Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
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
2.
Viral Immunol ; 36(1): 33-40, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36399689

RESUMO

Rift Valley fever virus (RVFV) is an emerging arbovirus that affects both ruminants and humans. RVFV causes severe and recurrent outbreaks in Africa and the Arabian Peninsula with a significant risk for emergence into new locations. Although there are a variety of RVFV veterinary vaccines for use in endemic areas, there is currently no licensed vaccine for human use; therefore, there is a need to develop and assess new vaccines. Herein, we report a live-attenuated recombinant vaccine candidate for RVFV, based on the previously described genomic reconfiguration of the conditionally licensed MP12 vaccine. There are two general strategies used to develop live-attenuated RVFV vaccines, one being serial passage of wild-type RVFV strains to select attenuated mutants such as Smithburn, Clone 13, and MP12 vaccine strains. The second strategy has utilized reverse genetics to attenuate RVFV strains by introducing deletions or insertions within the viral genome. The novel candidate vaccine characterized in this report contains a two-segmented genome that lacks the medium viral segment (M) and two virulence genes (nonstructural small and nonstructural medium). The vaccine candidate, named r2segMP12, was evaluated for the production of neutralizing antibodies to RVFV in outbred CD-1 mice. The immune response induced by the r2segMP12 vaccine candidate was directly compared to the immune response induced by the rMP12 parental strain vaccine. Our study demonstrated that a single immunization with the r2segMP12 vaccine candidate at 105 plaque-forming units elicited a higher neutralizing antibody response than the rMP12 vaccine at the same vaccination titer without the need for a booster.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Virais , Humanos , Animais , Camundongos , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/epidemiologia , Vacinas Atenuadas/genética , Vacinas Virais/genética , Anticorpos Neutralizantes
3.
J Anat ; 242(1): 64-75, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35255526

RESUMO

Chronic total occlusions (CTOs) occur in approximately 40% of individuals with symptomatic peripheral arterial disease and are indicative of critical limb ischaemia. Currently, few medical devices can effectively treat CTOs long-term, with amputation often required. This is due to a lack of knowledge of CTO anatomy, making device design and testing difficult. This study is a proof-of-concept study, which aimed to develop a workflow for further characterising the complex multi-material anatomy of CTOs and creating 3D models of CTO components, which may be useful in producing a vascular CTO biomimetic for device testing. Here, we establish such a workflow using samples of atheromatous plaques. We focus on a high-resolution, non-destructive microcomputed tomography (µCT) technique which enables visualisation of occlusion anatomy at a greater resolution than computed tomography angiography (CTA), which is the typical modality used for CTO clinical visualisation. Four arteries (n = 2 superficial femoral; n = 2 popliteal) with evidence of atheromatous plaques were cut into 8 cm segments, which were then stained with iodine and scanned at low resolution, with calcified regions rescanned at high resolution. Resulting files were manually segmented to generate 3D models, which were then 3D printed in resin using a stereolithography printer to produce parts suitable for creating a biomimetic. In total, µCT files from three arterial segments (n = 2 high resolution, n = 1 low resolution) were deemed suitably calcified for segmentation, and thus were segmented to produce 3D models. 3D models of the arterial wall, intima and atheromatous calcium deposits from a high-resolution popliteal artery scan were successfully 3D printed at several scales. While this research is at an early stage, it holds great promise. The workflow for segmentation and 3D printing various components of an atheromatous plaque established here is replicable and uses software and equipment which are accessible to research laboratories in both academia and industry. The ability to print detailed models on a desktop 3D printer is unprecedented and can be improved further, which is promising for future development of biomimetics with multi-material detail of both soft tissue and calcified components of a vascular occlusion. Indeed, this workflow provides a solid foundation for future studies of CTO anatomy and the creation of true, multi-material CTO biomimetics. Such biomimetics may enable the development of improved interventional devices, as they would mimic the general in vivo CTO environment. As this method cannot be applied in vivo, we cannot yet produce patient-specific biomimetics, however, these analogues would still be important in device development, which would improve patient outcomes in critical limb ischaemia.


Assuntos
Biomimética , Placa Aterosclerótica , Humanos , Isquemia Crônica Crítica de Membro , Microtomografia por Raio-X , Impressão Tridimensional , Resultado do Tratamento
4.
Curr Opin Virol ; 57: 101278, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36375406

RESUMO

The Bunyavirales order is the largest grouping of RNA viruses, comprising emerging and re-emerging human, plant and animal pathogens. Bunyaviruses have a global distribution and many members of the order are transmitted by arthropods. They have evolved a plethora of mechanisms to manipulate the regulatory processes of the infected cell to facilitate their own replicative cycle, in hosts of disparate phylogenies. Interest in virus-vector interactions is growing rapidly. However, current understanding of tick-borne bunyavirus cellular interaction is heavily biased to studies conducted in mammalian systems. In this short review, we summarise current understandings of how tick-borne bunyaviruses utilise major cellular pathways (innate immunity, apoptosis and RNAi responses) in mammalian or tick cells to facilitate virus replication.


Assuntos
Infecções por Bunyaviridae , Bunyaviridae , Orthobunyavirus , Doenças Transmitidas por Carrapatos , Carrapatos , Animais , Humanos , Orthobunyavirus/genética , Interações entre Hospedeiro e Microrganismos , Bunyaviridae/fisiologia , Mamíferos
5.
Vector Borne Zoonotic Dis ; 22(11): 553-558, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36354965

RESUMO

Background: The emergence or re-emergence of several orthobunyaviruses (order: Bunyavirales; family: Peribunyaviridae), including Cache Valley virus (CVV) and Oropouche virus, warrants the development and evaluation of candidate live-attenuated vaccines (LAVs). Ideally, these vaccines would elicit long-lasting immunity with one single immunization. Materials and Methods: Since the deletion of two virulence factors, NSs and NSm, has been shown to attenuate the virulence phenotype of orthobunyaviruses, phleboviruses, and nairoviruses, genetic manipulation of the viral genome is considered an effective strategy for the rational design of candidate LAVs for bunyaviruses across multiple families. In addition, the deletion of Rift Valley fever virus NSs and NSm genes has been shown to reduce transmission by mosquitoes. Results: In this study, the ability of a CVV mutant lacking the NSs and NSm genes (2delCVV) to replicate in intrathoracically injected Aedes albopictus was compared with the parental wild-type CVV (wtCVV) 6V633 strain. In contrast to the robust replication of wtCVV in injected mosquitoes, the multiplication kinetics of the 2delCVV mutant was reduced by more than a 100-fold. Conclusion: These results suggest that the deletion of NSm and NSs genes is a feasible approach to rationally design candidate orthobunyavirus LAVs that are highly attenuated in mosquitoes and, therefore, pose little risk of reversion to virulence and transmission.


Assuntos
Aedes , Vírus Bunyamwera , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Virais , Animais , Vacinas Atenuadas , Cinética , Vírus da Febre do Vale do Rift/genética , Replicação Viral
6.
Trans R Soc Trop Med Hyg ; 116(11): 1082-1090, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36040309

RESUMO

BACKGROUND: Human and animal cases of Rift Valley fever (RVF) are typically only reported during large outbreaks. The occurrence of RVF cases that go undetected by national surveillance systems in the period between these outbreaks is considered likely. The last reported cases of RVF in Tanzania occurred during a large outbreak in 2007-2008. METHODS: Samples collected between 2017 and 2019 from livestock suffering abortion across northern Tanzania were retrospectively tested for evidence of RVF virus infection using serology and reverse transcription quantitative polymerase chain reaction (RT-qPCR). RESULTS: A total of 14 RVF-associated cattle abortions were identified among dairy cattle in a peri-urban area surrounding the town of Moshi. RVF cases occurred from May to August 2018 and were considered to represent an undetected, small-scale RVF outbreak. Milk samples from 3 of 14 cases (21%) were found to be RT-qPCR positive. Genotyping revealed circulation of RVF viruses from two distinct lineages. CONCLUSIONS: RVF outbreaks can occur more often in endemic settings than would be expected on the basis of detection by national surveillance. The occurrence of RVF cases among peri-urban dairy cattle and evidence for viral shedding in milk, also highlights potentially emerging risks for RVF associated with increasing urban and peri-urban livestock populations.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Bovinos , Animais , Humanos , Febre do Vale de Rift/epidemiologia , Tanzânia/epidemiologia , Estudos Retrospectivos , Surtos de Doenças/veterinária , Gado
7.
PNAS Nexus ; 1(1): pgac024, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35529317

RESUMO

Dabie bandavirus (previously severe fever with thrombocytopenia syndrome virus; SFTSV), is an emerging tick-borne bunyavirus responsible for severe fever with thrombocytopenia syndrome (SFTS), a disease with high case fatality that is characterized by high fever, thrombocytopenia, and potentially lethal hemorrhagic manifestations. Currently, neither effective therapeutic strategies nor approved vaccines exist for SFTS. Therefore, there remains a pressing need to better understand the pathogenesis of the disease and to identify therapeutic strategies to ameliorate SFTS outcomes. Using a type I interferon (IFN)-deficient mouse model, we investigated the viral tropism, disease kinetics, and the role of the virulence factor nonstructural protein (NSs) in SFTS. Ly6C+ MHCII+ cells in the lymphatic tissues were identified as an important target cell for SFTSV. Advanced SFTS was characterized by significant migration of inflammatory leukocytes, notably neutrophils, into the lymph node and spleen, however, these cells were not required to orchestrate the disease phenotype. The development of SFTS was associated with significant upregulation of proinflammatory cytokines, including high levels of IFN-γ and IL-6 in the serum, lymph node, and spleen. Humoral immunity generated by inoculation with delNSs SFTSV was 100% protective. Importantly, NSs was critical to the inhibition of the host IFNɣ response or downstream IFN-stimulated gene production and allowed for the establishment of severe disease. Finally, therapeutic but not prophylactic use of anti-IL-6 antibodies significantly increased the survival of mice following SFTSV infection and, therefore, this treatment modality presents a novel therapeutic strategy for treating severe SFTS.

8.
PLoS Biol ; 19(2): e3001091, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33630831

RESUMO

The recent emergence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the underlying cause of Coronavirus Disease 2019 (COVID-19), has led to a worldwide pandemic causing substantial morbidity, mortality, and economic devastation. In response, many laboratories have redirected attention to SARS-CoV-2, meaning there is an urgent need for tools that can be used in laboratories unaccustomed to working with coronaviruses. Here we report a range of tools for SARS-CoV-2 research. First, we describe a facile single plasmid SARS-CoV-2 reverse genetics system that is simple to genetically manipulate and can be used to rescue infectious virus through transient transfection (without in vitro transcription or additional expression plasmids). The rescue system is accompanied by our panel of SARS-CoV-2 antibodies (against nearly every viral protein), SARS-CoV-2 clinical isolates, and SARS-CoV-2 permissive cell lines, which are all openly available to the scientific community. Using these tools, we demonstrate here that the controversial ORF10 protein is expressed in infected cells. Furthermore, we show that the promising repurposed antiviral activity of apilimod is dependent on TMPRSS2 expression. Altogether, our SARS-CoV-2 toolkit, which can be directly accessed via our website at https://mrcppu-covid.bio/, constitutes a resource with considerable potential to advance COVID-19 vaccine design, drug testing, and discovery science.


Assuntos
Vacinas contra COVID-19 , COVID-19/diagnóstico , COVID-19/virologia , Genética Reversa , SARS-CoV-2/genética , Células A549 , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Chlorocebus aethiops , Códon , Humanos , Hidrazonas/farmacologia , Camundongos , Morfolinas/farmacologia , Fases de Leitura Aberta , Plasmídeos/genética , Pirimidinas/farmacologia , Serina Endopeptidases/metabolismo , Células Vero , Proteínas Virais/metabolismo
9.
Front Sports Act Living ; 3: 670018, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34977565

RESUMO

Basketball games and training sessions are characterized by quick actions and many scoring attempts, which pose biomechanical loads on the bodies of the players. Inertial Measurement Units (IMUs) capture these biomechanical loads as PlayerLoad and Inertial Movement Analysis (IMA) and teams collect those data to monitor adaptations to training schedules. However, the association of biomechanical loads with game performance is a relatively unexplored area. The aims of the current study were to determine the statistical relations between biomechanical loads in games and training with game performance. Biomechanical training and game load measures and player-level and team-level game stats from one college basketball team of two seasons were included in the dataset. The training loads were obtained on the days before gameday. A three-step analysis pipeline modeled: (i) relations between team-level game stats and the win/loss probabilities of the team, (ii) associations between the player-level training and game loads and their game stats, and (iii) associations between player-level training loads and game loads. The results showed that offensive and defensive game stats increased the odds of winning, but several stats were subject to positional and individual performance variability. Further analyses, therefore, included total points [PTS], two-point field goals, and defensive rebounds (DEF REB) that were less subject to those influences. Increases in game loads were significantly associated with game stats. In addition, training loads significantly affected the game loads in the following game. In particular, increased loads 2 days before the game resulted in increased expected game loads. Those findings suggested that biomechanical loads were good predictors for game performance. Specifically, the game loads were good predictors for game stats, and training loads 2 days before gameday were good predictors for the expected game load. The current analyses accounted for the variation in loads of players and stats that enabled modeling the expected game performance for each individual. Coaches, trainers, and sports scientists can use these findings to further optimize training plans and possibly make in-game decisions for individual player performance.

10.
Viruses ; 12(4)2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32272808

RESUMO

Toscana virus (TOSV) is a Phlebovirus in the Phenuiviridae family, order Bunyavirales, found in the countries surrounding the Mediterranean. TOSV is an important cause of seasonal acute meningitis and encephalitis within its range. Here, we determined the full sequence of the TOSV strain 1500590, a lineage A virus obtained from an infected patient (Marseille, 2007) and used this in combination with other sequence information to construct functional cDNA plasmids encoding the viral L, M, and S antigenomic sequences under the control of the T7 RNA promoter to recover recombinant viruses. Importantly, resequencing identified two single nucleotide changes to a TOSV reference genome, which, when corrected, restored functionality to the polymerase L and made it possible to recover infectious recombinant TOSV (rTOSV) from cDNA, as well as establish a minigenome system. Using reverse genetics, we produced an NSs-deletant rTOSV and also obtained viruses expressing reporter genes instead of NSs. The availability of such a system assists investigating questions that require genetic manipulation of the viral genome, such as investigations into replication and tropism, and beyond these fundamental aspects, also the development of novel vaccine design strategies.


Assuntos
Genoma Viral , Genética Reversa/métodos , Vírus da Febre do Flebótomo Napolitano/genética , Células A549 , Humanos , Febre por Flebótomos/virologia , Regiões Promotoras Genéticas , Vírus da Febre do Flebótomo Napolitano/classificação , Proteínas Virais/genética
11.
Proc Natl Acad Sci U S A ; 116(52): 26900-26908, 2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31818942

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) virus (SFTSV) is an emerging tick-borne virus classified within the Banyangvirus genus. SFTS disease has been reported throughout East Asia since 2009 and is characterized by high fever, thrombocytopenia, and leukopenia and has a 12 to 30% case fatality rate. Due to the recent emergence of SFTSV, there has been little time to conduct research into preventative measures aimed at combatting the virus. SFTSV is listed as one of the World Health Organization's Prioritized Pathogens for research into antiviral therapeutics and vaccine development. Here, we report 2 attenuated recombinant SFTS viruses that induce a humoral immune response in immunized ferrets and confer complete cross-genotype protection to lethal challenge. Animals infected with rHB29NSsP102A or rHB2912aaNSs (both genotype D) had a reduced viral load in both serum and tissues and presented without high fever, thrombocytopenia, or mortality associated with infection. rHB29NSsP102A- or rHB2912aaNSs-immunized animals developed a robust anti-SFTSV immune response against cross-genotype isolates of SFTSV. This immune response was capable of neutralizing live virus in a focus-reduction neutralization test (FRNT) and was 100% protective against a cross-genotype lethal challenge with the CB1/2014 strain of SFTSV (genotype B). Thus, using our midsized, aged ferret infection model, we demonstrate 2 live attenuated vaccine candidates against the emerging pathogen SFTSV.

12.
Int J Rheum Dis ; 22(6): 1036-1040, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30838791

RESUMO

AIM: The modified Rodnan skin score (mRSS) is a validated outcome measure for skin thickness in systemic sclerosis (SSc). Training has been shown to reduce variability in the measurement of mRSS. Our objective was to assess the inter- and intra-observer variability of mRSS scoring using the proposed recommendations for training by the Scleroderma Clinical Trials Consortium (SCTC) and World Scleroderma Foundation (WSF). METHOD: Fifty-two trainees and eight adult SSc patients participated in the SSc skin scoring workshop that was conducted in two sessions by four teachers. Each session, attended by 26 trainees, had a teaching and evaluation phase. The teaching phase comprised of: (a) lecture on mRSS scoring; (b) video demonstration of mRSS scoring; and (c) live demonstration of mRSS on one SSc patient. In the evaluation phase, each trainee independently assessed the mRSS in four SSc patients. For intra-observer reliability, 14 trainees re-assessed the mRSS of two SSc patients whom they had previously examined. We computed the inter- and intra-observer variability using a linear mixed model. RESULTS: For the evaluation phase, 34 (65.4%) trainees were within five units of the established teachers' score in 3 out of 4 patients. Overall, the whole group had acceptable inter-observer variability (intra-class correlation coefficient [ICC] = 0.71, mean = 8.64 and within-patient standard deviation [SD] = 4.25). The intra-observer ICC was 0.85 and within-patient SD was 2.73. CONCLUSION: There was good inter-observer and excellent intra-observer reliability. This is the first study examining the training of assessors using the SCTC/WSF recommendations and our results support the importance of standardized training for skin scoring.


Assuntos
Técnicas de Apoio para a Decisão , Escleroderma Sistêmico/patologia , Pele/patologia , Estudos de Viabilidade , Humanos , Variações Dependentes do Observador , Valor Preditivo dos Testes , Prognóstico , Reprodutibilidade dos Testes , Índice de Gravidade de Doença
13.
Nat Microbiol ; 4(3): 429-437, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30617349

RESUMO

Severe fever with thrombocytopenia syndrome phlebovirus (SFTSV), listed in the World Health Organization Prioritized Pathogens, is an emerging phlebovirus with a high fatality1-4. Owing to the lack of therapies and vaccines5,6, there is a pressing need to understand SFTSV pathogenesis. SFSTV non-structural protein (NSs) has been shown to block type I interferon induction7-11 and facilitate disease progression12,13. Here, we report that SFTSV-NSs targets the tumour progression locus 2 (TPL2)-A20-binding inhibitor of NF-κB activation 2 (ABIN2)-p105 complex to induce the expression of interleukin-10 (IL-10) for viral pathogenesis. Using a combination of reverse genetics, a TPL2 kinase inhibitor and Tpl2-/- mice showed that NSs interacted with ABIN2 and promoted TPL2 complex formation and signalling activity, resulting in the marked upregulation of Il10 expression. Whereas SFTSV infection of wild-type mice led to rapid weight loss and death, Tpl2-/- mice or Il10-/- mice survived an infection. Furthermore, SFTSV-NSs P102A and SFTSV-NSs K211R that lost the ability to induce TPL2 signalling and IL-10 production showed drastically reduced pathogenesis. Remarkably, the exogenous administration of recombinant IL-10 effectively rescued the attenuated pathogenic activity of SFTSV-NSs P102A, resulting in a lethal infection. Our study demonstrates that SFTSV-NSs targets the TPL2 signalling pathway to induce immune-suppressive IL-10 cytokine production as a means to dampen the host defence and promote viral pathogenesis.


Assuntos
Interações Hospedeiro-Patógeno , MAP Quinase Quinase Quinases/metabolismo , Phlebovirus/patogenicidade , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/patologia , Feminino , Células HEK293 , Células HeLa , Humanos , Interleucina-10/administração & dosagem , Interleucina-10/genética , MAP Quinase Quinase Quinases/imunologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Phlebovirus/efeitos dos fármacos , Proteínas Proto-Oncogênicas/imunologia , Células RAW 264.7 , Genética Reversa
14.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30567991

RESUMO

Bunyaviruses have a tripartite negative-sense RNA genome. Due to the segmented nature of these viruses, if two closely related viruses coinfect the same host or vector cell, it is possible that RNA segments from either of the two parental viruses will be incorporated into progeny virions to give reassortant viruses. Little is known about the ability of tick-borne phleboviruses to reassort. The present study describes the development of minigenome assays for the tick-borne viruses Uukuniemi phlebovirus (UUKV) and Heartland phlebovirus (HRTV). We used these minigenome assays in conjunction with the existing minigenome system of severe fever with thrombocytopenia syndrome (SFTS) phlebovirus (SFTSV) to assess the abilities of viral N and L proteins to recognize, transcribe, and replicate the M segment-based minigenome of a heterologous virus. The highest minigenome activity was detected with the M segment-based minigenomes of cognate viruses. However, our findings indicate that several combinations utilizing N and L proteins of heterologous viruses resulted in M segment minigenome activity. This suggests that the M segment untranslated regions (UTRs) are recognized as functional promoters of transcription and replication by the N and L proteins of related viruses. Further, virus-like particle assays demonstrated that HRTV glycoproteins can package UUKV and SFTSV S and L segment-based minigenomes. Taken together, these results suggest that coinfection with these viruses could lead to the generation of viable reassortant progeny. Thus, the tools developed in this study could aid in understanding the role of genome reassortment in the evolution of these emerging pathogens in an experimental setting.IMPORTANCE In recent years, there has been a large expansion in the number of emerging tick-borne viruses that are assigned to the Phlebovirus genus. Bunyaviruses have a tripartite segmented genome, and infection of the same host cell by two closely related bunyaviruses can, in theory, result in eight progeny viruses with different genome segment combinations. We used genome analogues expressing reporter genes to assess the abilities of Phlebovirus nucleocapsid protein and RNA-dependent RNA polymerase to recognize the untranslated region of a genome segment of a related phlebovirus, and we used virus-like particle assays to assess whether viral glycoproteins can package genome analogues of related phleboviruses. Our results provide strong evidence that these emerging pathogens could reassort their genomes if they were to meet in nature in an infected host or vector. This reassortment process could result in viruses with new pathogenic properties.


Assuntos
Genoma Viral/genética , Phlebovirus/genética , Animais , Infecções por Bunyaviridae/virologia , Linhagem Celular , Mesocricetus , Filogenia , Regiões Promotoras Genéticas/genética , Carrapatos/virologia , Proteínas não Estruturais Virais/genética
15.
Cell Rep ; 25(13): 3750-3758.e4, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30590046

RESUMO

The Gn subcomponent of the Gn-Gc assembly that envelopes the human and animal pathogen, Rift Valley fever virus (RVFV), is a primary target of the neutralizing antibody response. To better understand the molecular basis for immune recognition, we raised a class of neutralizing monoclonal antibodies (nAbs) against RVFV Gn, which exhibited protective efficacy in a mouse infection model. Structural characterization revealed that these nAbs were directed to the membrane-distal domain of RVFV Gn and likely prevented virus entry into a host cell by blocking fusogenic rearrangements of the Gn-Gc lattice. Genome sequence analysis confirmed that this region of the RVFV Gn-Gc assembly was under selective pressure and constituted a site of vulnerability on the virion surface. These data provide a blueprint for the rational design of immunotherapeutics and vaccines capable of preventing RVFV infection and a model for understanding Ab-mediated neutralization of bunyaviruses more generally.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antivirais/farmacologia , Vírus da Febre do Vale do Rift/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/farmacologia , Chlorocebus aethiops , Feminino , Glicoproteínas/química , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Imunização , Imunoglobulina G/metabolismo , Camundongos Endogâmicos BALB C , Modelos Biológicos , Testes de Neutralização , Domínios Proteicos , Coelhos , Proteínas Recombinantes/farmacologia , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
16.
J Gen Virol ; 99(12): 1739-1745, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30394867

RESUMO

Many insect cell lines are persistently infected with insect-specific viruses (ISV) often unrecognized by the scientific community. Considering recent findings showing the possibility of interference between arbovirus and ISV infections, it is important to pay attention to ISV-infected cell lines. One example is the Entomobirnavirus, Culex Y virus (CYV). Here we describe the detection of CYV using a combination of small RNA sequencing, electron microscopy and PCR in mosquito cell lines Aag2, U4.4 and C7-10. We found CYV-specific small RNAs in all three cell lines. Interestingly, the magnitude of the detected viral RNA genome is variable among cell passages and leads to irregular detection via electron microscopy. Gaining insights into the presence of persistent ISV infection in commonly used mosquito cells and their interactions with the host immune system is beneficial for evaluating the outcome of co-infections with arboviruses of public health concern.


Assuntos
Birnaviridae/crescimento & desenvolvimento , Birnaviridae/isolamento & purificação , Culicidae/virologia , Pequeno RNA não Traduzido/análise , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Microscopia Eletrônica , Reação em Cadeia da Polimerase , Pequeno RNA não Traduzido/genética , Análise de Sequência de DNA
17.
PLoS Negl Trop Dis ; 12(10): e0006884, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30372452

RESUMO

Orthobunyaviruses such as Cache Valley virus (CVV) and Kairi virus (KRIV) are important animal pathogens. Periodic outbreaks of CVV have resulted in the significant loss of lambs on North American farms, whilst KRIV has mainly been detected in South and Central America with little overlap in geographical range. Vaccines or treatments for these viruses are unavailable. One approach to develop novel vaccine candidates is based on the use of reverse genetics to produce attenuated viruses that elicit immune responses but cannot revert to full virulence. The full genomes of both viruses were sequenced to obtain up to date genome sequence information. Following sequencing, minigenome systems and reverse genetics systems for both CVV and KRIV were developed. Both CVV and KRIV showed a wide in vitro cell host range, with BHK-21 cells a suitable host cell line for virus propagation and titration. To develop attenuated viruses, the open reading frames of the NSs proteins were disrupted. The recombinant viruses with no NSs protein expression induced the production of type I interferon (IFN), indicating that for both viruses NSs functions as an IFN antagonist and that such attenuated viruses could form the basis for attenuated viral vaccines. To assess the potential for reassortment between CVV and KRIV, which could be relevant during vaccination campaigns in areas of overlap, we attempted to produce M segment reassortants by reverse genetics. We were unable to obtain such viruses, suggesting that it is an unlikely event.


Assuntos
Infecções por Bunyaviridae/imunologia , Interações Hospedeiro-Patógeno , Orthobunyavirus/genética , Orthobunyavirus/imunologia , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Genética Reversa/métodos , Animais , Infecções por Bunyaviridae/virologia , Linhagem Celular , Técnicas de Inativação de Genes , Genoma Viral , Especificidade de Hospedeiro , Evasão da Resposta Imune , Imunidade Inata , Orthobunyavirus/crescimento & desenvolvimento , Vírus Reordenados/crescimento & desenvolvimento , Análise de Sequência de DNA , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/isolamento & purificação
18.
J Virol ; 92(13)2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29695422

RESUMO

Bunyaviruses pose a significant threat to human health, prosperity, and food security. In response to viral infections, interferons (IFNs) upregulate the expression of hundreds of interferon-stimulated genes (ISGs), whose cumulative action can potently inhibit the replication of bunyaviruses. We used a flow cytometry-based method to screen the ability of ∼500 unique ISGs from humans and rhesus macaques to inhibit the replication of Bunyamwera orthobunyavirus (BUNV), the prototype of both the Peribunyaviridae family and the Bunyavirales order. Candidates possessing antibunyaviral activity were further examined using a panel of divergent bunyaviruses. Interestingly, one candidate, ISG20, exhibited potent antibunyaviral activity against most viruses examined from the Peribunyaviridae, Hantaviridae, and Nairoviridae families, whereas phleboviruses (Phenuiviridae) largely escaped inhibition. Similar to the case against other viruses known to be targeted by ISG20, the antibunyaviral activity of ISG20 is dependent upon its functional RNase activity. Through use of an infectious virus-like particle (VLP) assay (based on the BUNV minigenome system), we confirmed that gene expression from all 3 viral segments is strongly inhibited by ISG20. Using in vitro evolution, we generated a substantially ISG20-resistant BUNV and mapped the determinants of ISG20 sensitivity/resistance. Taking all the data together, we report that ISG20 is a broad and potent antibunyaviral factor but that some bunyaviruses are remarkably ISG20 resistant. Thus, ISG20 sensitivity/resistance may influence the pathogenesis of bunyaviruses, many of which are emerging viruses of clinical or veterinary significance.IMPORTANCE There are hundreds of bunyaviruses, many of which cause life-threatening acute diseases in humans and livestock. The interferon (IFN) system is a key component of innate immunity, and type I IFNs limit bunyaviral propagation both in vitro and in vivo Type I IFN signaling results in the upregulation of hundreds of IFN-stimulated genes (ISGs), whose concerted action generates an "antiviral state." Although IFNs are critical in limiting bunyaviral replication and pathogenesis, much is still unknown about which ISGs inhibit bunyaviruses. Using ISG-expression screening, we examined the ability of ∼500 unique ISGs to inhibit Bunyamwera orthobunyavirus (BUNV), the prototypical bunyavirus. Using this approach, we identified ISG20, an interferon-stimulated exonuclease, as a potent inhibitor of BUNV. Interestingly, ISG20 possesses highly selective antibunyaviral activity, with multiple bunyaviruses being potently inhibited while some largely escape inhibition. We speculate that the ability of some bunyaviruses to escape ISG20 may influence their pathogenesis.


Assuntos
Antivirais/farmacologia , Vírus Bunyamwera/patogenicidade , Infecções por Bunyaviridae/prevenção & controle , Exonucleases/farmacologia , Genoma Viral , Interferons/metabolismo , Infecções por Bunyaviridae/metabolismo , Infecções por Bunyaviridae/virologia , Exonucleases/genética , Exorribonucleases , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos
19.
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
20.
Elife ; 62017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28915104

RESUMO

Rift Valley fever phlebovirus (RVFV) is a clinically and economically important pathogen increasingly likely to cause widespread epidemics. RVFV virulence depends on the interferon antagonist non-structural protein (NSs), which remains poorly characterized. We identified a stable core domain of RVFV NSs (residues 83-248), and solved its crystal structure, a novel all-helical fold organized into highly ordered fibrils. A hallmark of RVFV pathology is NSs filament formation in infected cell nuclei. Recombinant virus encoding the NSs core domain induced intranuclear filaments, suggesting it contains all essential determinants for nuclear translocation and filament formation. Mutations of key crystal fibril interface residues in viruses encoding full-length NSs completely abrogated intranuclear filament formation in infected cells. We propose the fibrillar arrangement of the NSs core domain in crystals reveals the molecular basis of assembly of this key virulence factor in cell nuclei. Our findings have important implications for fundamental understanding of RVFV virulence.


Assuntos
Vírus da Febre do Vale do Rift/química , Proteínas não Estruturais Virais/química , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Núcleo Celular/virologia , Cristalografia por Raios X , Análise Mutacional de DNA , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Vírus da Febre do Vale do Rift/genética , Proteínas não Estruturais Virais/genética , Fatores de Virulência/química , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...