Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 619(7969): 338-347, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37380775

RESUMO

Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.


Assuntos
Aves , Interações entre Hospedeiro e Microrganismos , Vírus da Influenza A , Influenza Aviária , Influenza Humana , Zoonoses Virais , Animais , Humanos , Aves/virologia , Vírus da Influenza A/classificação , Vírus da Influenza A/genética , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/transmissão , Influenza Aviária/virologia , Influenza Humana/prevenção & controle , Influenza Humana/transmissão , Influenza Humana/virologia , Primatas , Sistema Respiratório/metabolismo , Sistema Respiratório/virologia , Medição de Risco , Zoonoses Virais/prevenção & controle , Zoonoses Virais/transmissão , Zoonoses Virais/virologia , Replicação Viral
2.
mBio ; 14(3): e0010123, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37097030

RESUMO

Infected hosts possess two alternative strategies to protect themselves against the negative impact of virus infections: resistance, used to abrogate virus replication, and disease tolerance, used to avoid tissue damage without controlling viral burden. The principles governing pathogen resistance are well understood, while less is known about those involved in disease tolerance. Here, we studied bluetongue virus (BTV), the cause of bluetongue disease of ruminants, as a model system to investigate the mechanisms of virus-host interactions correlating with disease tolerance. BTV induces clinical disease mainly in sheep, while cattle are considered reservoirs of infection, rarely exhibiting clinical symptoms despite sustained viremia. Using primary cells from multiple donors, we show that BTV consistently reaches higher titers in ovine cells than cells from cattle. The variable replication kinetics of BTV in sheep and cow cells were mostly abolished by abrogating the cell type I interferon (IFN) response. We identified restriction factors blocking BTV replication, but both the sheep and cow orthologues of these antiviral genes possess anti-BTV properties. Importantly, we demonstrate that BTV induces a faster host cell protein synthesis shutoff in primary sheep cells than cow cells, which results in an earlier downregulation of antiviral proteins. Moreover, by using RNA sequencing (RNA-seq), we also show a more pronounced expression of interferon-stimulated genes (ISGs) in BTV-infected cow cells than sheep cells. Our data provide a new perspective on how the type I IFN response in reservoir species can have overall positive effects on both virus and host evolution. IMPORTANCE The host immune response usually aims to inhibit virus replication in order to avoid cell damage and disease. In some cases, however, the infected host avoids the deleterious effects of infection despite high levels of viral replication. This strategy is known as disease tolerance, and it is used by animal reservoirs of some zoonotic viruses. Here, using a virus of ruminants (bluetongue virus [BTV]) as an experimental system, we dissected virus-host interactions in cells collected from species that are susceptible (sheep) or tolerant (cow) to disease. We show that (i) virus modulation of the host antiviral type I interferon (IFN) responses, (ii) viral replication kinetics, and (iii) virus-induced cell damage differ in tolerant and susceptible BTV-infected cells. Understanding the complex virus-host interactions in disease tolerance can allow us to disentangle the critical balance between protective and damaging host immune responses.


Assuntos
Bluetongue , Interferon Tipo I , Feminino , Ovinos , Animais , Bovinos , Interferon Tipo I/genética , Bluetongue/metabolismo , Viremia , Antivirais
3.
PLoS Pathog ; 19(3): e1011283, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36996243

RESUMO

Toscana virus (TOSV) (Bunyavirales, Phenuiviridae, Phlebovirus, Toscana phlebovirus) and other related human pathogenic arboviruses are transmitted by phlebotomine sand flies. TOSV has been reported in nations bordering the Mediterranean Sea among other regions. Infection can result in febrile illness as well as meningitis and encephalitis. Understanding vector-arbovirus interactions is crucial to improving our knowledge of how arboviruses spread, and in this context, immune responses that control viral replication play a significant role. Extensive research has been conducted on mosquito vector immunity against arboviruses, with RNA interference (RNAi) and specifically the exogenous siRNA (exo-siRNA) pathway playing a critical role. However, the antiviral immunity of phlebotomine sand flies is less well understood. Here we were able to show that the exo-siRNA pathway is active in a Phlebotomus papatasi-derived cell line. Following TOSV infection, distinctive 21 nucleotide virus-derived small interfering RNAs (vsiRNAs) were detected. We also identified the exo-siRNA effector Ago2 in this cell line, and silencing its expression rendered the exo-siRNA pathway largely inactive. Thus, our data show that this pathway is active as an antiviral response against a sand fly transmitted bunyavirus, TOSV.


Assuntos
Arbovírus , Phlebotomus , Phlebovirus , Psychodidae , Vírus da Febre do Flebótomo Napolitano , Animais , Humanos , Vírus da Febre do Flebótomo Napolitano/genética , Phlebotomus/genética , Psychodidae/genética , Interferência de RNA , Phlebovirus/genética , Arbovírus/genética , RNA Interferente Pequeno/genética
4.
Proc Natl Acad Sci U S A ; 119(24): e2114309119, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35675424

RESUMO

Viruses transmitted by Aedes mosquitoes are an increasingly important global cause of disease. Defining common determinants of host susceptibility to this large group of heterogenous pathogens is key for informing the rational design of panviral medicines. Infection of the vertebrate host with these viruses is enhanced by mosquito saliva, a complex mixture of salivary-gland-derived factors and microbiota. We show that the enhancement of infection by saliva was dependent on vascular function and was independent of most antisaliva immune responses, including salivary microbiota. Instead, the Aedes gene product sialokinin mediated the enhancement of virus infection through a rapid reduction in endothelial barrier integrity. Sialokinin is unique within the insect world as having a vertebrate-like tachykinin sequence and is absent from Anopheles mosquitoes, which are incompetent for most arthropod-borne viruses, whose saliva was not proviral and did not induce similar vascular permeability. Therapeutic strategies targeting sialokinin have the potential to limit disease severity following infection with Aedes-mosquito-borne viruses.


Assuntos
Aedes , Infecções por Arbovirus , Arbovírus , Saliva , Taquicininas , Viroses , Aedes/genética , Aedes/virologia , Animais , Infecções por Arbovirus/transmissão , Arbovírus/genética , Arbovírus/metabolismo , Saliva/virologia , Taquicininas/genética , Taquicininas/metabolismo , Viroses/transmissão
5.
PLoS Pathog ; 18(1): e1010202, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34990484

RESUMO

The exogenous small interfering RNA (exo-siRNA) pathway is a key antiviral mechanism in the Aedes aegypti mosquito, a widely distributed vector of human-pathogenic arboviruses. This pathway is induced by virus-derived double-stranded RNAs (dsRNA) that are cleaved by the ribonuclease Dicer 2 (Dcr2) into predominantly 21 nucleotide (nt) virus-derived small interfering RNAs (vsiRNAs). These vsiRNAs are used by the effector protein Argonaute 2 within the RNA-induced silencing complex to cleave target viral RNA. Dcr2 contains several domains crucial for its activities, including helicase and RNase III domains. In Drosophila melanogaster Dcr2, the helicase domain has been associated with binding to dsRNA with blunt-ended termini and a processive siRNA production mechanism, while the platform-PAZ domains bind dsRNA with 3' overhangs and subsequent distributive siRNA production. Here we analyzed the contributions of the helicase and RNase III domains in Ae. aegypti Dcr2 to antiviral activity and to the exo-siRNA pathway. Conserved amino acids in the helicase and RNase III domains were identified to investigate Dcr2 antiviral activity in an Ae. aegypti-derived Dcr2 knockout cell line by reporter assays and infection with mosquito-borne Semliki Forest virus (Togaviridae, Alphavirus). Functionally relevant amino acids were found to be conserved in haplotype Dcr2 sequences from field-derived Ae. aegypti across different continents. The helicase and RNase III domains were critical for silencing activity and 21 nt vsiRNA production, with RNase III domain activity alone determined to be insufficient for antiviral activity. Analysis of 21 nt vsiRNA sequences (produced by functional Dcr2) to assess the distribution and phasing along the viral genome revealed diverse yet highly consistent vsiRNA pools, with predominantly short or long sequence overlaps including 19 nt overlaps (the latter representing most likely true Dcr2 cleavage products). Combined with the importance of the Dcr2 helicase domain, this suggests that the majority of 21 nt vsiRNAs originate by processive cleavage. This study sheds new light on Ae. aegypti Dcr2 functions and properties in this important arbovirus vector species.


Assuntos
Aedes/imunologia , Aedes/virologia , Infecções por Alphavirus/imunologia , Ribonuclease III/imunologia , Aedes/genética , Animais , Análise Mutacional de DNA , Mosquitos Vetores/virologia , RNA Interferente Pequeno/imunologia , RNA Viral/imunologia , Ribonuclease III/genética , Vírus da Floresta de Semliki
6.
Science ; 374(6567): eabj3624, 2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-34581622

RESUMO

Inherited genetic factors can influence the severity of COVID-19, but the molecular explanation underpinning a genetic association is often unclear. Intracellular antiviral defenses can inhibit the replication of viruses and reduce disease severity. To better understand the antiviral defenses relevant to COVID-19, we used interferon-stimulated gene (ISG) expression screening to reveal that 2'-5'-oligoadenylate synthetase 1 (OAS1), through ribonuclease L, potently inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We show that a common splice-acceptor single-nucleotide polymorphism (Rs10774671) governs whether patients express prenylated OAS1 isoforms that are membrane-associated and sense-specific regions of SARS-CoV-2 RNAs or if they only express cytosolic, nonprenylated OAS1 that does not efficiently detect SARS-CoV-2. In hospitalized patients, expression of prenylated OAS1 was associated with protection from severe COVID-19, suggesting that this antiviral defense is a major component of a protective antiviral response.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/metabolismo , COVID-19/genética , COVID-19/fisiopatologia , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , SARS-CoV-2/fisiologia , Regiões 5' não Traduzidas , Células A549 , Animais , COVID-19/enzimologia , COVID-19/imunologia , Quirópteros/genética , Quirópteros/virologia , Coronaviridae/enzimologia , Coronaviridae/genética , Coronaviridae/fisiologia , Endorribonucleases/metabolismo , Humanos , Interferons/imunologia , Isoenzimas/genética , Isoenzimas/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Polimorfismo de Nucleotídeo Único , Prenilação de Proteína , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/genética , RNA Viral/química , RNA Viral/genética , Retroelementos , SARS-CoV-2/genética , Índice de Gravidade de Doença , Replicação Viral
7.
PLoS Pathog ; 17(9): e1009870, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34473801

RESUMO

As mosquito females require a blood meal to reproduce, they can act as vectors of numerous pathogens, such as arboviruses (e.g. Zika, dengue and chikungunya viruses), which constitute a substantial worldwide public health burden. In addition to blood meals, mosquito females can also take sugar meals to get carbohydrates for their energy reserves. It is now recognised that diet is a key regulator of health and disease outcome through interactions with the immune system. However, this has been mostly studied in humans and model organisms. So far, the impact of sugar feeding on mosquito immunity and in turn, how this could affect vector competence for arboviruses has not been explored. Here, we show that sugar feeding increases and maintains antiviral immunity in the digestive tract of the main arbovirus vector Aedes aegypti. Our data demonstrate that the gut microbiota does not mediate the sugar-induced immunity but partly inhibits it. Importantly, sugar intake prior to an arbovirus-infected blood meal further protects females against infection with arboviruses from different families. Sugar feeding blocks arbovirus initial infection and dissemination from the gut and lowers infection prevalence and intensity, thereby decreasing the transmission potential of female mosquitoes. Finally, we show that the antiviral role of sugar is mediated by sugar-induced immunity. Overall, our findings uncover a crucial role of sugar feeding in mosquito antiviral immunity which in turn decreases vector competence for arboviruses. Since Ae. aegypti almost exclusively feed on blood in some natural settings, our findings suggest that this lack of sugar intake could increase the spread of mosquito-borne arboviral diseases.


Assuntos
Aedes/virologia , Infecções por Arbovirus , Dieta , Insetos Vetores/virologia , Intestinos/imunologia , Aedes/imunologia , Animais , Arbovírus , Insetos Vetores/imunologia , Açúcares
8.
Viruses ; 13(6)2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34205194

RESUMO

Mosquitoes are known as important vectors of many arthropod-borne (arbo)viruses causing disease in humans. These include dengue (DENV) and Zika (ZIKV) viruses. The exogenous small interfering (si)RNA (exo-siRNA) pathway is believed to be the main antiviral defense in arthropods, including mosquitoes. During infection, double-stranded RNAs that form during viral replication and infection are cleaved by the enzyme Dicer 2 (Dcr2) into virus-specific 21 nt vsiRNAs, which are subsequently loaded into Argonaute 2 (Ago2). Ago2 then targets and subsequently cleaves complementary RNA sequences, resulting in degradation of the target viral RNA. Although various studies using silencing approaches have supported the antiviral activity of the exo-siRNA pathway in mosquitoes, and despite strong similarities between the siRNA pathway in the Drosophila melanogaster model and mosquitoes, important questions remain unanswered. The antiviral activity of Ago2 against different arboviruses has been previously demonstrated. However, silencing of Ago2 had no effect on ZIKV replication, whereas Dcr2 knockout enhanced its replication. These findings raise the question as to the role of Ago2 and Dcr2 in the control of arboviruses from different viral families in mosquitoes. Using a newly established Ago2 knockout cell line, alongside the previously reported Dcr2 knockout cell line, we investigated the impact these proteins have on the modulation of different arboviral infections. Infection of Ago2 knockout cell line with alpha- and bunyaviruses resulted in an increase of viral replication, but not in the case of ZIKV. Analysis of small RNA sequencing data in the Ago2 knockout cells revealed a lack of methylated siRNAs from different sources, such as acute and persistently infecting viruses-, TE- and transcriptome-derived RNAs. The results confirmed the importance of the exo-siRNA pathway in the defense against arboviruses, but highlights variability in its response to different viruses and the impact the siRNA pathway proteins have in controlling viral replication. Moreover, this established Ago2 knockout cell line can be used for functional Ago2 studies, as well as research on the interplay between the RNAi pathways.


Assuntos
Aedes/genética , Aedes/virologia , Infecções por Arbovirus/transmissão , Infecções por Arbovirus/virologia , Arbovírus/fisiologia , Proteínas Argonautas/deficiência , Mosquitos Vetores/genética , Mosquitos Vetores/virologia , Animais , Linhagem Celular , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno , Interferência de RNA , Replicação Viral
9.
Nat Commun ; 12(1): 2766, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33986255

RESUMO

The escalating global prevalence of arboviral diseases emphasizes the need to improve our understanding of their biology. Research in this area has been hindered by the lack of molecular tools for studying virus-mosquito interactions. Here, we develop an Aedes aegypti cell line which stably expresses Zika virus (ZIKV) capsid proteins in order to study virus-vector protein-protein interactions through quantitative label-free proteomics. We identify 157 interactors and show that eight have potentially pro-viral activity during ZIKV infection in mosquito cells. Notably, silencing of transitional endoplasmic reticulum protein TER94 prevents ZIKV capsid degradation and significantly reduces viral replication. Similar results are observed if the TER94 ortholog (VCP) functioning is blocked with inhibitors in human cells. In addition, we show that an E3 ubiquitin-protein ligase, UBR5, mediates the interaction between TER94 and ZIKV capsid. Our study demonstrates a pro-viral function for TER94/VCP during ZIKV infection that is conserved between human and mosquito cells.


Assuntos
Proteínas do Capsídeo/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Proteína com Valosina/metabolismo , Zika virus/metabolismo , Células A549 , Aedes/virologia , Animais , Capsídeo/metabolismo , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Humanos , Mapas de Interação de Proteínas , Interferência de RNA , RNA Interferente Pequeno/genética , Proteína com Valosina/genética , Replicação Viral/fisiologia , Zika virus/genética , Infecção por Zika virus/patologia
10.
Antiviral Res ; 183: 104939, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32980446

RESUMO

Yellow fever virus (YFV), a member of the Flaviviridae family, is an arthropod-borne virus that can cause severe disease in humans with a lethality rate of up to 60%. Since 2017, increases in YFV activity in areas of South America and Africa have been described. Although a vaccine is available, named strain 17D (Theiler and Smith, 1937), it is contraindicated for use in the elderly, expectant mothers, immunocompromised people, among others. To this day there is no antiviral treatment against YFV to reduce the severity of viral infection. Here, we used a circular polymerase extension reaction (CPER)-based reverse genetics approach to generate a full-length reporter virus (YFVhb) by introducing a small HiBit tag in the NS1 protein. The reporter virus replicates at a similar rate to the parental YFV in HuH-7 cells. Using YFVhb, we designed a high throughput antiviral screening luciferase-based assay to identify inhibitors that target any step of the viral replication cycle. We validated our assay by using a range of inhibitors including drugs, immune sera and neutralizing single chain variable fragments (scFv). In light of the recent upsurge in YFV and a potential spread of the virus, this assay is a further tool in the development of antiviral therapy against YFV.


Assuntos
Antivirais/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Genética Reversa/métodos , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/genética , Animais , Linhagem Celular , Descoberta de Drogas/métodos , Genes Reporter , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Replicação Viral/efeitos dos fármacos , Vírus da Febre Amarela/isolamento & purificação , Vírus da Febre Amarela/fisiologia
11.
Viruses ; 12(7)2020 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-32664591

RESUMO

Mosquitoes, such as Aedes aegypti, can transmit arboviruses to humans. The exogenous short interfering RNA (exo-siRNA) pathway plays a major antiviral role in controlling virus infection in mosquito cells. The Dicer 2 (Dcr2) nuclease is a key effector protein in this pathway, which cleaves viral double-stranded RNA into virus-derived siRNAs that are further loaded onto an effector called Argonaute 2 (Ago2), which as part of the multiprotein RNA-induced silencing complex (RISC) targets and cleaves viral RNA. In order to better understand the effector protein Dcr2, proteomics experiments were conducted to identify interacting cellular partners. We identified several known interacting partners including Ago2, as well as two novel and previously uncharacterized Ae. aegypti proteins. The role of these two proteins was further investigated, and their interactions with Dcr2 verified by co-immunoprecipitation. Interestingly, despite their ability to interact with Ago2 and Piwi4, neither of these proteins was found to affect exo-siRNA silencing in a reporter assay. However, one of these proteins, Q0IFK9, subsequently called aBravo (aedine broadly active antiviral protein), was found to mediate antiviral activity against positive strand RNA arboviruses. Intriguingly the presence of Dcr2 was not necessary for this effect, suggesting that this interacting antiviral effector may act as part of protein complexes with potentially separate antiviral activities.


Assuntos
Aedes/metabolismo , Arbovírus/metabolismo , Proteínas de Insetos/metabolismo , RNA Interferente Pequeno/metabolismo , Aedes/virologia , Animais , Western Blotting , Imunoprecipitação , Proteínas de Insetos/isolamento & purificação , Reação em Cadeia da Polimerase , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo
12.
Viruses ; 12(5)2020 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-32397571

RESUMO

Zika virus (ZIKV; Flaviviridae) is a mosquito-borne flavivirus shown to cause fetal abnormalities collectively known as congenital Zika syndrome and Guillain-Barré syndrome in recent outbreaks. Currently, there is no specific treatment or vaccine available, and more effort is needed to identify cellular factors in the viral life cycle. Here, we investigated interactors of ZIKV envelope (E) protein by combining protein pull-down with mass spectrometry. We found that E interacts with the endoplasmic reticulum (ER) resident chaperone, glucose regulated protein 78 (GRP78). Although other flaviviruses are known to co-opt ER resident proteins, including GRP78, to enhance viral infectivity, the role ER proteins play during the ZIKV life cycle is yet to be elucidated. We showed that GRP78 levels increased during ZIKV infection and localised to sites coincident with ZIKV E staining. Depletion of GRP78 using specific siRNAs significantly reduced reporter-virus luciferase readings, viral protein synthesis, and viral titres. Additionally, GRP78 depletion reduced the ability of ZIKV to disrupt host cell translation and altered the localisation of viral replication factories, though there was no effect on viral RNA synthesis. In summary, we showed GRP78 is a vital host-factor during ZIKV infection, which may be involved in the coordination of viral replication factories.


Assuntos
Proteínas de Choque Térmico/metabolismo , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Infecção por Zika virus/metabolismo , Zika virus/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/genética , Interações Hospedeiro-Patógeno , Humanos , Ligação Proteica , Proteínas do Envelope Viral/genética , Zika virus/genética , Infecção por Zika virus/genética , Infecção por Zika virus/virologia
13.
mSphere ; 5(2)2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32269152

RESUMO

Arboviruses are pathogens of humans and animals. A better understanding of the interactions between these pathogens and the arthropod vectors, such as mosquitoes, that transmit them is necessary to develop novel control measures. A major antiviral pathway in the mosquito vector is the exogenous small interfering RNA (exo-siRNA) pathway, which is induced by arbovirus-derived double-stranded RNA in infected cells. Although recent work has shown the key role played by Argonaute-2 (Ago-2) and Dicer-2 (Dcr-2) in this pathway, the regulatory mechanisms that govern these pathways have not been studied in mosquitoes. Here, we show that the Domino ortholog p400 has antiviral activity against the alphavirus Semliki Forest virus (Togaviridae) both in Aedes aegypti-derived cells and in vivo Antiviral activity of p400 was also demonstrated against chikungunya virus (Togaviridae) and Bunyamwera virus (Peribunyaviridae) but not Zika virus (Flaviviridae). p400 was found to be expressed across mosquito tissues and regulated ago-2 but not dcr-2 transcript levels in A. aegypti mosquitoes. These findings provide novel insights into the regulation of an important aedine exo-siRNA pathway effector protein, Ago-2, by the Domino ortholog p400. They add functional insights to previous observations of this protein's antiviral and RNA interference regulatory activities in Drosophila melanogasterIMPORTANCE Female Aedes aegypti mosquitoes are vectors of human-infecting arthropod-borne viruses (arboviruses). In recent decades, the incidence of arthropod-borne viral infections has grown dramatically. Vector competence is influenced by many factors, including the mosquito's antiviral defenses. The exogenous small interfering RNA (siRNA) pathway is a major antiviral response restricting arboviruses in mosquitoes. While the roles of the effectors of this pathway, Argonaute-2 and Dicer-2 are well characterized, nothing is known about its regulation in mosquitoes. In this study, we demonstrate that A. aegypti p400, whose ortholog Domino in Drosophila melanogaster is a chromatin-remodeling ATPase member of the Tip60 complex, regulates siRNA pathway activity and controls ago-2 expression levels. In addition, we found p400 to have antiviral activity against different arboviruses. Therefore, our study provides new insights into the regulation of the antiviral response in A. aegypti mosquitoes.


Assuntos
Aedes/genética , Proteínas Argonautas/genética , Proteínas de Insetos/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Aedes/virologia , Animais , Arbovírus/fisiologia , Feminino , Regulação da Expressão Gênica , Mosquitos Vetores/genética , Mosquitos Vetores/virologia
14.
Sci Transl Med ; 12(527)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31969486

RESUMO

Arthropod-borne viruses (arboviruses) are important human pathogens for which there are no specific antiviral medicines. The abundance of genetically distinct arbovirus species, coupled with the unpredictable nature of their outbreaks, has made the development of virus-specific treatments challenging. Instead, we have defined and targeted a key aspect of the host innate immune response to virus at the arthropod bite that is common to all arbovirus infections, potentially circumventing the need for virus-specific therapies. Using mouse models and human skin explants, we identify innate immune responses by dermal macrophages in the skin as a key determinant of disease severity. Post-exposure treatment of the inoculation site by a topical TLR7 agonist suppressed both the local and subsequent systemic course of infection with a variety of arboviruses from the Alphavirus, Flavivirus, and Orthobunyavirus genera. Clinical outcome was improved in mice after infection with a model alphavirus. In the absence of treatment, antiviral interferon expression to virus in the skin was restricted to dermal dendritic cells. In contrast, stimulating the more populous skin-resident macrophages with a TLR7 agonist elicited protective responses in key cellular targets of virus that otherwise proficiently replicated virus. By defining and targeting a key aspect of the innate immune response to virus at the mosquito bite site, we have identified a putative new strategy for limiting disease after infection with a variety of genetically distinct arboviruses.


Assuntos
Infecções por Arbovirus/imunologia , Infecções por Arbovirus/metabolismo , Arbovírus/imunologia , Arbovírus/patogenicidade , Macrófagos/metabolismo , Pele/citologia , Alphavirus/imunologia , Alphavirus/patogenicidade , Animais , Flavivirus/imunologia , Flavivirus/patogenicidade , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos , Orthobunyavirus/imunologia , Orthobunyavirus/patogenicidade , Receptor 7 Toll-Like/metabolismo
15.
Viruses ; 10(12)2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30563205

RESUMO

Arthropod vectors control the replication of arboviruses through their innate antiviral immune responses. In particular, the RNA interference (RNAi) pathways are of notable significance for the control of viral infections. Although much has been done to understand the role of RNAi in vector populations, little is known about its importance in non-vector mosquito species. In this study, we investigated the presence of an RNAi response in Toxorhynchites amboinensis, which is a non-blood feeding species proposed as a biological control agent against pest mosquitoes. Using a derived cell line (TRA-171), we demonstrate that these mosquitoes possess a functional RNAi response that is active against a mosquito-borne alphavirus, Semliki Forest virus. As observed in vector mosquito species, small RNAs are produced that target viral sequences. The size and characteristics of these small RNAs indicate that both the siRNA and piRNA pathways are induced in response to infection. Taken together, this data suggests that Tx. amboinensis are able to control viral infections in a similar way to natural arbovirus vector mosquito species. Understanding their ability to manage arboviral infections will be advantageous when assessing these and similar species as biological control agents.


Assuntos
Culicidae/genética , Culicidae/virologia , Interferência de RNA , Vírus da Floresta de Semliki/genética , Infecções por Alphavirus/imunologia , Animais , Agentes de Controle Biológico , Linhagem Celular , Culicidae/citologia , Sequenciamento de Nucleotídeos em Larga Escala , Imunidade Inata , Mosquitos Vetores/genética , Mosquitos Vetores/virologia , RNA Interferente Pequeno/genética , Vírus da Floresta de Semliki/imunologia , Replicação Viral
16.
J Gen Virol ; 99(12): 1551-1562, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30372405

RESUMO

There are several RNA interference (RNAi) pathways in insects. The small interfering RNA pathway is considered to be the main antiviral mechanism of the innate immune system; however, virus-specific P-element-induced Wimpy testis gene (PIWI)-interacting RNAs (vpiRNAs) have also been described, especially in mosquitoes. Understanding the antiviral potential of the RNAi pathways is important, given that many human and animal pathogens are transmitted by mosquitoes, such as Zika virus, dengue virus and chikungunya virus. In recent years, significant progress has been made to characterize the piRNA pathway in mosquitoes (including the possible antiviral activity) and to determine the differences between mosquitoes and the model organism Drosophila melanogaster. The new findings, especially regarding vpiRNA in mosquitoes, as well as important questions that need to be tackled in the future, are discussed in this review.


Assuntos
Culicidae/imunologia , Culicidae/virologia , Imunidade Inata , Fatores Imunológicos/metabolismo , Interferência de RNA , Vírus de RNA/imunologia , RNA Interferente Pequeno/metabolismo , Animais , Antivirais/metabolismo , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia
17.
Viruses ; 10(5)2018 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-29702546

RESUMO

Infection by Chikungunya virus (CHIKV) of the Old World alphaviruses (family Togaviridae) in humans can cause arthritis and arthralgia. The virus encodes four non-structural proteins (nsP) (nsP1, nsp2, nsP3 and nsP4) that act as subunits of the virus replicase. These proteins also interact with numerous host proteins and some crucial interactions are mediated by the unstructured C-terminal hypervariable domain (HVD) of nsP3. In this study, a human cell line expressing EGFP tagged with CHIKV nsP3 HVD was established. Using quantitative proteomics, it was found that CHIKV nsP3 HVD can bind cytoskeletal proteins, including CD2AP, SH3KBP1, CAPZA1, CAPZA2 and CAPZB. The interaction with CD2AP was found to be most evident; its binding site was mapped to the second SH3 ligand-like element in nsP3 HVD. Further assessment indicated that CD2AP can bind to nsP3 HVDs of many different New and Old World alphaviruses. Mutation of the short binding element hampered the ability of the virus to establish infection. The mutation also abolished ability of CD2AP to co-localise with nsP3 and replication complexes of CHIKV; the same was observed for Semliki Forest virus (SFV) harbouring a similar mutation. Similar to CD2AP, its homolog SH3KBP1 also bound the identified motif in CHIKV and SFV nsP3.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Alphavirus/fisiologia , Motivos de Aminoácidos/genética , Sítios de Ligação/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Alphavirus/genética , Animais , Proteína de Capeamento de Actina CapZ/metabolismo , Linhagem Celular , Vírus Chikungunya/genética , Vírus Chikungunya/fisiologia , Cricetinae , Interações Hospedeiro-Patógeno , Humanos , Mutação , Ligação Proteica , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/fisiologia , Proteínas não Estruturais Virais/genética , Replicação Viral/genética
18.
Viruses ; 10(2)2018 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-29463033

RESUMO

Mosquitoes transmit several human- and animal-pathogenic alphaviruses (Togaviridae family). In alphavirus-infected mosquito cells two different types of virus-specific small RNAs are produced as part of the RNA interference response: short-interfering (si)RNAs and PIWI-interacting (pi)RNAs. The siRNA pathway is generally thought to be the main antiviral pathway. Although an antiviral activity has been suggested for the piRNA pathway its role in host defences is not clear. Knock down of key proteins of the piRNA pathway (Ago3 and Piwi5) in Aedesaegypti-derived cells reduced the production of alphavirus chikungunya virus (CHIKV)-specific piRNAs but had no effect on virus replication. In contrast, knock down of the siRNA pathway key protein Ago2 resulted in an increase in virus replication. Similar results were obtained when expression of Piwi4 was silenced. Knock down of the helicase Spindle-E (SpnE), an essential co-factor of the piRNA pathway in Drosophila melanogaster, resulted in increased virus replication indicating that SpnE acts as an antiviral against alphaviruses such as CHIKV and the related Semliki Forest virus (SFV). Surprisingly, this effect was found to be independent of the siRNA and piRNA pathways in Ae. aegypti cells and specific for alphaviruses. This suggests a small RNA-independent antiviral function for this protein in mosquitoes.


Assuntos
Infecções por Alphavirus/virologia , Alphavirus , Culicidae/virologia , Animais , Células Cultivadas , Proteínas de Insetos/genética , Interferência de RNA , Pequeno RNA não Traduzido/genética , Replicação Viral
19.
J Gen Virol ; 99(2): 258-264, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29076805

RESUMO

The rapid spread of Zika virus (ZIKV) in the Americas raised many questions about the role of Culex quinquefasciatus mosquitoes in transmission, in addition to the key role played by the vector Aedes aegypti. Here we analysed the competence of Cx. quinquefasciatus (with or without Wolbachia endosymbionts) for a ZIKV isolate. We also examined the induction of RNA interference pathways after viral challenge and the production of small virus-derived RNAs. We did not observe any infection nor such small virus-derived RNAs, regardless of the presence or absence of Wolbachia. Thus, Cx. quinquefasciatus does not support ZIKV replication and Wolbachia is not involved in producing this phenotype. In short, these mosquitoes are very unlikely to play a role in transmission of ZIKV.


Assuntos
Culex/virologia , Mosquitos Vetores/virologia , Replicação Viral , Wolbachia/fisiologia , Infecção por Zika virus/transmissão , Zika virus/fisiologia , Animais , Feminino , Fenótipo , Infecção por Zika virus/virologia
20.
PLoS Negl Trop Dis ; 11(12): e0006155, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29267287

RESUMO

Rift Valley fever phlebovirus (RVFV; Phenuiviridae, Phlebovirus) is an important mosquito-borne pathogen of both humans and ruminants. The RVFV genome is composed of tripartite, single stranded, negative or ambisense RNAs. The small (S) segment encodes both the nucleocapsid protein (N) and the non-structural protein (NSs). The N protein is responsible for the formation of the viral ribonucleoprotein (RNP) complexes, which are essential in the virus life cycle and for the transcription and replication of the viral genome. There is currently limited knowledge surrounding the roles of the RVFV nucleocapsid protein in viral infection other than its key functions: N protein multimerisation, encapsidation of the RNA genome and interactions with the RNA-dependent RNA polymerase, L. By bioinformatic comparison of the N sequences of fourteen phleboviruses, mutational analysis, minigenome assays and packaging assays, we have further characterised the RVFV N protein. Amino acids P11 and F149 in RVFV N play an essential role in the function of RNPs and are neither associated with N protein multimerisation nor known nucleocapsid protein functions and may have additional roles in the virus life cycle. Amino acid Y30 exhibited increased minigenome activity despite reduced RNA binding capacity. Additionally, we have determined that the N-terminal arm of N protein is not involved in N-L interactions. Elucidating the fundamental processes that involve the nucleocapsid protein will add to our understanding of this important viral protein and may influence future studies in the development of novel antiviral strategies.


Assuntos
Análise Mutacional de DNA , Genoma Viral/genética , Proteínas do Nucleocapsídeo/genética , RNA Viral/genética , Vírus da Febre do Vale do Rift/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Multimerização Proteica/genética , Multimerização Proteica/fisiologia , Proteínas de Ligação a RNA/genética , Febre do Vale de Rift/virologia , Alinhamento de Sequência , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...