was also seen with heat-inactivated serum devoid of complement activity and with serum from a range of animals including mouse, ferret, and non-human primates. Depletion of antibodies from serum resulted in loss of enhancement of infectivity and sucrose gradient sedimentation assays showed IgG co-sedimenting with I-LACV particles. In agreement with our results with I-LACV, HaCaT-derived LACV was not neutralized by complement or antibodies in normal human serum. However, in contrast to I-LACV, HaCaT-derived LACV infectivity was stable when incubated alone and treatment with serum did not enhance infectivity. Our results indicate that LACV derived from insect cells differs substantially from virus derived from human cells, with I-LACV being dependent on serum factors to enhance infectivity. These findings suggest that understanding differential composition of insect versus animal cell-derived LACV may form the foundation for potential new antiviral approaches.


Assuntos
Encefalite da Califórnia/virologia , Insetos/virologia , Queratinócitos/virologia , Vírus La Crosse/fisiologia , Soro/imunologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Encefalite da Califórnia/imunologia , Furões , Interações Hospedeiro-Patógeno , Humanos , Queratinócitos/imunologia , Vírus La Crosse/genética , Vírus La Crosse/imunologia , Camundongos , Testes de Neutralização , Primatas , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...