Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 635
Filtrar
1.
Nature ; 602(7896): 307-313, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34937050

RESUMEN

Emerging variants of concern (VOCs) are driving the COVID-19 pandemic1,2. Experimental assessments of replication and transmission of major VOCs and progenitors are needed to understand the mechanisms of replication and transmission of VOCs3. Here we show that the spike protein (S) from Alpha (also known as B.1.1.7) and Beta (B.1.351) VOCs had a greater affinity towards the human angiotensin-converting enzyme 2 (ACE2) receptor than that of the progenitor variant S(D614G) in vitro. Progenitor variant virus expressing S(D614G) (wt-S614G) and the Alpha variant showed similar replication kinetics in human nasal airway epithelial cultures, whereas the Beta variant was outcompeted by both. In vivo, competition experiments showed a clear fitness advantage of Alpha over wt-S614G in ferrets and two mouse models-the substitutions in S were major drivers of the fitness advantage. In hamsters, which support high viral replication levels, Alpha and wt-S614G showed similar fitness. By contrast, Beta was outcompeted by Alpha and wt-S614G in hamsters and in mice expressing human ACE2. Our study highlights the importance of using multiple models to characterize fitness of VOCs and demonstrates that Alpha is adapted for replication in the upper respiratory tract and shows enhanced transmission in vivo in restrictive models, whereas Beta does not overcome Alpha or wt-S614G in naive animals.


Asunto(s)
COVID-19/transmisión , COVID-19/virología , Mutación , SARS-CoV-2/clasificación , SARS-CoV-2/fisiología , Replicación Viral , Sustitución de Aminoácidos , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Animales de Laboratorio/virología , COVID-19/veterinaria , Cricetinae , Modelos Animales de Enfermedad , Células Epiteliales/virología , Femenino , Hurones/virología , Humanos , Masculino , Mesocricetus/virología , Ratones , Ratones Transgénicos , SARS-CoV-2/genética , SARS-CoV-2/crecimiento & desarrollo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Virulencia/genética
2.
Nature ; 592(7852): 122-127, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636719

RESUMEN

During the evolution of SARS-CoV-2 in humans, a D614G substitution in the spike glycoprotein (S) has emerged; virus containing this substitution has become the predominant circulating variant in the COVID-19 pandemic1. However, whether the increasing prevalence of this variant reflects a fitness advantage that improves replication and/or transmission in humans or is merely due to founder effects remains unknown. Here we use isogenic SARS-CoV-2 variants to demonstrate that the variant that contains S(D614G) has enhanced binding to the human cell-surface receptor angiotensin-converting enzyme 2 (ACE2), increased replication in primary human bronchial and nasal airway epithelial cultures as well as in a human ACE2 knock-in mouse model, and markedly increased replication and transmissibility in hamster and ferret models of SARS-CoV-2 infection. Our data show that the D614G substitution in S results in subtle increases in binding and replication in vitro, and provides a real competitive advantage in vivo-particularly during the transmission bottleneck. Our data therefore provide an explanation for the global predominance of the variant that contains S(D614G) among the SARS-CoV-2 viruses that are currently circulating.


Asunto(s)
COVID-19/transmisión , COVID-19/virología , Mutación , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/genética , Replicación Viral/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Bronquios/citología , Bronquios/virología , COVID-19/epidemiología , Línea Celular , Células Cultivadas , Cricetinae , Modelos Animales de Enfermedad , Células Epiteliales/virología , Femenino , Hurones/virología , Efecto Fundador , Técnicas de Sustitución del Gen , Aptitud Genética , Humanos , Masculino , Mesocricetus , Ratones , Mucosa Nasal/citología , Mucosa Nasal/virología , Unión Proteica , ARN Viral/análisis , Receptores de Coronavirus/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad
3.
PLoS Pathog ; 20(1): e1011880, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38271294

RESUMEN

BACKGROUND: West Nile virus (WNV) outbreaks in birds, humans, and livestock have occurred in multiple areas in Europe and have had a significant impact on animal and human health. The patterns of emergence and spread of WNV in Europe are very different from those in the US and understanding these are important for guiding preparedness activities. METHODS: We mapped the evolution and spread history of WNV in Europe by incorporating viral genome sequences and epidemiological data into phylodynamic models. Spatially explicit phylogeographic models were developed to explore the possible contribution of different drivers to viral dispersal direction and velocity. A "skygrid-GLM" approach was used to identify how changes in environments would predict viral genetic diversity variations over time. FINDINGS: Among the six lineages found in Europe, WNV-2a (a sub-lineage of WNV-2) has been predominant (accounting for 73% of all sequences obtained in Europe that have been shared in the public domain) and has spread to at least 14 countries. In the past two decades, WNV-2a has evolved into two major co-circulating clusters, both originating from Central Europe, but with distinct dynamic history and transmission patterns. WNV-2a spreads at a high dispersal velocity (88km/yr-215 km/yr) which is correlated to bird movements. Notably, amongst multiple drivers that could affect the spread of WNV, factors related to land use were found to strongly influence the spread of WNV. Specifically, the intensity of agricultural activities (defined by factors related to crops and livestock production, such as coverage of cropland, pasture, cultivated and managed vegetation, livestock density) were positively associated with both spread direction and velocity. In addition, WNV spread direction was associated with high coverage of wetlands and migratory bird flyways. CONCLUSION: Our results suggest that-in addition to ecological conditions favouring bird- and mosquito- presence-agricultural land use may be a significant driver of WNV emergence and spread. Our study also identified significant gaps in data and the need to strengthen virological surveillance in countries of Central Europe from where WNV outbreaks are likely seeded. Enhanced monitoring for early detection of further dispersal could be targeted to areas with high agricultural activities and habitats of migratory birds.


Asunto(s)
Fiebre del Nilo Occidental , Virus del Nilo Occidental , Animales , Humanos , Virus del Nilo Occidental/genética , Fiebre del Nilo Occidental/epidemiología , Fiebre del Nilo Occidental/veterinaria , Filogeografía , Europa (Continente)/epidemiología , Brotes de Enfermedades
4.
Nature ; 586(7829): 424-428, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33029010

RESUMEN

Since 1814, when rubella was first described, the origins of the disease and its causative agent, rubella virus (Matonaviridae: Rubivirus), have remained unclear1. Here we describe ruhugu virus and rustrela virus in Africa and Europe, respectively, which are, to our knowledge, the first known relatives of rubella virus. Ruhugu virus, which is the closest relative of rubella virus, was found in apparently healthy cyclops leaf-nosed bats (Hipposideros cyclops) in Uganda. Rustrela virus, which is an outgroup to the clade that comprises rubella and ruhugu viruses, was found in acutely encephalitic placental and marsupial animals at a zoo in Germany and in wild yellow-necked field mice (Apodemus flavicollis) at and near the zoo. Ruhugu and rustrela viruses share an identical genomic architecture with rubella virus2,3. The amino acid sequences of four putative B cell epitopes in the fusion (E1) protein of the rubella, ruhugu and rustrela viruses and two putative T cell epitopes in the capsid protein of the rubella and ruhugu viruses are moderately to highly conserved4-6. Modelling of E1 homotrimers in the post-fusion state predicts that ruhugu and rubella viruses have a similar capacity for fusion with the host-cell membrane5. Together, these findings show that some members of the family Matonaviridae can cross substantial barriers between host species and that rubella virus probably has a zoonotic origin. Our findings raise concerns about future zoonotic transmission of rubella-like viruses, but will facilitate comparative studies and animal models of rubella and congenital rubella syndrome.


Asunto(s)
Mamíferos/virología , Filogenia , Virus de la Rubéola/clasificación , Virus de la Rubéola/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Animales de Zoológico/inmunología , Animales de Zoológico/virología , Membrana Celular/virología , Quirópteros/virología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Equidae/inmunología , Equidae/virología , Evolución Molecular , Femenino , Mapeo Geográfico , Alemania , Especificidad del Huésped , Humanos , Masculino , Mamíferos/inmunología , Marsupiales/inmunología , Marsupiales/virología , Fusión de Membrana , Ratones , Modelos Animales , Modelos Moleculares , Rubéola (Sarampión Alemán)/congénito , Rubéola (Sarampión Alemán)/virología , Virus de la Rubéola/química , Virus de la Rubéola/inmunología , Alineación de Secuencia , Uganda , Proteínas del Envoltorio Viral/química
5.
Nature ; 588(7836): E2, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33199919

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Nature ; 567(7746): 109-112, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30787439

RESUMEN

Zoonotic influenza A viruses of avian origin can cause severe disease in individuals, or even global pandemics, and thus pose a threat to human populations. Waterfowl and shorebirds are believed to be the reservoir for all influenza A viruses, but this has recently been challenged by the identification of novel influenza A viruses in bats1,2. The major bat influenza A virus envelope glycoprotein, haemagglutinin, does not bind the canonical influenza A virus receptor, sialic acid or any other glycan1,3,4, despite its high sequence and structural homology with conventional haemagglutinins. This functionally uncharacterized plasticity of the bat influenza A virus haemagglutinin means the tropism and zoonotic potential of these viruses has not been fully determined. Here we show, using transcriptomic profiling of susceptible versus non-susceptible cells in combination with genome-wide CRISPR-Cas9 screening, that the major histocompatibility complex class II (MHC-II) human leukocyte antigen DR isotype (HLA-DR) is an essential entry determinant for bat influenza A viruses. Genetic ablation of the HLA-DR α-chain rendered cells resistant to infection by bat influenza A virus, whereas ectopic expression of the HLA-DR complex in non-susceptible cells conferred susceptibility. Expression of MHC-II from different bat species, pigs, mice or chickens also conferred susceptibility to infection. Notably, the infection of mice with bat influenza A virus resulted in robust virus replication in the upper respiratory tract, whereas mice deficient for MHC-II were resistant. Collectively, our data identify MHC-II as a crucial entry mediator for bat influenza A viruses in multiple species, which permits a broad vertebrate tropism.


Asunto(s)
Quirópteros/virología , Antígenos de Histocompatibilidad Clase II/metabolismo , Especificidad del Huésped , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Zoonosis/inmunología , Zoonosis/virología , Animales , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Pollos/genética , Pollos/inmunología , Quirópteros/genética , Quirópteros/inmunología , Quirópteros/metabolismo , Femenino , Perfilación de la Expresión Génica , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Humanos , Masculino , Ratones , Ratones Noqueados , Sistema Respiratorio/virología , Porcinos/genética , Porcinos/inmunología , Tropismo Viral/genética , Tropismo Viral/inmunología , Replicación Viral , Zoonosis/genética , Zoonosis/metabolismo
7.
Emerg Infect Dis ; 30(2): 399-401, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38270110

RESUMEN

We identified a novel lineage of lymphocytic choriomeningitis virus, tentatively named lineage V, in wood mice (Apodemus sylvaticus) from Germany. Wood mouse-derived lymphocytic choriomeningitis virus can be found across a substantially greater range than previously thought. Increased surveillance is needed to determine its geographic range and zoonotic potential.


Asunto(s)
Virus de la Coriomeningitis Linfocítica , Ratones , Animales , Virus de la Coriomeningitis Linfocítica/genética , Alemania/epidemiología
9.
J Gen Virol ; 105(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38921821

RESUMEN

Schmallenberg virus (SBV) belongs to the Simbu serogroup within the family Peribunyaviridae, genus Orthobunyavirus and is transmitted by Culicoides biting midges. Infection of naïve ruminants in a critical phase of gestation may lead to severe congenital malformations. Sequence analysis from viremic animals revealed a very high genome stability. In contrast, sequence variations are frequently described for SBV from malformed fetuses. In addition to S segment mutations, especially within the M segment encoding the major immunogen Gc, point mutations or genomic deletions are also observed. Analysis of the SBV_D281/12 isolate from a malformed fetus revealed multiple point mutations in all three genome segments. It also has a large genomic deletion in the antigenic domain encoded by the M segment compared to the original SBV reference strain 'BH80/11' isolated from viremic blood in 2011. Interestingly, SBV_D281/12 showed a marked replication deficiency in vitro in Culicoides sonorensis cells (KC cells), but not in standard baby hamster kidney cells (BHK-21). We therefore generated a set of chimeric viruses of rSBV_D281/12 and wild-type rSBV_BH80/11 by reverse genetics, which were characterized in both KC and BHK-21 cells. It could be shown that the S segment of SBV_D281/12 is responsible for the replication deficit and that it acts independently from the large deletion within Gc. In addition, a single point mutation at position 111 (S to N) of the nucleoprotein was identified as the critical mutation. Our results suggest that virus variants found in malformed fetuses and carrying characteristic genomic mutations may have a clear 'loss of fitness' for their insect hosts in vitro. It can also be concluded that such mutations lead to virus variants that are no longer part of the natural transmission cycle between mammalian and insect hosts. Interestingly, analysis of a series of SBV sequences confirmed the S111N mutation exclusively in samples of malformed fetuses and not in blood from viremic animals. The characterization of these changes will allow the definition of protein functions that are critical for only one group of hosts.


Asunto(s)
Infecciones por Bunyaviridae , Ceratopogonidae , Genoma Viral , Orthobunyavirus , Animales , Orthobunyavirus/genética , Orthobunyavirus/clasificación , Orthobunyavirus/aislamiento & purificación , Infecciones por Bunyaviridae/virología , Infecciones por Bunyaviridae/veterinaria , Ceratopogonidae/virología , Cricetinae , Línea Celular , Replicación Viral , Mutación Puntual , Bovinos , Ovinos , Filogenia , ARN Viral/genética
10.
J Gen Virol ; 105(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38695722

RESUMEN

High-pathogenicity avian influenza viruses (HPAIVs) of the goose/Guangdong lineage are enzootically circulating in wild bird populations worldwide. This increases the risk of entry into poultry production and spill-over to mammalian species, including humans. Better understanding of the ecological and epizootiological networks of these viruses is essential to optimize mitigation measures. Based on full genome sequences of 26 HPAIV samples from Iceland, which were collected between spring and autumn 2022, as well as 1 sample from the 2023 summer period, we show that 3 different genotypes of HPAIV H5N1 clade 2.3.4.4b were circulating within the wild bird population in Iceland in 2022. Furthermore, in 2023 we observed a novel introduction of HPAIV H5N5 of the same clade to Iceland. The data support the role of Iceland as an utmost northwestern distribution area in Europe that might act also as a potential bridging point for intercontinental spread of HPAIV across the North Atlantic.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Filogenia , Islandia/epidemiología , Animales , Gripe Aviar/virología , Gripe Aviar/epidemiología , Gripe Aviar/transmisión , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Genotipo , Animales Salvajes/virología , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Genoma Viral , Aves/virología
11.
Eur J Immunol ; 53(12): e2250332, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37609807

RESUMEN

Knowledge about early immunity to SARS-CoV-2 variants of concern mainly comes from the analysis of human blood. Such data provide limited information about host responses at the site of infection and largely miss the initial events. To gain insights into compartmentalization and the early dynamics of host responses to different SARS-CoV-2 variants, we utilized human angiotensin converting enzyme 2 (hACE2) transgenic mice and tracked immune changes during the first days after infection by RNAseq, multiplex assays, and flow cytometry. Viral challenge infection led to divergent viral loads in the lungs, distinct inflammatory patterns, and innate immune cell accumulation in response to ancestral SARS-CoV-2, Beta (B.1.351) and Delta (B.1.617.2) variant of concern (VOC). Compared to other SARS-CoV-2 variants, infection with Beta (B.1.351) VOC spread promptly to the lungs, leading to increased inflammatory responses. SARS-CoV-2-specific antibodies and T cells developed within the first 7 days postinfection and were required to reduce viral spread and replication. Our studies show that VOCs differentially trigger transcriptional profiles and inflammation. This information contributes to the basic understanding of immune responses immediately postexposure to SARS-CoV-2 and is relevant for developing pan-VOC interventions including prophylactic vaccines.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Ratones , Humanos , Enzima Convertidora de Angiotensina 2/genética , COVID-19/genética , Anticuerpos Antivirales , Ratones Transgénicos , Inmunidad
12.
J Virol ; 97(10): e0107623, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37811996

RESUMEN

IMPORTANCE: The influenza A virus genome consists of eight distinct viral RNAs (vRNAs) that are typically packaged into a single virion as an octameric complex. How this genome complex is assembled and incorporated into the virion is poorly understood, but previous research suggests a coordinative role for packaging signals present in all vRNAs. Here, we show that disruption of two packaging signals in a model H7N7 influenza A virus results in a mixture of virions with unusual vRNA content, including empty virions, virions with one to four vRNAs, and virions with octameric complexes composed of vRNA duplicates. Our results suggest that (i) the assembly of error-free octameric complexes proceeds through a series of defined vRNA sub-complexes and (ii) virions can bud without incorporating complete octameric complexes.


Asunto(s)
Subtipo H7N7 del Virus de la Influenza A , Virus de la Influenza A , Empaquetamiento del Genoma Viral , Ensamble de Virus , Genoma Viral , Virus de la Influenza A/genética , Subtipo H7N7 del Virus de la Influenza A/genética , ARN Viral/genética , Virión/genética
13.
J Virol ; 97(4): e0193222, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37022231

RESUMEN

High-throughput sequences were generated from DNA and cDNA from four Southern white rhinoceros (Ceratotherium simum simum) located in the Taronga Western Plain Zoo in Australia. Virome analysis identified reads that were similar to Mus caroli endogenous gammaretrovirus (McERV). Previous analysis of perissodactyl genomes did not recover gammaretroviruses. Our analysis, including the screening of the updated white rhinoceros (Ceratotherium simum) and black rhinoceros (Diceros bicornis) draft genomes identified high-copy orthologous gammaretroviral ERVs. Screening of Asian rhinoceros, extinct rhinoceros, domestic horse, and tapir genomes did not identify related gammaretroviral sequences in these species. The newly identified proviral sequences were designated SimumERV and DicerosERV for the white and black rhinoceros retroviruses, respectively. Two long terminal repeat (LTR) variants (LTR-A and LTR-B) were identified in the black rhinoceros, with different copy numbers associated with each (n = 101 and 373, respectively). Only the LTR-A lineage (n = 467) was found in the white rhinoceros. The African and Asian rhinoceros lineages diverged approximately 16 million years ago. Divergence age estimation of the identified proviruses suggests that the exogenous retroviral ancestor of the African rhinoceros ERVs colonized their genomes within the last 8 million years, a result consistent with the absence of these gammaretroviruses from Asian rhinoceros and other perissodactyls. The black rhinoceros germ line was colonized by two lineages of closely related retroviruses and white rhinoceros by one. Phylogenetic analysis indicates a close evolutionary relationship with ERVs of rodents including sympatric African rats, suggesting a possible African origin of the identified rhinoceros gammaretroviruses. IMPORTANCE Rhinoceros genomes were thought to be devoid of gammaretroviruses, as has been determined for other perissodactyls (horses, tapirs, and rhinoceros). While this may be true of most rhinoceros, the African white and black rhinoceros genomes have been colonized by evolutionarily young gammaretroviruses (SimumERV and DicerosERV for the white and black rhinoceros, respectively). These high-copy endogenous retroviruses (ERVs) may have expanded in multiple waves. The closest relative of SimumERV and DicerosERV is found in rodents, including African endemic species. Restriction of the ERVs to African rhinoceros suggests an African origin for the rhinoceros gammaretroviruses.


Asunto(s)
Evolución Biológica , Retrovirus Endógenos , Gammaretrovirus , Perisodáctilos , Animales , Ratones , Ratas , Retrovirus Endógenos/clasificación , Retrovirus Endógenos/genética , Gammaretrovirus/clasificación , Gammaretrovirus/genética , Caballos/genética , Caballos/virología , Perisodáctilos/genética , Perisodáctilos/virología , Filogenia , Provirus/genética
14.
PLoS Pathog ; 18(1): e1010161, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35025969

RESUMEN

The global response to Coronavirus Disease 2019 (COVID-19) is now facing new challenges such as vaccine inequity and the emergence of SARS-CoV-2 variants of concern (VOCs). Preclinical models of disease, in particular animal models, are essential to investigate VOC pathogenesis, vaccine correlates of protection and postexposure therapies. Here, we provide an update from the World Health Organization (WHO) COVID-19 modeling expert group (WHO-COM) assembled by WHO, regarding advances in preclinical models. In particular, we discuss how animal model research is playing a key role to evaluate VOC virulence, transmission and immune escape, and how animal models are being refined to recapitulate COVID-19 demographic variables such as comorbidities and age.


Asunto(s)
COVID-19/etiología , Modelos Animales de Enfermedad , SARS-CoV-2 , Factores de Edad , Animales , COVID-19/prevención & control , COVID-19/terapia , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/inmunología , Comorbilidad , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad
15.
Emerg Infect Dis ; 29(6): 1202-1205, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37209672

RESUMEN

We detected a novel poxvirus from a gray seal (Halichoerus grypus) from the North Sea, Germany. The juvenile animal showed pox-like lesions and deteriorating overall health condition and was finally euthanized. Histology, electron microscopy, sequencing, and PCR confirmed a previously undescribed poxvirus of the Chordopoxvirinae subfamily, tentatively named Wadden Sea poxvirus.


Asunto(s)
Chordopoxvirinae , Poxviridae , Phocidae , Animales , Poxviridae/genética , Mar del Norte , Alemania/epidemiología
16.
Emerg Infect Dis ; 29(7): 1492-1495, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37347930

RESUMEN

We found that nasal and alimentary experimental exposure of pigs to highly pathogenic avian influenza virus H5N1 clade 2.3.4.4b was associated with marginal viral replication, without inducing any clinical manifestation or pathological changes. Only 1 of 8 pigs seroconverted, pointing to high resistance of pigs to clade 2.3.4.4b infection.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Animales , Porcinos , Replicación Viral
17.
Emerg Infect Dis ; 29(5): 1042-1045, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37081716

RESUMEN

Retrospective investigation of archived tissue samples from 3 lions displaying nonsuppurative meningoencephalitis and vasculitis led to the detection of rustrela virus (RusV). We confirmed RusV antigen and RNA in cortical neurons, axons, astrocytes and Purkinje cells by reverse transcription quantitative PCR, immunohistochemistry, and in situ hybridization.


Asunto(s)
Leones , Meningoencefalitis , Virus , Animales , Estudios Retrospectivos , Meningoencefalitis/diagnóstico , Meningoencefalitis/veterinaria , Inmunohistoquímica
18.
Emerg Infect Dis ; 29(3): 631-634, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36823667

RESUMEN

Lymphocytic choriomeningitis mammarenavirus (LCMV) is a globally distributed zoonotic pathogen transmitted by house mice (Mus musculus). We report the reemergence of LCMV (lineages I and II) in wild house mice (Mus musculus domesticus) and LCMV lineage I in a diseased golden lion tamarin (Leontopithecus rosalia) from a zoo in Germany.


Asunto(s)
Coriomeningitis Linfocítica , Enfermedades de los Roedores , Animales , Ratones , Virus de la Coriomeningitis Linfocítica , Alemania
19.
J Gen Virol ; 104(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37014781

RESUMEN

Mass mortality was observed among colony-breeding seabirds in the German Wadden Sea area of the North Sea during the summer months of 2022. Several species' colonies were affected, most notably sandwich terns (Thalasseus sandvicensis), common terns (Sterna hirundo) and Germany's only northern gannet (Morus bassanus) colony on the island of Heligoland. Mortality in some tern colonies reached 40%, while other colonies were almost spared. In all cases, infections with the high-pathogenicity avian influenza virus (HPAIV) subtype H5N1 of clade 2.3.4.4b were identified to have caused the epidemic. Phylogenetic analysis of whole-genome sequences revealed that the outbreaks were dominated by two genotypes, Ger-10-21 N1.2 and Ger-10-21 N1.5, previously identified in Germany. Spatiotemporal analyses of phylogenetic data suggested that these viruses could have entered the continental North Sea coastal region via the British Isles. A close linkage of viruses from tern colonies in the German Wadden Sea was evident with further connections to breeding colonies in Belgium and the Netherlands, and further spread to Denmark and Poland. Several of the affected species are endangered, such that negative effects of epizootic HPAIV infections on populations are feared, with uncertain long-term consequences.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Animales , Gripe Aviar/epidemiología , Subtipo H5N1 del Virus de la Influenza A/genética , Filogenia , Virulencia , Aves , Genotipo
20.
J Gen Virol ; 104(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37622664

RESUMEN

In April 2023, following the annual International Committee on Taxonomy of Viruses (ICTV) ratification vote on newly proposed taxa, the phylum Negarnaviricota was amended and emended. The phylum was expanded by one new family, 14 new genera, and 140 new species. Two genera and 538 species were renamed. One species was moved, and four were abolished. This article presents the updated taxonomy of Negarnaviricota as now accepted by the ICTV.


Asunto(s)
Virus ARN de Sentido Negativo , Virus ARN , Virus ARN/genética , ARN Polimerasa Dependiente del ARN/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda