Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
PLoS Biol ; 20(11): e3001870, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36378688

RESUMO

Bunyaviruses lack a specific mechanism to ensure the incorporation of a complete set of genome segments into each virion, explaining the generation of incomplete virus particles lacking one or more genome segments. Such incomplete virus particles, which may represent the majority of particles produced, are generally considered to interfere with virus infection and spread. Using the three-segmented arthropod-borne Rift Valley fever virus as a model bunyavirus, we here show that two distinct incomplete virus particle populations unable to spread autonomously are able to efficiently complement each other in both mammalian and insect cells following co-infection. We further show that complementing incomplete virus particles can co-infect mosquitoes, resulting in the reconstitution of infectious virus that is able to disseminate to the mosquito salivary glands. Computational models of infection dynamics predict that incomplete virus particles can positively impact virus spread over a wide range of conditions, with the strongest effect at intermediate multiplicities of infection. Our findings suggest that incomplete particles may play a significant role in within-host spread and between-host transmission, reminiscent of the infection cycle of multipartite viruses.


Assuntos
Arbovírus , Culicidae , Orthobunyavirus , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Viroses , Animais , Humanos , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/genética , Febre do Vale de Rift/metabolismo , Vírion/metabolismo , Mamíferos
2.
J Virol ; 97(6): e0041523, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37306574

RESUMO

Rift Valley fever virus (RVFV) (family Phenuiviridae) can cause severe disease, and outbreaks of this mosquito-borne pathogen pose a significant threat to public and animal health. Yet many molecular aspects of RVFV pathogenesis remain incompletely understood. Natural RVFV infections are acute, characterized by a rapid onset of peak viremia during the first days post-infection, followed by a rapid decline. Although in vitro studies identified a major role of interferon (IFN) responses in counteracting the infection, a comprehensive overview of the specific host factors that play a role in RVFV pathogenesis in vivo is still lacking. Here, the host in vivo transcriptional profiles in the liver and spleen tissues of lambs exposed to RVFV are studied using RNA sequencing (RNA-seq) technology. We validate that IFN-mediated pathways are robustly activated in response to infection. We also link the observed hepatocellular necrosis with severely compromised organ function, which is reflected as a marked downregulation of multiple metabolic enzymes essential for homeostasis. Furthermore, we associate the elevated basal expression of LRP1 in the liver with RVFV tissue tropism. Collectively, the results of this study deepen the knowledge of the in vivo host response during RVFV infection and reveal new insights into the gene regulation networks underlying pathogenesis in a natural host. IMPORTANCE Rift Valley fever virus (RVFV) is a mosquito-transmitted pathogen capable of causing severe disease in animals and humans. Outbreaks of RVFV pose a significant threat to public health and can result in substantial economic losses. Little is known about the molecular basis of RVFV pathogenesis in vivo, particularly in its natural hosts. We employed RNA-seq technology to investigate genome-wide host responses in the liver and spleen of lambs during acute RVFV infection. We show that RVFV infection drastically decreases the expression of metabolic enzymes, which impairs normal liver function. Moreover, we highlight that basal expression levels of the host factor LRP1 may be a determinant of RVFV tissue tropism. This study links the typical pathological phenotype induced by RVFV infection with tissue-specific gene expression profiles, thereby improving our understanding of RVFV pathogenesis.


Assuntos
Homeostase , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Febre do Vale de Rift/patologia , Vírus da Febre do Vale do Rift/patogenicidade , Ovinos , Transcriptoma , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Fígado , Interações Hospedeiro-Patógeno , Interferons/metabolismo
3.
Biologicals ; 74: 10-15, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34736782

RESUMO

The Zoonoses Anticipation and Preparedness Initiative (ZAPI) was set up to prepare for future outbreaks and to develop and implement new technologies to accelerate development and manufacturing of vaccines and monoclonal antibodies. To be able to achieve surge capacity, an easy deployment and production at multiple sites is needed. This requires a straightforward manufacturing system with a limited number of steps in upstream and downstream processes, a minimum number of in vitro Quality Control assays, and robust and consistent platforms. Three viruses were selected as prototypes: Middle East Respiratory Syndrome (MERS) coronavirus, Rift Valley fever virus, and Schmallenberg virus. Selected antibodies against the viral surface antigens were manufactured by transient gene expression in Chinese Hamster Ovary (CHO) cells, scaling up to 200 L. For vaccine production, viral antigens were fused to multimeric protein scaffold particles using the SpyCatcher/SpyTag system. In vivo models demonstrated the efficacy of both antibodies and vaccines. The final step in speeding up vaccine (and antibody) development is the regulatory appraisal of new platform technologies. Towards this end, within ZAPI, a Platform Master File (PfMF) was developed, as part of a licensing dossier, to facilitate and accelerate the scientific assessment by avoiding repeated discussion of already accepted platforms. The veterinary PfMF was accepted, whereas the human PfMF is currently under review by the European Medicines Agency, aiming for publication of the guideline by January 2022.


Assuntos
Infecções por Coronavirus , Vacinas Virais , Zoonoses , Animais , Anticorpos Antivirais , Antígenos Virais , Células CHO , Congressos como Assunto , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , Cricetinae , Cricetulus , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio , Vírus da Febre do Vale do Rift , Zoonoses/prevenção & controle
4.
BMC Vet Res ; 16(1): 51, 2020 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-32046722

RESUMO

BACKGROUND: Porcine teschovirus (PTV) circulates among wild and domesticated pig populations without causing clinical disease, however neuroinvasive strains have caused high morbidity and mortality in the past. In recent years, several reports appeared with viral agents as a cause for neurologic signs in weanling and growing pigs among which PTV and new strains of PTV were described. CASE PRESENTATION: On two unrelated pig farms in the Netherlands the weanling pig population showed a staggering gate, which developed progressively to paresis or paralysis of the hind legs with a morbidity up to 5%. After necropsy we diagnosed a non-suppurative encephalomyelitis on both farms, which was most consistent with a viral infection. PTV was detected within the central nervous system by qPCR. From both farms PTV full-length genomes were sequenced, which clustered closely with PTV-3 (98%) or PTV-11 (85%). Other common swine viruses were excluded by qPCR and sequencing of the virus. CONCLUSION: Our results show that new neuroinvasive PTV strains still emerge in pigs in the Netherlands. Further research is needed to investigate the impact of PTV and other viral agents causing encephalomyelitis within wild and domestic pig populations supported by the awareness of veterinarians.


Assuntos
Encefalomielite/veterinária , Infecções por Picornaviridae/veterinária , Doenças dos Suínos/virologia , Teschovirus/classificação , Animais , Encefalomielite/epidemiologia , Encefalomielite/virologia , Países Baixos/epidemiologia , Filogenia , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/virologia , Suínos , Doenças dos Suínos/epidemiologia , Teschovirus/genética , Teschovirus/isolamento & purificação
5.
Biologicals ; 65: 46-49, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32209300

RESUMO

On the 17th of October 2019, a workshop was held at Wageningen Bioveterinary Research in Lelystad, the Netherlands, to discuss the obstacles to vaccination in the veterinary field. Participants from academia, OIE, FAO, EC, EMA, USDA, national regulatory and veterinary health authorities, and the animal health industry discussed how availability and access to animal vaccines can be improved not just in the EU and US but also in Low to Middle Income Countries (LMIC) across the world and agreed that this requires innovations in both the scientific and the regulatory field. The workshop called for engaging all stakeholders to improve regulatory acceptance of novel vaccine technologies and encourage their registration. There is a need for better mutual understanding between academia, industry and regulators, and more openness to discuss framework, requirements, and product authorisations, and to converge the regulatory rules between regions. The next leap forward could be a broader application of novel technologies using RNA- or DNA-based vaccine platforms, where the "backbone" is maintained, while the gene of interest coding for an immunogenic protein can be exchanged in a standardised manner. This approach enables rapid response in outbreak situations and should lower the risk and cost of vaccine development.


Assuntos
Doenças Transmissíveis Emergentes/prevenção & controle , Doenças Transmissíveis Emergentes/veterinária , Vacinas de DNA/uso terapêutico , Vacinas Sintéticas/uso terapêutico , Criação de Animais Domésticos , Animais , Animais Domésticos , Animais Selvagens , Desenvolvimento de Medicamentos , Participação dos Interessados , Vacinas de DNA/economia , Vacinas Sintéticas/economia , Drogas Veterinárias , Vacinas de mRNA
6.
J Gen Virol ; 100(8): 1187-1199, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31310198

RESUMO

Rift Valley fever (RVF) is a mosquito-borne viral zoonosis that was first discovered in Kenya in 1930 and is now endemic throughout multiple African countries and the Arabian Peninsula. RVF virus primarily infects domestic livestock (sheep, goats, cattle) causing high rates of neonatal mortality and abortion, with human infection resulting in a wide variety of clinical outcomes, ranging from self-limiting febrile illness to life-threatening haemorrhagic diatheses, and miscarriage in pregnant women. Since its discovery, RVF has caused many outbreaks in Africa and the Arabian Peninsula with major impacts on human and animal health. However, options for the control of RVF outbreaks are limited by the lack of licensed human vaccines or therapeutics. For this reason, RVF is prioritized by the World Health Organization for urgent research and development of countermeasures for the prevention and control of future outbreaks. In this review, we highlight the current understanding of RVF, including its epidemiology, pathogenesis, clinical manifestations and status of vaccine development.


Assuntos
Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/fisiologia , Animais , Humanos , Febre do Vale de Rift/epidemiologia , Febre do Vale de Rift/transmissão , Vírus da Febre do Vale do Rift/classificação , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/isolamento & purificação , Zoonoses/epidemiologia , Zoonoses/transmissão , Zoonoses/virologia
7.
PLoS Pathog ; 12(8): e1005800, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27548280

RESUMO

The bunyavirus genome comprises a small (S), medium (M), and large (L) RNA segment of negative polarity. Although genome segmentation confers evolutionary advantages by enabling genome reassortment events with related viruses, genome segmentation also complicates genome replication and packaging. Accumulating evidence suggests that genomes of viruses with eight or more genome segments are incorporated into virions by highly selective processes. Remarkably, little is known about the genome packaging process of the tri-segmented bunyaviruses. Here, we evaluated, by single-molecule RNA fluorescence in situ hybridization (FISH), the intracellular spatio-temporal distribution and replication kinetics of the Rift Valley fever virus (RVFV) genome and determined the segment composition of mature virions. The results reveal that the RVFV genome segments start to replicate near the site of infection before spreading and replicating throughout the cytoplasm followed by translocation to the virion assembly site at the Golgi network. Despite the average intracellular S, M and L genome segments approached a 1:1:1 ratio, major differences in genome segment ratios were observed among cells. We also observed a significant amount of cells lacking evidence of M-segment replication. Analysis of two-segmented replicons and four-segmented viruses subsequently confirmed the previous notion that Golgi recruitment is mediated by the Gn glycoprotein. The absence of colocalization of the different segments in the cytoplasm and the successful rescue of a tri-segmented variant with a codon shuffled M-segment suggested that inter-segment interactions are unlikely to drive the copackaging of the different segments into a single virion. The latter was confirmed by direct visualization of RNPs inside mature virions which showed that the majority of virions lack one or more genome segments. Altogether, this study suggests that RVFV genome packaging is a non-selective process.


Assuntos
Genoma Viral/fisiologia , Vírus da Febre do Vale do Rift/fisiologia , Montagem de Vírus/fisiologia , Linhagem Celular , Humanos , Hibridização in Situ Fluorescente , Febre do Vale de Rift/metabolismo
8.
Acta Neuropathol ; 131(2): 159-184, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26659576

RESUMO

A wide range of viruses from different virus families in different geographical areas, may cause immediate or delayed neuropathological changes and neurological manifestations in humans and animals. Infection by neurotropic viruses as well as the resulting immune response can irreversibly disrupt the complex structural and functional architecture of the central nervous system, frequently leaving the patient or affected animal with a poor or fatal prognosis. Mechanisms that govern neuropathogenesis and immunopathogenesis of viral infections are highlighted, using examples of well-studied virus infections that are associated with these alterations in different populations throughout the world. A better understanding of the molecular, epidemiological and biological characteristics of these infections and in particular of mechanisms that underlie their clinical manifestations may be expected to provide tools for the development of more effective intervention strategies and treatment regimens.


Assuntos
Viroses do Sistema Nervoso Central/patologia , Viroses do Sistema Nervoso Central/fisiopatologia , Animais , Humanos
9.
J Virol ; 88(18): 10883-93, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25008937

RESUMO

UNLABELLED: Bunyavirus genomes comprise a small (S), a medium (M), and a large (L) RNA segment of negative polarity. Although the untranslated regions have been shown to comprise signals required for transcription, replication, and encapsidation, the mechanisms that drive the packaging of at least one S, M, and L segment into a single virion to generate infectious virus are largely unknown. One of the most important members of the Bunyaviridae family that causes devastating disease in ruminants and occasionally humans is the Rift Valley fever virus (RVFV). We studied the flexibility of RVFV genome packaging by splitting the glycoprotein precursor gene, encoding the (NSm)GnGc polyprotein, into two individual genes encoding either (NSm)Gn or Gc. Using reverse genetics, six viruses with a segmented glycoprotein precursor gene were rescued, varying from a virus comprising two S-type segments in the absence of an M-type segment to a virus consisting of four segments (RVFV-4s), of which three are M-type. Despite that all virus variants were able to grow in mammalian cell lines, they were unable to spread efficiently in cells of mosquito origin. Moreover, in vivo studies demonstrated that RVFV-4s is unable to cause disseminated infection and disease in mice, even in the presence of the main virulence factor NSs, but induced a protective immune response against a lethal challenge with wild-type virus. In summary, splitting bunyavirus glycoprotein precursor genes provides new opportunities to study bunyavirus genome packaging and offers new methods to develop next-generation live-attenuated bunyavirus vaccines. IMPORTANCE: Rift Valley fever virus (RVFV) causes devastating disease in ruminants and occasionally humans. Virions capable of productive infection comprise at least one copy of the small (S), medium (M), and large (L) RNA genome segments. The M segment encodes a glycoprotein precursor (GPC) protein that is cotranslationally cleaved into Gn and Gc, which are required for virus entry and fusion. We studied the flexibility of RVFV genome packaging and developed experimental live-attenuated vaccines by applying a unique strategy based on the splitting of the GnGc open reading frame. Several RVFV variants, varying from viruses comprising two S-type segments to viruses consisting of four segments (RVFV-4s), of which three are M-type, could be rescued and were shown to induce a rapid protective immune response. Altogether, the segmentation of bunyavirus GPCs provides a new method for studying bunyavirus genome packaging and facilitates the development of novel live-attenuated bunyavirus vaccines.


Assuntos
Genoma Viral , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/genética , Vírion/fisiologia , Montagem de Vírus , Animais , Culicidae/virologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , RNA Viral/genética , Vírus da Febre do Vale do Rift/fisiologia , Vírion/genética
10.
Emerg Infect Dis ; 20(8): 1355-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25061813

RESUMO

In June 2013, a subclinical infection with severe fever with thrombocytopenia syndrome virus (SFTSV) was detected in Zhejiang Province, China, prompting seroprevalence studies in 6 districts within the province. Of 986 healthy persons tested, 71 had IgG antibodies against SFTSV. This finding suggests that most natural infections with SFTSV are mild or subclinical.


Assuntos
Anticorpos Antivirais/imunologia , Voluntários Saudáveis , Phlebovirus/imunologia , Vigilância em Saúde Pública , Anticorpos Antivirais/sangue , China/epidemiologia , Evolução Fatal , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Masculino , Pessoa de Meia-Idade , Febre por Flebótomos/virologia , Estudos Soroepidemiológicos
11.
J Gen Virol ; 95(Pt 12): 2638-2648, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25209808

RESUMO

Replicon-particle-based vaccines combine the efficacy of live-attenuated vaccines with the safety of inactivated or subunit vaccines. Recently, we developed Rift Valley fever virus (RVFV) replicon particles, also known as nonspreading RVFV (NSR), and demonstrated that a single vaccination with these particles can confer sterile immunity in target animals. NSR particles can be produced by transfection of replicon cells, which stably maintain replicating RVFV S and L genome segments, with an expression plasmid encoding the RVFV glycoproteins, Gn and Gc, normally encoded by the M-genome segment. Here, we explored the possibility to produce NSR with the use of a helper virus. We show that replicon cells infected with a Newcastle disease virus expressing Gn and Gc (NDV-GnGc) were able to produce high levels of NSR particles. In addition, using reverse genetics and site-directed mutagenesis, we were able to create an NDV-GnGc variant that lacks the NDV fusion protein and contains two amino acid substitutions in, respectively, Gn and HN. The resulting virus uses a unique entry pathway that facilitates the efficient production of NSR in a one-component system. The novel system provides a promising alternative for transfection-based NSR production.


Assuntos
Vírus da Doença de Newcastle/imunologia , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/fisiologia , Vacinas Virais/imunologia , Replicação Viral , Animais , Linhagem Celular , Cricetinae , Feminino , Camundongos , Camundongos Endogâmicos BALB C
12.
Arch Virol ; 159(11): 2883-92, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24957652

RESUMO

Shuni virus (SHUV), a member of the genus Orthobunyavirus, has in a recent study been associated with neurological disease in horses in South Africa. After its first isolation in 1966 from an asymptomatic bovine, very little attention was given to the genetic characterisation of SHUV. The association of SHUV with severe neurological disease in several horses in South Africa prompted us to determine the full genome sequence of a horse neurovirulent isolate to compare it to other members of the genus Orthobunyavirus, as well as the partially sequenced genome of the prototype SHUV strain. The availability of a full genome sequence will facilitate the development of a reverse genetics system to study SHUV molecular biology and pathogenesis.


Assuntos
Infecções por Bunyaviridae/veterinária , Genoma Viral , Doenças dos Cavalos/virologia , Orthobunyavirus/classificação , Orthobunyavirus/genética , Filogenia , Sequência de Aminoácidos , Animais , Infecções por Bunyaviridae/virologia , Bovinos , Doenças dos Bovinos/virologia , Genômica , Cavalos , Dados de Sequência Molecular , Orthobunyavirus/isolamento & purificação , Alinhamento de Sequência , África do Sul , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
13.
Emerg Microbes Infect ; 13(1): 2373313, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38946528

RESUMO

Rift Valley fever (RVF) is a mosquito-borne zoonotic disease caused by RVF virus (RVFV). RVFV infections in humans are usually asymptomatic or associated with mild febrile illness, although more severe cases of haemorrhagic disease and encephalitis with high mortality also occur. Currently, there are no licensed human vaccines available. The safety and efficacy of a genetically engineered four-segmented RVFV variant (hRVFV-4s) as a potential live-attenuated human vaccine has been tested successfully in mice, ruminants, and marmosets though the correlates of protection of this vaccine are still largely unknown. In the present study, we have assessed hRVFV-4s-induced humoral and cellular immunity in a mouse model of RVFV infection. Our results confirm that a single dose of hRVFV-4s is highly efficient in protecting naïve mice from developing severe disease following intraperitoneal challenge with a highly virulent RVFV strain and data show that virus neutralizing (VN) serum antibody titres in a prime-boost regimen are significantly higher compared to the single dose. Subsequently, VN antibodies from prime-boost-vaccinated recipients were shown to be protective when transferred to naïve mice. In addition, hRVFV-4s vaccination induced a significant virus-specific T cell response as shown by IFN-γ ELISpot assay, though these T cells did not provide significant protection upon passive transfer to naïve recipient mice. Collectively, this study highlights hRVFV-4s-induced VN antibodies as a major correlate of protection against lethal RVFV infection.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Atenuadas , Vacinas Virais , Animais , Vírus da Febre do Vale do Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Camundongos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Feminino , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Modelos Animais de Doenças , Imunidade Celular , Linfócitos T/imunologia , Imunidade Humoral , Camundongos Endogâmicos BALB C , Interferon gama/imunologia , Vacinação
14.
Virus Genes ; 47(3): 505-14, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23996608

RESUMO

A novel orthobunyavirus, named "Schmallenberg virus" (SBV), was first detected in the blood of cattle at the end of the summer in Germany in 2011, and subsequently in late autumn from the brain of a stillborn malformed lamb in The Netherlands. Full genome sequences, including 5' and 3' terminal "panhandle" sequences of the L, M, and S segments of the SBV isolated from lamb brain tissue (named HL1) were determined. In addition, a second SBV strain was isolated from the blood of a dairy cow (named F6) also in The Netherlands. This isolate was passaged on Vero cells, and its genome sequence was determined by next-generation sequencing. Alignments of the two genome sequences revealed 4, 12, and 2 amino acid differences in the open reading frames of the L, M, and S segments, respectively. Eleven of a total of 12 amino acid differences were detected in the M segment encoding the ectodomain of the putative structural glycoprotein Gc. Notably, in the HL1 isolate, positions 737-739 are occupied by isoleucine, arginine, and leucine (IRL), whereas in the majority of other sequenced SBV isolates these positions are occupied by threonine, histidine, and proline, respectively. Moreover, in all sheep, goat, and cattle SBV isolates sequenced and published so far, an IRL sequence was never found. This has brought us to the conclusion that the M segment of the HL1 isolate differed markedly from that of other lamb and cow isolates. Whether this atypical variant resulted from adaptation to the ewe, fetus, or insect vector remains to be investigated.


Assuntos
Encéfalo/virologia , Infecções por Bunyaviridae/veterinária , Orthobunyavirus/genética , Orthobunyavirus/isolamento & purificação , Doenças dos Ovinos/virologia , Sequência de Aminoácidos , Animais , Infecções por Bunyaviridae/virologia , Bovinos , Dados de Sequência Molecular , Orthobunyavirus/química , Orthobunyavirus/classificação , Alinhamento de Sequência , Ovinos , Proteínas Virais/química , Proteínas Virais/genética
15.
Vaccines (Basel) ; 11(3)2023 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-36992291

RESUMO

Live-attenuated Rift Valley fever (RVF) vaccines transiently replicate in the vaccinated host, thereby effectively initiating an innate and adaptive immune response. Rift Valley fever virus (RVFV)-specific neutralizing antibodies are considered the main correlate of protection. Vaccination with classical live-attenuated RVF vaccines during gestation in livestock has been associated with fetal malformations, stillbirths, and fetal demise. Facilitated by an increased understanding of the RVFV infection and replication cycle and availability of reverse genetics systems, novel rationally-designed live-attenuated candidate RVF vaccines with improved safety profiles have been developed. Several of these experimental vaccines are currently advancing beyond the proof-of-concept phase and are being evaluated for application in both animals and humans. We here provide perspectives on some of these next-generation live-attenuated RVF vaccines and highlight the opportunities and challenges of these approaches to improve global health.

16.
Front Cell Infect Microbiol ; 13: 1206089, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38170150

RESUMO

Rift Valley fever virus (RVFV) is a (re)emerging mosquito-borne pathogen impacting human and animal health. How RVFV spreads through a population depends on population-level and individual-level interactions between vector, host and pathogen. Here, we estimated the probability for RVFV to transmit to naive animals by experimentally exposing lambs to a bite of an infectious mosquito, and assessed if and how RVFV infection subsequently developed in the exposed animal. Aedes aegypti mosquitoes, previously infected via feeding on a viremic lamb, were used to expose naive lambs to the virus. Aedes aegypti colony mosquitoes were used as they are easy to maintain and readily feed in captivity. Other mosquito spp. could be examined with similar methodology. Lambs were exposed to either 1-3 (low exposure) or 7-9 (high exposure) infectious mosquitoes. All lambs in the high exposure group became viremic and showed characteristic signs of Rift Valley fever within 2-4 days post exposure. In contrast, 3 out of 12 lambs in the low exposure group developed viremia and disease, with similar peak-levels of viremia as the high exposure group but with some heterogeneity in the onset of viremia. These results suggest that the likelihood for successful infection of a ruminant host is affected by the number of infectious mosquitoes biting, but also highlights that a single bite of an infectious mosquito can result in disease. The per bite mosquito-to-host transmission efficiency was estimated at 28% (95% confidence interval: 15 - 47%). We subsequently combined this transmission efficiency with estimates for life traits of Aedes aegypti or related mosquitoes into a Ross-McDonald mathematical model to illustrate scenarios under which major RVFV outbreaks could occur in naïve populations (i.e., R0 >1). The model revealed that relatively high vector-to-host ratios as well as mosquitoes feeding preferably on competent hosts are required for R0 to exceed 1. Altogether, this study highlights the importance of experiments that mimic natural exposure to RVFV. The experiments facilitate a better understanding of the natural progression of disease and a direct way to obtain epidemiological parameters for mathematical models.


Assuntos
Aedes , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Mosquitos Vetores , Febre do Vale de Rift/epidemiologia , Ruminantes , Ovinos , Viremia/veterinária
17.
Sci Rep ; 13(1): 21627, 2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-38062065

RESUMO

The distribution of tick-borne encephalitis virus (TBEV) is expanding to Western European countries, including the Netherlands, but the contribution of different rodent species to the transmission of TBEV is poorly understood. We investigated whether two species of wild rodents native to the Netherlands, the wood mouse Apodemus sylvaticus and the yellow-necked mouse Apodemus flavicollis, differ in their relative susceptibility to experimental infection with TBEV. Wild-caught individuals were inoculated subcutaneously with the classical European subtype of TBEV (Neudoerfl) or with TBEV-NL, a genetically divergent TBEV strain from the Netherlands. Mice were euthanised and necropsied between 3 and 21 days post-inoculation. None of the mice showed clinical signs or died during the experimental period. Nevertheless, TBEV RNA was detected up to 21 days in the blood of both mouse species and TBEV was also isolated from the brain of some mice. Moreover, no differences in infection rates between virus strains and mouse species were found in blood, spleen, or liver samples. Our results suggest that the wood mouse and the yellow-necked mouse may equally contribute to the transmission cycle of TBEV in the Netherlands. Future experimental infection studies that include feeding ticks will help elucidate the relative importance of viraemic transmission in the epidemiology of TBEV.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Encefalite Transmitida por Carrapatos , Carrapatos , Animais , Camundongos , Vírus da Encefalite Transmitidos por Carrapatos/genética , Encefalite Transmitida por Carrapatos/epidemiologia , Encefalite Transmitida por Carrapatos/veterinária , Murinae , Países Baixos
18.
J Virol ; 85(23): 12622-30, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21957302

RESUMO

Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic bunyavirus of the genus Phlebovirus and a serious human and veterinary pathogen. RVFV contains a three-segmented RNA genome, which is comprised of the large (L), medium (M), and small (S) segments. The proteins that are essential for genome replication are encoded by the L and S segments, whereas the structural glycoproteins are encoded by the M segment. We have produced BHK replicon cell lines (BHK-Rep) that maintain replicating L and S genome segments. Transfection of BHK-Rep cells with a plasmid encoding the structural glycoproteins results in the efficient production of RVFV replicon particles (RRPs). To facilitate monitoring of infection, the NSs gene was replaced with an enhanced green fluorescent protein gene. RRPs are infectious for both mammalian and insect cells but are incapable of autonomous spreading, rendering their application outside biosafety containment completely safe. We demonstrate that a single intramuscular vaccination with RRPs protects mice from a lethal dose of RVFV and show that RRPs can be used for rapid virus neutralization tests that do not require biocontainment facilities. The methods reported here will greatly facilitate vaccine and drug development as well as fundamental studies on RVFV biology. Moreover, it may be possible to develop similar systems for other members of the bunyavirus family as well.


Assuntos
Genoma Viral , Proteínas de Fluorescência Verde/metabolismo , Replicon/genética , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/patogenicidade , Replicação Viral , Animais , Northern Blotting , Cricetinae , Ensaio de Imunoadsorção Enzimática , Feminino , Engenharia Genética , Proteínas de Fluorescência Verde/genética , Injeções Intramusculares , Rim/citologia , Rim/metabolismo , Rim/virologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos , Recombinação Genética , Febre do Vale de Rift/genética , Taxa de Sobrevida , Vacinação , Proteínas não Estruturais Virais/metabolismo , Internalização do Vírus
19.
Methods Mol Biol ; 2446: 313-326, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35157280

RESUMO

Multimerization of single-domain antibodies (sdAbs) is instrumental for construction of antibody molecules with high avidity, extended in vivo half-life, and tailor-made biological activity. Two-component superglues, based on bacterium-derived peptides (Tags) and small protein domains (Catchers) that form isopeptide bonds when in close proximity, enable the creation of multimers by simply mixing of the individual components. Here, we provide detailed methods for the construction of sdAbs and scaffolds bearing genetically fused superglue components and their assembly into multimeric complexes.


Assuntos
Anticorpos de Domínio Único , Bactérias/genética , Peptídeos/química , Domínios Proteicos , Anticorpos de Domínio Único/genética
20.
NPJ Vaccines ; 7(1): 54, 2022 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-35585071

RESUMO

Rift Valley fever virus (RVFV) is an emerging mosquito-borne bunyavirus that is highly pathogenic to wild and domesticated ruminants, camelids, and humans. While animals are exclusively infected via mosquito bites, humans can also be infected via contact with contaminated tissues or blood. No human vaccine is available and commercialized veterinary vaccines do not optimally combine efficacy with safety. We previously reported the development of two novel live-attenuated RVF vaccines, created by splitting the M genome segment and deleting the major virulence determinant NSs. The vaccine candidates, referred to as the veterinary vaccine vRVFV-4s and the human vaccine hRVFV-4s, were shown to induce protective immunity in multiple species after a single vaccination. Anticipating accidental exposure of humans to the veterinary vaccine and the application of hRVFV-4s to humans, the safety of each vaccine was evaluated in the most susceptible nonhuman primate model, the common marmoset (Callithrix jacchus). Marmosets were inoculated with high doses of each vaccine and were monitored for clinical signs as well as for vaccine virus dissemination, shedding, and spreading to the environment. To accurately assess the attenuation of both vaccine viruses, separate groups of marmosets were inoculated with the parent wild-type RVFV strains. Both wild-type strains induced high viremia and disseminated to primary target organs, associated with mild-to-severe morbidity. In contrast, both vaccines were well tolerated with no evidence of dissemination and shedding while inducing potent neutralizing antibody responses. The results of the studies support the unprecedented safety profile of both vaccines for animals and humans.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa