Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 18(4): e3000673, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32343693

RESUMO

The mechanisms underlying virus emergence are rarely well understood, making the appearance of outbreaks largely unpredictable. Bluetongue virus serotype 8 (BTV-8), an arthropod-borne virus of ruminants, emerged in livestock in northern Europe in 2006, spreading to most European countries by 2009 and causing losses of billions of euros. Although the outbreak was successfully controlled through vaccination by early 2010, puzzlingly, a closely related BTV-8 strain re-emerged in France in 2015, triggering a second outbreak that is still ongoing. The origin of this virus and the mechanisms underlying its re-emergence are unknown. Here, we performed phylogenetic analyses of 164 whole BTV-8 genomes sampled throughout the two outbreaks. We demonstrate consistent clock-like virus evolution during both epizootics but found negligible evolutionary change between them. We estimate that the ancestor of the second outbreak dates from the height of the first outbreak in 2008. This implies that the virus had not been replicating for multiple years prior to its re-emergence in 2015. Given the absence of any known natural mechanism that could explain BTV-8 persistence over this long period without replication, we hypothesise that the second outbreak could have been initiated by accidental exposure of livestock to frozen material contaminated with virus from approximately 2008. Our work highlights new targets for pathogen surveillance programmes in livestock and illustrates the power of genomic epidemiology to identify pathways of infectious disease emergence.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/virologia , Genoma Viral , Animais , Evolução Biológica , Bluetongue/epidemiologia , Vírus Bluetongue/genética , Surtos de Doenças , Europa (Continente)/epidemiologia , França , Gado/virologia , Mutação , Filogenia
2.
Int J Mol Sci ; 24(7)2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37047816

RESUMO

Bioinformatic analyses have predicted that orbiviruses encode an additional, small non-structural protein (NS5) from a secondary open reading frame on genome segment 10. However, this protein has not previously been detected in infected mammalian or insect cells. NS5-specific antibodies were generated in mice and were used to identify NS5 synthesised in orbivirus-infected BSR cells or cells transfected with NS5 expression plasmids. Confocal microscopy shows that although NS5 accumulates in the nucleus, particularly in the nucleolus, which becomes disrupted, it also appears in the cell cytoplasm, co-localising with mitochondria. NS5 helps to prevent the degradation of ribosomal RNAs during infection and reduces host-cell protein synthesis However, it helps to extend cell viability by supporting viral protein synthesis and virus replication. Pulldown studies showed that NS5 binds to ssRNAs and supercoiled DNAs and demonstrates interactions with ZBP1, suggesting that it modulates host-cell responses.


Assuntos
Orbivirus , Animais , Camundongos , Núcleo Celular/metabolismo , DNA , Orbivirus/genética , Orbivirus/metabolismo , RNA Viral/genética , Proteínas de Ligação a RNA , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
3.
J Gen Virol ; 103(11)2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36394457

RESUMO

Spinareoviridae is a large family of icosahedral viruses that are usually regarded as non-enveloped with segmented (9-12 linear segments) dsRNA genomes of 23-29 kbp. Spinareovirids have a broad host range, infecting animals, fungi and plants. Some have important pathogenic potential for humans (e.g. Colorado tick fever virus), livestock (e.g. avian orthoreoviruses), fish (e.g. aquareoviruses) and plants (e.g. rice ragged stunt virus and rice black streaked dwarf virus). This is a summary of the ICTV Report on the family Spinareoviridae, which is available at ictv.global/report/spinareoviridae.


Assuntos
Fungos , RNA de Cadeia Dupla , Animais , Humanos , Plantas , Especificidade de Hospedeiro , Filogenia
4.
J Gen Virol ; 103(10)2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36215107

RESUMO

Sedoreoviridae is a large family of icosahedral viruses that are usually regarded as non-enveloped with segmented (10-12 linear segments) dsRNA genomes of 18-26 kbp. Sedoreovirids have a broad host range, infecting mammals, birds, crustaceans, arthropods, algae and plants. Some of them have important pathogenic potential for humans (e.g. rotavirus A), livestock (e.g. bluetongue virus) and plants (e.g. rice dwarf virus). This is a summary of the ICTV Report on the family Sedoreoviridae, which is available at ictv.global/report/sedoreoviridae.


Assuntos
Mamíferos , RNA de Cadeia Dupla , Animais , Aves , Genoma Viral , Humanos , Plantas , Vírion , Replicação Viral
5.
J Gen Virol ; 100(4): 568-582, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30843784

RESUMO

Bluetongue virus (BTV) causes an economically important disease in domestic and wildlife ruminants and is transmitted by Culicoides biting midges. In ruminants, BTV has a wide cell tropism that includes endothelial cells of vascular and lymphatic vessels as important cell targets for virus replication, and several cell types of the immune system including monocytes, macrophages and dendritic cells. Thus, cell-entry represents a particular challenge for BTV as it infects many different cell types in widely diverse vertebrate and invertebrate hosts. Improved understanding of BTV cell-entry could lead to novel antiviral approaches that can block virus transmission from cell to cell between its invertebrate and vertebrate hosts. Here, we have investigated BTV cell-entry using endothelial cells derived from the natural bovine host (BFA cells) and purified whole virus particles of a low-passage, insect-cell isolate of a virulent strain of BTV-1. Our results show that the main entry pathway for infection of BFA cells is dependent on actin and dynamin, and shares certain characteristics with macropinocytosis. The ability to use a macropinocytosis-like entry route could explain the diverse cell tropism of BTV and contribute to the efficiency of transmission between vertebrate and invertebrate hosts.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/virologia , Doenças dos Bovinos/virologia , Insetos/virologia , Pinocitose , Internalização do Vírus , Actinas/genética , Actinas/metabolismo , Animais , Bluetongue/genética , Bluetongue/metabolismo , Bluetongue/fisiopatologia , Vírus Bluetongue/genética , Vírus Bluetongue/crescimento & desenvolvimento , Bovinos , Doenças dos Bovinos/genética , Doenças dos Bovinos/metabolismo , Doenças dos Bovinos/fisiopatologia , Células Cultivadas , Dinaminas/genética , Dinaminas/metabolismo , Células Endoteliais/virologia , Inoculações Seriadas , Ovinos , Doenças dos Ovinos/virologia , Replicação Viral
6.
PLoS Comput Biol ; 12(7): e1004901, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27384712

RESUMO

Infectious disease surveillance is key to limiting the consequences from infectious pathogens and maintaining animal and public health. Following the detection of a disease outbreak, a response in proportion to the severity of the outbreak is required. It is thus critical to obtain accurate information concerning the origin of the outbreak and its forward trajectory. However, there is often a lack of situational awareness that may lead to over- or under-reaction. There is a widening range of tests available for detecting pathogens, with typically different temporal characteristics, e.g. in terms of when peak test response occurs relative to time of exposure. We have developed a statistical framework that combines response level data from multiple diagnostic tests and is able to 'hindcast' (infer the historical trend of) an infectious disease epidemic. Assuming diagnostic test data from a cross-sectional sample of individuals infected with a pathogen during an outbreak, we use a Bayesian Markov Chain Monte Carlo (MCMC) approach to estimate time of exposure, and the overall epidemic trend in the population prior to the time of sampling. We evaluate the performance of this statistical framework on simulated data from epidemic trend curves and show that we can recover the parameter values of those trends. We also apply the framework to epidemic trend curves taken from two historical outbreaks: a bluetongue outbreak in cattle, and a whooping cough outbreak in humans. Together, these results show that hindcasting can estimate the time since infection for individuals and provide accurate estimates of epidemic trends, and can be used to distinguish whether an outbreak is increasing or past its peak. We conclude that if temporal characteristics of diagnostics are known, it is possible to recover epidemic trends of both human and animal pathogens from cross-sectional data collected at a single point in time.


Assuntos
Biologia Computacional/métodos , Epidemias/estatística & dados numéricos , Modelos Estatísticos , Vigilância da População/métodos , Algoritmos , Animais , Bluetongue , Bovinos , Doenças dos Bovinos , Estudos Transversais , Epidemias/prevenção & controle , Humanos , Coqueluche
7.
J Virol ; 86(18): 10255-6, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22923810

RESUMO

The entire genome of the reference strain of bluetongue virus (BTV) serotype 16 (strain RSArrrr/16) was sequenced (a total of 23,518 base pairs). The virus was obtained from the Orbivirus Reference Collection (ORC) at IAH, Pirbright, United Kingdom. The virus strain, which was previously provided by the Onderstepoort Veterinary Research Institute in South Africa, was originally isolated from the Indian subcontinent (Hazara, West Pakistan) in 1960. Previous phylogenetic comparisons show that BTV RNA sequences cluster according to the geographic origins of the virus isolate/lineage, identifying distinct BTV topotypes. Sequence comparisons of segments Seg-1 to Seg-10 show that RSArrrr/16 belongs to the major eastern topotype of BTV (BTV-16e) and can be regarded as a reference strain of BTV-16e for phylogenetic and molecular epidemiology studies. All 10 genome segments of RSArrrr/16 group closely with the vaccine strain of BTV-16 (RSAvvvv/16) that was derived from it, as well as those recently published for a Chinese isolate of BTV-16 (>99% nucleotide identity), suggesting a very recent common ancestry for all three viruses.


Assuntos
Vírus Bluetongue/genética , Animais , Bluetongue/virologia , Vírus Bluetongue/classificação , Genoma Viral , Índia , Dados de Sequência Molecular , Filogenia , Sorotipagem
8.
J Virol ; 86(12): 7011-2, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22628397

RESUMO

The full genome sequence (19,177 bp) of an Indian strain (IND1988/02) of bluetongue virus (BTV) serotype 23 was determined. This virus was isolated from a sheep that had been killed during a severe bluetongue outbreak that occurred in Rahuri, Maharashtra State, western India, in 1988. Phylogenetic analyses of these data demonstrate that most of the genome segments from IND1988/02 belong to the major "eastern" BTV topotype. However, genome segment 5 belongs to the major "western" BTV topotype, demonstrating that IND1988/02 is a reassortant. This may help to explain the increased virulence that was seen during this outbreak in 1988. Genome segment 5 of IND1988/02 shows >99% sequence identity with some other BTV isolates from India (e.g., BTV-3 IND2003/08), providing further evidence of the existence and circulation of reassortant strains on the subcontinent.


Assuntos
Vírus Bluetongue/genética , Bluetongue/virologia , Genoma Viral , Vírus Reordenados/genética , Animais , Sequência de Bases , Vírus Bluetongue/classificação , Vírus Bluetongue/isolamento & purificação , Índia , Dados de Sequência Molecular , Vírus Reordenados/classificação , Vírus Reordenados/isolamento & purificação , Ovinos
9.
J Virol ; 86(11): 6375-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22570246

RESUMO

All 10 genome segments (Seg-1 to 10-a total of 19,188 bp) were sequenced from a strain of bluetongue virus serotype 3 (BTV-3) from India (strain IND2003/08). Sequence comparisons showed that nine of the genome segments from this virus group with other eastern topotype strains. Genome Seg-2 and Seg-6 group with eastern BTV-3 strains from Japan. However, Seg-5 (the NS1 gene) from IND2003/08 belongs to a western lineage, demonstrating that IND2003/08 is a reassortant between eastern and western topotype bluetongue viruses. This confirms that western BTV strains have been imported and are circulating within the subcontinent.


Assuntos
Vírus Bluetongue/genética , Genoma Viral , RNA Viral/genética , Vírus Reordenados/genética , Análise de Sequência de DNA , Animais , Vírus Bluetongue/isolamento & purificação , Índia , Dados de Sequência Molecular , Filogenia , Vírus Reordenados/isolamento & purificação , Homologia de Sequência
10.
J Virol ; 86(10): 5967-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22532533

RESUMO

Bluetongue virus type 2, isolated in India in 1982 (IND1982/01), was obtained from the Orbivirus Reference Collection at IAH Pirbright (http://www.reoviridae.org/dsRNA_virus_proteins/ReoID/btv-2.htm#IND1982/01). Full genome sequencing and phylogenetic analyses show that IND1982/01 is a reassortant virus containing genome segments derived from both eastern and western topotypes. These data will help to identify further reassortment events involving this or other virus lineages in the subcontinent.


Assuntos
Vírus Bluetongue/genética , Bluetongue/virologia , Genoma Viral , Recombinação Genética , Animais , Sequência de Bases , Vírus Bluetongue/classificação , Vírus Bluetongue/isolamento & purificação , Índia , Dados de Sequência Molecular , Filogenia , Ruminantes
11.
J Virol ; 86(10): 5971-2, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22532535

RESUMO

Bluetongue virus is the type species of the genus Orbivirus in the family Reoviridae. We report the first complete genome sequence of an isolate (IND2004/01) of bluetongue virus serotype 10 (BTV-10) from Andhra Pradesh, India. This isolate, which is stored in the Orbivirus Reference Collection (ORC) at IAH Pirbright, shows >99% nucleotide identity in all 10 genome segments with a vaccine strain of BTV-10 from the United States.


Assuntos
Vírus Bluetongue/genética , Bluetongue/virologia , Genoma Viral , Sequência de Bases , Vírus Bluetongue/classificação , Vírus Bluetongue/isolamento & purificação , Índia , Dados de Sequência Molecular , Estados Unidos , Vacinas Virais/genética
12.
J Virol ; 86(9): 5404-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22492927

RESUMO

Bluetongue virus serotype 2 (IND2003/02) was isolated in Tiruneveli City, Tamil Nadu State, India, and is stored in the Orbivirus Reference Collection at the Institute for Animal Health, Pirbright, United Kingdom. The entire genome of this isolate was sequenced, showing that it is composed of a total of 19,203 bp (all 10 genome segments). This is the first report of the entire genome sequence of a western strain of BTV-2 isolated in India, indicating that this virus has been introduced and is circulating in the region. These data will aid in the development of diagnostics and molecular epidemiology studies of BTV-2 in the subcontinent.


Assuntos
Vírus Bluetongue/genética , Genoma Viral , Animais , Vírus Bluetongue/isolamento & purificação , Índia , Anotação de Sequência Molecular , Dados de Sequência Molecular
13.
J Virol ; 86(8): 4717-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22457532

RESUMO

We report the full-genome sequence of an Indian isolate of bluetongue virus serotype 1 (BTV-1), strain IND1992/01. This is the first report of the entire genome sequence (Seg-1 to Seg-10) of an Eastern (e) strain of BTV-1. These sequence data provide a reference for BTV-1e that will help to define the phylogenetic relationships and geographic origins of distinct Indian lineages of BTV-1 as well as their relationships with other BTV strains from around the world. The availability of data for all 10 genome segments of this strain will also help to identify reassortment events involving this and other virus lineages.


Assuntos
Vírus Bluetongue/classificação , Vírus Bluetongue/genética , Genoma Viral , Índia , Dados de Sequência Molecular , Sorotipagem
14.
Virol J ; 10: 319, 2013 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-24165208

RESUMO

BACKGROUND: Bluetongue virus (BTV) is an arbovirus that is responsible for 'bluetongue', an economically important disease of livestock. Although BTV is well characterised at the protein level, less is known regarding its interaction with host cells. During studies of virus inclusion body formation we observed what appeared to be a large proportion of cells in mitosis. Although the modulation of the cell cycle is well established for many viruses, this was a novel observation for BTV. We therefore undertook a study to reveal in more depth the impact of BTV upon cell division. METHODS: We used a confocal microscopy approach to investigate the localisation of BTV proteins in a cellular context with their respective position relative to cellular proteins. In addition, to quantitatively assess the frequency of aberrant mitosis induction by the viral non-structural protein (NS) 2 we utilised live cell imaging to monitor HeLa-mCherry tubulin cells transfected with a plasmid expressing NS2. RESULTS: Our data showed that these 'aberrant mitoses' can be induced in multiple cell types and by different strains of BTV. Further study confirmed multiplication of the centrosomes, each resulting in a separate mitotic spindle during mitosis. Interestingly, the BTV NS1 protein was strongly localised to the centrosomal regions. In a separate, yet related observation, the BTV NS2 protein was co-localised with the condensed chromosomes to a region suggestive of the kinetochore. Live cell imaging revealed that expression of an EGFP-NS2 fusion protein in HeLa-mCherry tubulin cells also results in mitotic defects. CONCLUSIONS: We hypothesise that NS2 is a microtubule cargo protein that may inadvertently disrupt the interaction of microtubule tips with the kinetochores during mitosis. Furthermore, the BTV NS1 protein was distinctly localised to a region encompassing the centrosome and may therefore be, at least in part, responsible for the disruption of the centrosome as observed in BTV infected mammalian cells.


Assuntos
Vírus Bluetongue/fisiologia , Interações Hospedeiro-Patógeno , Mitose , Animais , Linhagem Celular , Citosol/química , Citosol/virologia , Microscopia Confocal , Proteínas Virais/análise
15.
Biomolecules ; 13(6)2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37371457

RESUMO

At least 12 serotypes of 'atypical' bluetongue virus (BTV-25 to BTV-36) have been identified to date. These atypical serotypes fail to infect/replicate in Culicoides-derived cell lines and/or adult Culicoides vectors and hence can no longer be transmitted by these vectors. They appear to be horizontally transmitted from infected to in-contact ruminants, although the route(s) of infection remain to be identified. Viral genome segments 1, 2 and 3 (Seg-1, Seg2 and Seg-3) of BTV-26 were identified as involved in blocking virus replication in KC cells. We have developed Culicoides-specific expression plasmids, which we used in transfected insect cells to assess the stability of viral mRNAs and protein expression from full-length open reading frames of Seg-1, -2 and -3 of BTV-1 (a Culicoides-vectored BTV) or BTV-26. Our results indicate that the blocked replication of BTV-26 in KC cells is not due to an RNAi response, which would lead to rapid degradation of viral mRNAs. A combination of degradation/poor expression and/or modification of the proteins encoded by these segments appears to drive the failure of BTV-26 core/whole virus-particles to assemble and replicate effectively in Culicoides cells.


Assuntos
Vírus Bluetongue , Ceratopogonidae , Animais , Vírus Bluetongue/genética , Vírus Bluetongue/metabolismo , Ceratopogonidae/genética , Sorogrupo , Genoma Viral , Linhagem Celular , Replicação Viral/genética
16.
Viruses ; 15(9)2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37766314

RESUMO

Non-structural protein 4 (NS4) of insect-borne and tick-borne orbiviruses is encoded by genome segment 9, from a secondary open reading frame. Though a protein dispensable for bluetongue virus (BTV) replication, it has been shown to counter the interferon response in cells infected with BTV or African horse sickness virus. We further explored the functional role(s) of NS4 proteins of BTV and the tick-borne Great Island virus (GIV). We show that NS4 of BTV or GIV helps an E3L deletion mutant of vaccinia virus to replicate efficiently in interferon-treated cells, further confirming the role of NS4 as an interferon antagonist. Our results indicate that ectopically expressed NS4 of BTV localised with caspase 3 within the nucleus and was found in a protein complex with active caspase 3 in a pull-down assay. Previous studies have shown that pro-apoptotic caspases (including caspase 3) suppress type I interferon response by cleaving mediators involved in interferon signalling. Our data suggest that orbivirus NS4 plays a role in modulating the apoptotic process and/or regulating the interferon response in mammalian cells, thus acting as a virulence factor in pathogenesis.


Assuntos
Vírus Bluetongue , Interferon Tipo I , Orbivirus , Thogotovirus , Animais , Orbivirus/genética , Caspase 3 , Vírus Bluetongue/genética , Apoptose , Mamíferos
17.
Pathogens ; 12(4)2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-37111488

RESUMO

Bluetongue is an economically important disease of domesticated and wild ruminants caused by bluetongue virus (BTV). There are at least 36 different serotypes of BTV (the identity of which is determined by its outer-capsid protein VP2), most of which are transmitted by Culicoides biting midges. IFNAR(-/-) mice immunised with plant-expressed outer-capsid protein VP2 (rVP2) of BTV serotypes -1, -4 or -8, or the smaller outer-capsid protein rVP5 of BTV-10, or mock-immunised with PBS, were subsequently challenged with virulent strains of BTV-4 or BTV-8, or with an attenuated clone of BTV-1 (BTV-1RGC7). The mice that had received rVP2 generated a protective immune response against the homologous BTV serotype, reducing viraemia (as detected by qRT-PCR), the severity of clinical signs and mortality levels. No cross-serotype protection was observed after challenge with the heterologous BTV serotypes. However, the severity of clinical signs, viraemia and fatality levels after challenge with the attenuated strain of BTV-1 were all increased in mice immunised with rVP2 of BTV-4 and BTV-8, or with rVP5 of BTV10. The possibility is discussed that non-neutralising antibodies, reflecting serological relationships between the outer-capsid proteins of these different BTV serotypes, could lead to 'antibody-dependent enhancement of infection' (ADE). Such interactions could affect the epidemiology and emergence of different BTV strains in the field and would therefore be relevant to the design and implementation of vaccination campaigns.

18.
Transbound Emerg Dis ; 69(4): e671-e681, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34921513

RESUMO

African horse sickness (AHS) is a vector-borne disease transmitted by Culicoides spp., endemic to sub-Saharan Africa. There have been many examples of historic and recent outbreaks in the Middle East, Asia and Europe. However, not much is known about infection dynamics and outbreak potential in these naive populations. In order to better inform a previously published ordinary differential equation model, we performed a systematic literature search to identify studies documenting experimental infection of naive (control) equids in vaccination trials. Data on the time until the onset of viraemia, clinical signs and death after experimental infection of a naive equid and duration of viraemia were extracted. The time to viraemia was 4.6 days and the time to clinical signs was 4.9 days, longer than the previously estimated latent period of 3.7 days. The infectious periods of animals that died/were euthanized or survived were found to be 3.9 and 8.7 days, whereas previous estimations were 4.4 and 6 days, respectively. The case fatality was also found to be higher than previous estimations. The updated parameter values (along with other more recently published estimates from literature) resulted in an increase in the number of host deaths, decrease in the duration of the outbreak and greater prevalence in vectors.


Assuntos
Vírus da Doença Equina Africana , Doença Equina Africana , Ceratopogonidae , Doenças dos Cavalos , Doença Equina Africana/epidemiologia , Animais , Cavalos , Modelos Teóricos , Viremia/veterinária
19.
Emerg Infect Dis ; 17(5): 886-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21529403

RESUMO

Sheep and goats sampled in Kuwait during February 2010 were seropositive for bluetongue virus (BTV). BTV isolate KUW2010/02, from 1 of only 2 sheep that also tested positive for BTV by real-time reverse transcription-PCR, caused mild clinical signs in sheep. Nucleotide sequencing identified KUW2010/02 as a novel BTV serotype.


Assuntos
Vírus Bluetongue/classificação , Vírus Bluetongue/isolamento & purificação , Bluetongue/virologia , Animais , Vírus Bluetongue/genética , Proteínas do Capsídeo/genética , Cabras , Kuweit , Filogenia , RNA Viral/genética , Homologia de Sequência , Sorotipagem , Ovinos
20.
J Virol ; 84(4): 2013-26, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19939934

RESUMO

Although few simian rotaviruses (RVs) have been isolated, such strains have been important for basic research and vaccine development. To explore the origins of simian RVs, the complete genome sequences of strains PTRV (G8P[1]), RRV (G3P[3]), and TUCH (G3P[24]) were determined. These data allowed the genotype constellations of each virus to be determined and the phylogenetic relationships of the simian strains with each other and with nonsimian RVs to be elucidated. The results indicate that PTRV was likely transmitted from a bovine or other ruminant into pig-tailed macaques (its host of origin), since its genes have genotypes and encode outer-capsid proteins similar to those of bovine RVs. In contrast, most of the genes of rhesus-macaque strains, RRV and TUCH, have genotypes more typical of canine-feline RVs. However, the sequences of the canine and/or feline (canine/feline)-like genes of RRV and TUCH are only distantly related to those of modern canine/feline RVs, indicating that any potential transmission of a progenitor of these viruses from a canine/feline host to a simian host was not recent. The remaining genes of RRV and TUCH appear to have originated through reassortment with bovine, human, or other RV strains. Finally, comparison of PTRV, RRV, and TUCH genes with those of the vervet-monkey RV SA11-H96 (G3P[2]) indicates that SA11-H96 shares little genetic similarity to other simian strains and likely has evolved independently. Collectively, our data indicate that simian RVs are of diverse ancestry with genome constellations that originated largely by interspecies transmission and reassortment with nonhuman animal RVs.


Assuntos
Haplorrinos/virologia , Vírus Reordenados/genética , Rotavirus/genética , Sequência de Aminoácidos , Animais , Antígenos Virais/química , Antígenos Virais/genética , Gatos , Bovinos , Cães , Genoma Viral , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Estrutura Terciária de Proteína , Vírus Reordenados/classificação , Vírus Reordenados/imunologia , Vírus Reordenados/patogenicidade , Rotavirus/classificação , Rotavirus/imunologia , Rotavirus/patogenicidade , Infecções por Rotavirus/transmissão , Infecções por Rotavirus/veterinária , Infecções por Rotavirus/virologia , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa