Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Vet Pathol ; 61(4): 562-573, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38415450

RESUMEN

Lymphoproliferative disease virus (LPDV) was first documented in wild turkeys in North America in 2009. LPDV infection is often subclinical but can manifest as lymphoid proliferation or round cell neoplasia. Despite high prevalence across many sampled areas corresponding to declining populations of wild turkeys, knowledge regarding LPDV pathogenesis, risk factors for disease development, and associated impacts on population dynamics are unknown. To understand transmission, viral shedding, and tissue tropism, we inoculated 21 domestic turkeys via the oral cavity, crop, nasal cavity, subcutis, or coelomic cavity. For 12 weeks, oropharyngeal swabs, cloacal swabs, and whole blood were collected weekly. At 1 week postinoculation, 3 turkeys (3/21; 14%) had detectable LPDV proviral DNA in blood by polymerase chain reaction, and 10 developed DNAemia (50%; 10/20) by 12 weeks. LPDV proviral DNA was intermittently detected in oropharyngeal and cloacal swabs. Splenomegaly was the most consistent gross finding in DNAemic birds (8/11; 73%). Lymphoid hyperplasia in the spleen was the most significant microscopic finding (9/11; 82%). Three turkeys (3/11; 27%) developed round cell neoplasia characterized by sheets of pleomorphic, round to polygonal cells in the adrenal gland, bone marrow, skin, small intestine, and/or spleen. LPDV was detected in the spleen and bone marrow from all turkeys with DNAemia and all neoplasms. Our study establishes that infection and disease with North American LPDV from wild turkeys can be experimentally reproduced in domestic turkeys, laying the groundwork for future investigations into LPDV pathogenesis, development of diagnostic techniques, and understanding the impacts of LPDV on wild turkey populations.


Asunto(s)
Enfermedades de las Aves de Corral , Pavos , Animales , Pavos/virología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/patología , Enfermedades de las Aves de Corral/epidemiología , Trastornos Linfoproliferativos/veterinaria , Trastornos Linfoproliferativos/virología , Trastornos Linfoproliferativos/patología , ADN Viral/genética , Femenino , Infecciones Tumorales por Virus/veterinaria , Infecciones Tumorales por Virus/virología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/epidemiología , Esparcimiento de Virus , América del Norte/epidemiología , Masculino , Infecciones por Retroviridae/veterinaria , Infecciones por Retroviridae/virología , Infecciones por Retroviridae/patología , Bazo/patología , Bazo/virología
2.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31619551

RESUMEN

Canine parvovirus (CPV) is a highly successful pathogen that has sustained pandemic circulation in dogs for more than 40 years. Here, integrating full-genome and deep-sequencing analyses, structural information, and in vitro experimentation, we describe the macro- and microscale features that accompany CPV's evolutionary success. Despite 40 years of viral evolution, all CPV variants are more than ∼99% identical in nucleotide sequence, with only a limited number (<40) of substitutions becoming fixed or widespread during this time. Notably, most substitutions in the major capsid protein (VP2) gene are nonsynonymous, altering amino acid residues that fall within, or adjacent to, the overlapping receptor footprint or antigenic regions, suggesting that natural selection has channeled much of CPV evolution. Among the limited number of variable sites, CPV genomes exhibit complex patterns of variation that include parallel evolution, reversion, and recombination, compromising phylogenetic inference. At the intrahost level, deep sequencing of viral DNA in original clinical samples from dogs and other host species sampled between 1978 and 2018 revealed few subconsensus single nucleotide variants (SNVs) above ∼0.5%, and experimental passages demonstrate that substantial preexisting genetic variation is not necessarily required for rapid host receptor-driven adaptation. Together, these findings suggest that although CPV is capable of rapid host adaptation, a relatively low mutation rate, pleiotropy, and/or a lack of selective challenges since its initial emergence have inhibited the long-term accumulation of genetic diversity. Hence, continuously high levels of inter- and intrahost diversity are not necessarily required for virus host adaptation.IMPORTANCE Rapid mutation rates and correspondingly high levels of intra- and interhost diversity are often cited as key features of viruses with the capacity for emergence and sustained transmission in a new host species. However, most of this information comes from studies of RNA viruses, with relatively little known about evolutionary processes in viruses with single-stranded DNA (ssDNA) genomes. Here, we provide a unique model of virus evolution, integrating both long-term global-scale and short-term intrahost evolutionary processes of an ssDNA virus that emerged to cause a pandemic in a new host animal. Our analysis reveals that successful host jumping and sustained transmission does not necessarily depend on a high level of intrahost diversity nor result in the continued accumulation of high levels of long-term evolution change. These findings indicate that all aspects of the biology and ecology of a virus are relevant when considering their adaptability.


Asunto(s)
Proteínas de la Cápside/genética , ADN Viral/genética , Enfermedades de los Perros/epidemiología , Genoma Viral , Infecciones por Parvoviridae/veterinaria , Parvovirus Canino/genética , Proteínas no Estructurales Virales/genética , Adaptación Fisiológica/genética , Animales , Evolución Biológica , Proteínas de la Cápside/clasificación , Proteínas de la Cápside/metabolismo , ADN Viral/metabolismo , Enfermedades de los Perros/transmisión , Enfermedades de los Perros/virología , Perros , Zorros/virología , Especificidad del Huésped/genética , Modelos Moleculares , Mutación , Infecciones por Parvoviridae/epidemiología , Infecciones por Parvoviridae/transmisión , Infecciones por Parvoviridae/virología , Parvovirus Canino/clasificación , Parvovirus Canino/patogenicidad , Filogenia , Conformación Proteica , Perros Mapache/virología , Mapaches/virología , Proteínas no Estructurales Virales/clasificación , Proteínas no Estructurales Virales/metabolismo , Secuenciación Completa del Genoma
3.
J Virol ; 92(13)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29695427

RESUMEN

Antibody and receptor binding are key virus-host interactions that control host range and determine the success of infection. Canine and feline parvovirus capsids bind the transferrin receptor type 1 (TfR) to enter host cells, and specific structural interactions appear necessary to prepare the stable capsids for infection. Here, we define the details of binding, competition, and occupancy of wild-type and mutant parvovirus capsids with purified receptors and antibodies. TfR-capsid binding interactions depended on the TfR species and varied widely, with no direct relationship between binding affinity and infection. Capsids bound feline, raccoon, and black-backed jackal TfRs at high affinity but barely bound canine TfRs, which mediated infection efficiently. TfRs from different species also occupied capsids to different levels, with an estimated 1 to 2 feline TfRs but 12 black-backed jackal TfRs binding each capsid. Multiple alanine substitutions within loop 1 on the capsid surface reduced TfR binding but substitutions within loop 3 did not, suggesting that loop 1 directly engaged the TfR and loop 3 sterically affected that interaction. Binding and competition between different TfRs and/or antibodies showed complex relationships. Both antibodies 14 and E competed capsids off TfRs, but antibody E could also compete capsids off itself and antibody 14, likely by inducing capsid structural changes. In some cases, the initial TfR or antibody binding event affected subsequent TfR binding, suggesting that capsid structure changes occur after TfR or antibody binding and may impact infection. This shows that precise, host-specific TfR-capsid interactions, beyond simple attachment, are important for successful infection.IMPORTANCE Host receptor binding is a key step during viral infection and may control both infection and host range. In addition to binding, some viruses require specific interactions with host receptors in order to infect, and anti-capsid antibodies can potentially disrupt these interactions, leading to neutralization. Here, we examine the interactions between parvovirus capsids, the receptors from different hosts, and anti-capsid antibodies. We show that interactions between parvovirus capsids and host-specific TfRs vary in both affinity and in the numbers of receptors bound, with complex effects on infection. In addition, antibodies binding to two sites on the capsids had different effects on TfR-capsid binding. These experiments confirm that receptor and antibody binding to parvovirus capsids are complex processes, and the infection outcome is not determined simply by the affinity of attachment.


Asunto(s)
Anticuerpos Antivirales/metabolismo , Cápside/metabolismo , Mutación , Parvovirus/patogenicidad , Receptores de Transferrina/metabolismo , Animales , Cápside/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Gatos , Línea Celular , Perros , Especificidad del Huésped , Humanos , Chacales , Modelos Moleculares , Parvovirus/inmunología , Mapaches , Receptores de Transferrina/química
4.
J Virol ; 91(2)2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807242

RESUMEN

Since the first description of adenoviruses in bats in 2006, a number of micro- and megabat species in Europe, Africa, and Asia have been shown to carry a wide diversity of adenoviruses. Here, we report on the evolutionary, biological, and structural characterization of a novel bat adenovirus (BtAdV) recovered from a Rafinesque's big-eared bat (Corynorhinus rafinesquii) in Kentucky, USA, which is the first adenovirus isolated from North American bats. This virus (BtAdV 250-A) exhibits a close phylogenetic relationship with Canine mastadenovirus A (CAdV A), as previously observed with other BtAdVs. To further investigate the relationships between BtAdVs and CAdVs, we conducted mass spectrometric analysis and single-particle cryo-electron microscopy reconstructions of the BtAdV 250-A capsid and also analyzed the in vitro host ranges of both viruses. Our results demonstrate that BtAdV 250-A represents a new mastadenovirus species that, in contrast to CAdV, has a unique capsid morphology that contains more prominent extensions of protein IX and can replicate efficiently in a phylogenetically diverse range of species. These findings, in addition to the recognition that both the genetic diversity of BtAdVs and the number of different bat species from disparate geographic regions infected with BtAdVs appears to be extensive, tentatively suggest that bats may have served as a potential reservoir for the cross-species transfer of adenoviruses to other hosts, as theorized for CAdV. IMPORTANCE: Although many adenoviruses are host specific and likely codiverged with their hosts over millions of years, other adenoviruses appear to have emerged through successful cross-species transmission events on more recent time scales. The wide geographic distribution and genetic diversity of adenoviruses in bats and their close phylogenetic relationship to Canine mastadenovirus A (CAdV A) has raised important questions about how CAdV A, and possibly other mammalian adenoviruses, may have emerged. Although most adenoviruses tend to cause limited disease in their natural hosts, CAdV A is unusual in that it may cause high morbidity and sometimes fatal infections in immunocompetent hosts and is thus an important pathogen of carnivores. Here, we performed a comparative evolutionary and structural study of representative bat and canine adenoviruses to better understand the relationship between these two viral groups.


Asunto(s)
Infecciones por Adenoviridae/transmisión , Infecciones por Adenoviridae/virología , Evolución Biológica , Cápside/metabolismo , Cápside/ultraestructura , Microscopía por Crioelectrón , Mastadenovirus/fisiología , Mastadenovirus/ultraestructura , Animales , Quirópteros , Perros , Orden Génico , Genoma Viral , Especificidad del Huésped , Espectrometría de Masas , Mastadenovirus/clasificación , Sistemas de Lectura Abierta , Filogenia , ARN Viral , Homología de Secuencia , Virión
5.
J Virol ; 91(2)2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27847360

RESUMEN

Parvovirus capsids are small but complex molecular machines responsible for undertaking many of the steps of cell infection, genome packing, and cell-to-cell as well as host-to-host transfer. The details of parvovirus infection of cells are still not fully understood, but the processes must involve small changes in the capsid structure that allow the endocytosed virus to escape from the endosome, pass through the cell cytoplasm, and deliver the single-stranded DNA (ssDNA) genome to the nucleus, where viral replication occurs. Here, we examine capsid substitutions that eliminate canine parvovirus (CPV) infectivity and identify how those mutations changed the capsid structure or altered interactions with the infectious pathway. Amino acid substitutions on the exterior surface of the capsid (Gly299Lys/Ala300Lys) altered the binding of the capsid to transferrin receptor type 1 (TfR), particularly during virus dissociation from the receptor, but still allowed efficient entry into both feline and canine cells without successful infection. These substitutions likely control specific capsid structural changes resulting from TfR binding required for infection. A second set of changes on the interior surface of the capsid reduced viral infectivity by >100-fold and included two cysteine residues and neighboring residues. One of these substitutions, Cys270Ser, modulates a VP2 cleavage event found in ∼10% of the capsid proteins that also was shown to alter capsid stability. A neighboring substitution, Pro272Lys, significantly reduced capsid assembly, while a Cys273Ser change appeared to alter capsid transport from the nucleus. These mutants reveal additional structural details that explain cell infection processes of parvovirus capsids. IMPORTANCE: Parvoviruses are commonly found in both vertebrate and invertebrate animals and cause widespread disease. They are also being developed as oncolytic therapeutics and as gene therapy vectors. Most functions involved in infection or transduction are mediated by the viral capsid, but the structure-function correlates of the capsids and their constituent proteins are still incompletely understood, especially in relation to identifying capsid processes responsible for infection and release from the cell. Here, we characterize the functional effects of capsid protein mutations that result in the loss of virus infectivity, giving a better understanding of the portions of the capsid that mediate essential steps in successful infection pathways and how they contribute to viral infectivity.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Infecciones por Parvoviridae/virología , Parvovirus/fisiología , Conformación Proteica , Secuencia de Aminoácidos , Proteínas de la Cápside/genética , Endopeptidasas/metabolismo , Interacciones Huésped-Patógeno , Modelos Moleculares , Mutación , Transporte de Proteínas , Proteolisis , Receptores Virales/metabolismo , Relación Estructura-Actividad , Acoplamiento Viral
6.
Emerg Infect Dis ; 23(12): 1958-1965, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28841405

RESUMEN

Wellfleet Bay virus (WFBV), a novel orthomyxovirus in the genus Quaranjavirus, was first isolated in 2006 from carcasses of common eider (Somateria mollissima) during a mortality event in Wellfleet Bay (Barnstable County, Massachusetts, USA) and has since been repeatedly isolated during recurrent mortality events in this location. Hepatic, pancreatic, splenic, and intestinal necrosis was observed in dead eiders. We inoculated 6-week-old common eider ducklings with WFBV in an attempt to recreate the naturally occurring disease. Approximately 25% of inoculated eiders had onset of clinical disease and required euthanasia; an additional 18.75% were adversely affected based on net weight loss during the trial. Control ducklings did not become infected and did not have clinical disease. Infected ducklings with clinical disease had pathologic lesions consistent with those observed during natural mortality events. WFBV was reisolated from 37.5% of the inoculated ducklings. Ducklings surviving to 5 days postinoculation developed serum antibody titers to WFBV.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Enfermedades de las Aves/virología , Patos/virología , Necrosis/veterinaria , Infecciones por Orthomyxoviridae/veterinaria , Orthomyxoviridae/fisiología , Animales , Bahías , Enfermedades de las Aves/inmunología , Enfermedades de las Aves/patología , Modelos Animales de Enfermedad , Patos/inmunología , Intestinos/inmunología , Intestinos/patología , Intestinos/virología , Hígado/inmunología , Hígado/patología , Hígado/virología , Massachusetts , Necrosis/inmunología , Necrosis/patología , Necrosis/virología , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Páncreas/inmunología , Páncreas/patología , Páncreas/virología , Bazo/inmunología , Bazo/patología , Bazo/virología , Pérdida de Peso
7.
J Virol ; 90(9): 4849-53, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26889026

RESUMEN

Determining how viruses infect new hosts via receptor-binding mechanisms is important for understanding virus emergence. We studied the binding kinetics of canine parvovirus (CPV) variants isolated from raccoons-a newly recognized CPV host-to different carnivore transferrin receptors (TfRs) using single-particle tracking. Our data suggest that CPV may utilize adhesion-strengthening mechanisms during TfR binding and that a single mutation in the viral capsid at VP2 position 300 can profoundly alter receptor binding and infectivity.


Asunto(s)
Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Parvovirus Canino/fisiología , Mutación Puntual , Receptores de Transferrina/metabolismo , Tropismo Viral , Animales , Proteínas de la Cápside/química , Línea Celular , Perros , Cinética , Microscopía Fluorescente/métodos , Imagen Molecular/métodos , Unión Proteica , Mapaches , Coloración y Etiquetado
8.
J Virol ; 90(2): 753-67, 2016 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-26512077

RESUMEN

UNLABELLED: Sylvatic carnivores, such as raccoons, have recently been recognized as important hosts in the evolution of canine parvovirus (CPV), a pandemic pathogen of domestic dogs. Although viruses from raccoons do not efficiently bind the dog transferrin receptor (TfR) or infect dog cells, a single mutation changing an aspartic acid to a glycine at capsid (VP2) position 300 in the prototype raccoon CPV allows dog cell infection. Because VP2 position 300 exhibits extensive amino acid variation among the carnivore parvoviruses, we further investigated its role in determining host range by analyzing its diversity and evolution in nature and by creating a comprehensive set of VP2 position 300 mutants in infectious clones. Notably, some position 300 residues rendered CPV noninfectious for dog, but not cat or fox, cells. Changes of adjacent residues (residues 299 and 301) were also observed often after cell culture passage in different hosts, and some of the mutations mimicked changes seen in viruses recovered from natural infections of alternative hosts, suggesting that compensatory mutations were selected to accommodate the new residue at position 300. Analysis of the TfRs of carnivore hosts used in the experimental evolution studies demonstrated that their glycosylation patterns varied, including a glycan present only on the domestic dog TfR that dictates susceptibility to parvoviruses. Overall, there were significant differences in the abilities of viruses with alternative position 300 residues to bind TfRs and infect different carnivore hosts, demonstrating that the process of infection is highly host dependent and that VP2 position 300 is a key determinant of host range. IMPORTANCE: Although the emergence and pandemic spread of canine parvovirus (CPV) are well documented, the carnivore hosts and evolutionary pathways involved in its emergence remain enigmatic. We recently demonstrated that a region in the capsid structure of CPV, centered around VP2 position 300, varies after transfer to alternative carnivore hosts and may allow infection of previously nonsusceptible hosts in vitro. Here we show that VP2 position 300 is the most variable residue in the parvovirus capsid in nature, suggesting that it is a critical determinant in the cross-species transfer of viruses between different carnivores due to its interactions with the transferrin receptor to mediate infection. To this end, we demonstrated that there are substantial differences in receptor binding and infectivity of various VP2 position 300 mutants for different carnivore species and that single mutations in this region can influence whether a host is susceptible or refractory to virus infection.


Asunto(s)
Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Especificidad del Huésped , Mutación Missense , Parvovirus Canino/fisiología , Animales , Gatos , Línea Celular , Perros , Zorros , Glicosilación , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Polisacáridos/análisis , Receptores de Transferrina/química , Receptores de Transferrina/metabolismo , Pase Seriado
9.
J Virol ; 89(3): 1909-12, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25410876

RESUMEN

Canine parvovirus type 2 (CPV-2) emerged in 1978 and spread worldwide within 2 years. Subsequently, CPV-2 was completely replaced by the variant CPV-2a, which is characterized by four specific capsid (VP2) mutations. The X-ray crystal structure of the CPV-2a capsid shows that each mutation confers small local changes. The loss of a hydrogen bond and introduction of a glycine residue likely introduce flexibility to sites that control interactions with the host receptor, antibodies, and sialic acids.


Asunto(s)
Enfermedades de los Perros/virología , Especificidad del Huésped , Infecciones por Parvoviridae/veterinaria , Parvovirus Canino/fisiología , Animales , Proteínas de la Cápside/química , Cristalografía por Rayos X , Enfermedades de los Perros/epidemiología , Perros , Modelos Moleculares , Proteínas Mutantes/química , Pandemias , Infecciones por Parvoviridae/epidemiología , Infecciones por Parvoviridae/virología , Parvovirus Canino/química , Parvovirus Canino/aislamiento & purificación , Conformación Proteica
10.
J Virol ; 89(2): 1389-403, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392223

RESUMEN

UNLABELLED: Since 1998, cyclic mortality events in common eiders (Somateria mollissima), numbering in the hundreds to thousands of dead birds, have been documented along the coast of Cape Cod, MA, USA. Although longitudinal disease investigations have uncovered potential contributing factors responsible for these outbreaks, detecting a primary etiological agent has proven enigmatic. Here, we identify a novel orthomyxovirus, tentatively named Wellfleet Bay virus (WFBV), as a potential causative agent of these outbreaks. Genomic analysis of WFBV revealed that it is most closely related to members of the Quaranjavirus genus within the family Orthomyxoviridae. Similar to other members of the genus, WFBV contains an alphabaculovirus gp64-like glycoprotein that was demonstrated to have fusion activity; this also tentatively suggests that ticks (and/or insects) may vector the virus in nature. However, in addition to the six RNA segments encoding the prototypical structural proteins identified in other quaranjaviruses, a previously unknown RNA segment (segment 7) encoding a novel protein designated VP7 was discovered in WFBV. Although WFBV shows low to moderate levels of sequence similarity to Quaranfil virus and Johnston Atoll virus, the original members of the Quaranjavirus genus, additional antigenic and genetic analyses demonstrated that it is closely related to the recently identified Cygnet River virus (CyRV) from South Australia, suggesting that WFBV and CyRV may be geographic variants of the same virus. Although the identification of WFBV in part may resolve the enigma of these mass mortality events, the details of the ecology and epidemiology of the virus remain to be determined. IMPORTANCE: The emergence or reemergence of viral pathogens resulting in large-scale outbreaks of disease in humans and/or animals is one of the most important challenges facing biomedicine. For example, understanding how orthomyxoviruses such as novel influenza A virus reassortants and/or mutants emerge to cause epidemic or pandemic disease is at the forefront of current global health concerns. Here, we describe the emergence of a novel orthomyxovirus, Wellfleet Bay virus (WFBV), which has been associated with cyclic large-scale bird die-offs in the northeastern United States. This initial characterization study provides a foundation for further research into the evolution, epidemiology, and ecology of newly emerging orthomyxoviruses, such as WFBV, and their potential impacts on animal and/or human health.


Asunto(s)
Enfermedades de las Aves/epidemiología , Enfermedades de las Aves/mortalidad , Brotes de Enfermedades , Infecciones por Orthomyxoviridae/epidemiología , Infecciones por Orthomyxoviridae/mortalidad , Orthomyxoviridae/aislamiento & purificación , Animales , Anseriformes , Enfermedades de las Aves/patología , Enfermedades de las Aves/virología , Análisis por Conglomerados , Femenino , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , New England/epidemiología , Orthomyxoviridae/clasificación , Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Filogenia , Conformación Proteica , ARN Viral/genética , Análisis de Secuencia de ADN , Proteínas Virales/química , Proteínas Virales/genética
11.
PLoS Pathog ; 10(11): e1004475, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375184

RESUMEN

Canine parvovirus (CPV) emerged as a new pandemic pathogen of dogs in the 1970s and is closely related to feline panleukopenia virus (FPV), a parvovirus of cats and related carnivores. Although both viruses have wide host ranges, analysis of viral sequences recovered from different wild carnivore species, as shown here, demonstrated that>95% were derived from CPV-like viruses, suggesting that CPV is dominant in sylvatic cycles. Many viral sequences showed host-specific mutations in their capsid proteins, which were often close to sites known to control binding to the transferrin receptor (TfR), the host receptor for these carnivore parvoviruses, and which exhibited frequent parallel evolution. To further examine the process of host adaptation, we passaged parvoviruses with alternative backgrounds in cells from different carnivore hosts. Specific mutations were selected in several viruses and these differed depending on both the background of the virus and the host cells in which they were passaged. Strikingly, these in vitro mutations recapitulated many specific changes seen in viruses from natural populations, strongly suggesting they are host adaptive, and which were shown to result in fitness advantages over their parental virus. Comparison of the sequences of the transferrin receptors of the different carnivore species demonstrated that many mutations occurred in and around the apical domain where the virus binds, indicating that viral variants were likely selected through their fit to receptor structures. Some of the viruses accumulated high levels of variation upon passage in alternative hosts, while others could infect multiple different hosts with no or only a few additional mutations. Overall, these studies demonstrate that the evolutionary history of a virus, including how long it has been circulating and in which hosts, as well as its phylogenetic background, has a profound effect on determining viral host range.


Asunto(s)
Evolución Molecular , Interacciones Huésped-Patógeno/fisiología , Parvovirus Canino/fisiología , Animales , Gatos , Perros , Especificidad de la Especie
12.
J Med Entomol ; 52(5): 1050-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26336204

RESUMEN

Replication of arboviruses, including orbiviruses, within the vector has been shown to be temperature dependent. Cooler ambient temperatures slow virus replication in arthropod vectors, whereas viruses replicate faster and to higher titers at warmer ambient temperatures. Previous research with epizootic hemorrhagic disease virus (EHDV) serotype 1 demonstrated that higher temperatures were associated with shorter extrinsic incubation periods in Culicoides sonorensis Wirth & Jones, a confirmed vector of EHDV in North America. To further our understanding of the effect of temperature on replication of EHDV within the vector, C. sonorensis were experimentally infected with one of three EHDV strains representing three serotypes (1, 2, and 7). Midges were fed defibrinated white-tailed deer (Odocoileus virginianus) blood spiked with EHDV (≥10(6.5) TCID(50)/ml) through a parafilm membrane using an artificial feeding device and were then held at 20, 25, or 30°C. In addition to this in vitro method, a white-tailed deer experimentally infected with EHDV-7 was used to provide an infectious bloodmeal to determine if the results were comparable with those from the in vitro feeding method. Whole midges were processed for virus isolation and titration at regular intervals following feeding; midges with ≥10(2.7) TCID(50) were considered potentially competent to transmit virus. The virus recovery rates were high throughout the study and all three viruses replicated within C. sonorensis to high titer (≥ 10(2.7) TCID(50)/midge). Across all virus strains, the time to detection of potentially competent midges decreased with increasing temperature: 12-16 d postfeeding (dpf) at 20°C, 4-6 dpf at 25°C, and 2-4 dpf at 30°C. Significant differences in replication of the three viruses in C. sonorensis were observed, with EHDV-2 replicating to a high titer in a smaller proportion of midges and with lower peak titers. The findings are consistent with previous studies of related orbiviruses, showing that increasing temperature can shorten the apparent extrinsic incubation period for multiple EHDV strains (endemic and exotic) in C. sonorensis.


Asunto(s)
Ceratopogonidae/virología , Virus de la Enfermedad Hemorrágica Epizoótica/fisiología , Replicación Viral , Animales , Ciervos/parasitología , Ciervos/virología , Virus de la Enfermedad Hemorrágica Epizoótica/genética , Serogrupo , Temperatura
13.
Am J Primatol ; 77(10): 1075-85, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26119266

RESUMEN

Sexually transmitted diseases (STDs) can persist endemically, are known to cause sterility and infant mortality in humans, and could have similar impacts in wildlife populations. African apes (i.e., chimpanzees, bonobos, and to a lesser extent gorillas) show multi-male mating behavior that could offer opportunities for STD transmission, yet little is known about the prevalence and impact of STDs in this endangered primate group. We used serology and PCR-based detection methods to screen biological samples from wild and orphaned eastern chimpanzees and gorillas (N = 172 individuals, including adults, and juveniles) for four classes of pathogens that either commonly cause human STDs or were previously detected in captive apes: trichomonads, Chlamydia spp., Treponema pallidum (syphilis and yaws), and papillomaviruses. Based on results from prior modeling and comparative research, we expected STD prevalence to be highest in females versus males and in sexually mature versus immature individuals. All samples were negative for Chlamydia, Treponema pallidum, and papillomaviruses; however, a high percentage of wild chimpanzee urine and fecal samples showed evidence of trichomonads (protozoa). Analysis revealed that females were more likely than males to have positive urine-but not fecal-samples; however, there was no evidence of age (sexual maturity) differences in infection status. Sequence analysis of chimpanzee trichomonad samples revealed a close relationship to previously described trichomonads within the genus Tetratrichomonas. Phylogenetic comparisons to archived sequences from multiple vertebrate hosts suggests that many of the chimpanzee parasites from our study are likely transmitted via fecal-oral contact, but the transmission of some Tetratrichomonas sequence-types remains unknown and could include sexual contact. Our work emphasizes that only a fraction of infectious agents affecting wild apes are presently known to science, and that further work on great ape STDs could offer insights for the management of endangered great apes and for understanding human STD origins.


Asunto(s)
Chlamydia/aislamiento & purificación , Papillomaviridae/aislamiento & purificación , Enfermedades de los Primates/parasitología , Enfermedades de Transmisión Sexual/veterinaria , Treponema pallidum/aislamiento & purificación , Trichomonadida/aislamiento & purificación , Animales , Heces/parasitología , Femenino , Gorilla gorilla , Masculino , Pan troglodytes , Prevalencia , Enfermedades de los Primates/microbiología , Enfermedades de los Primates/virología , Infecciones Protozoarias en Animales , Factores Sexuales , Orina/parasitología
14.
J Virol ; 87(4): 2342-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221559

RESUMEN

Although parvoviruses are commonly described in domestic carnivores, little is known about their biodiversity in nondomestic species. A phylogenetic analysis of VP2 gene sequences from puma, coyote, gray wolf, bobcat, raccoon, and striped skunk revealed two major groups related to either feline panleukopenia virus ("FPV-like") or canine parvovirus ("CPV-like"). Cross-species transmission was commonplace, with multiple introductions into each host species but, with the exception of raccoons, relatively little evidence for onward transmission in nondomestic species.


Asunto(s)
Carnívoros/virología , Variación Genética , Infecciones por Parvoviridae/veterinaria , Parvovirus/clasificación , Parvovirus/aislamiento & purificación , Animales , Proteínas de la Cápside/genética , Análisis por Conglomerados , ADN Viral/genética , Datos de Secuencia Molecular , Infecciones por Parvoviridae/transmisión , Parvovirus/genética , Filogenia , Análisis de Secuencia de ADN
15.
PLoS Pathog ; 8(5): e1002666, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22570610

RESUMEN

Parvoviruses exploit transferrin receptor type-1 (TfR) for cellular entry in carnivores, and specific interactions are key to control of host range. We show that several key mutations acquired by TfR during the evolution of Caniforms (dogs and related species) modified the interactions with parvovirus capsids by reducing the level of binding. These data, along with signatures of positive selection in the TFRC gene, are consistent with an evolutionary arms race between the TfR of the Caniform clade and parvoviruses. As well as the modifications of amino acid sequence which modify binding, we found that a glycosylation site mutation in the TfR of dogs which provided resistance to the carnivore parvoviruses which were in circulation prior to about 1975 predates the speciation of coyotes and dogs. Because the closely-related black-backed jackal has a TfR similar to their common ancestor and lacks the glycosylation site, reconstructing this mutation into the jackal TfR shows the potency of that site in blocking binding and infection and explains the resistance of dogs until recent times. This alters our understanding of this well-known example of viral emergence by indicating that canine parvovirus emergence likely resulted from the re-adaptation of a parvovirus to the resistant receptor of a former host.


Asunto(s)
Canidae/genética , Infecciones por Parvoviridae/veterinaria , Parvovirus Canino/genética , Parvovirus Canino/patogenicidad , Receptores de Transferrina/genética , Receptores Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Evolución Biológica , Células CHO , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Cricetinae , Enfermedades de los Perros/virología , Perros/genética , Glicosilación , Interacciones Huésped-Patógeno , Mutación , Infecciones por Parvoviridae/virología , Parvovirus Canino/metabolismo , Filogenia , Unión Proteica , Receptores de Transferrina/química , Receptores de Transferrina/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Selección Genética , Análisis de Secuencia de ADN , Especificidad de la Especie , Transferrina/metabolismo
16.
Commun Biol ; 7(1): 476, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637646

RESUMEN

Since late 2021, highly pathogenic avian influenza (HPAI) viruses of A/goose/Guangdong/1/1996 (H5N1) lineage have caused widespread mortality in wild birds and poultry in the United States. Concomitant with the spread of HPAI viruses in birds are increasing numbers of mammalian infections, including wild and captive mesocarnivores and carnivores with central nervous system involvement. Here we report HPAI, A(H5N1) of clade 2.3.4.4b, in a common bottlenose dolphin (Tursiops truncatus) from Florida, United States. Pathological findings include neuronal necrosis and inflammation of the brain and meninges, and quantitative real time RT-PCR reveal the brain carried the highest viral load. Virus isolated from the brain contains a S246N neuraminidase substitution which leads to reduced inhibition by neuraminidase inhibitor oseltamivir. The increased prevalence of A(H5N1) viruses in atypical avian hosts and its cross-species transmission into mammalian species highlights the public health importance of continued disease surveillance and biosecurity protocols.


Asunto(s)
Delfín Mular , Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Animales , Gripe Aviar/epidemiología , Subtipo H5N1 del Virus de la Influenza A/genética , Florida/epidemiología , Neuraminidasa , Virus de la Influenza A/fisiología , Aves
17.
J Virol ; 86(2): 865-72, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22072763

RESUMEN

Understanding the mechanisms of cross-species virus transmission is critical to anticipating emerging infectious diseases. Canine parvovirus type 2 (CPV-2) emerged as a variant of a feline parvovirus when it acquired mutations that allowed binding to the canine transferrin receptor type 1 (TfR). However, CPV-2 was soon replaced by a variant virus (CPV-2a) that differed in antigenicity and receptor binding. Here we show that the emergence of CPV involved an additional host range variant virus that has circulated undetected in raccoons for at least 24 years, with transfers to and from dogs. Raccoon virus capsids showed little binding to the canine TfR, showed little infection of canine cells, and had altered antigenic structures. Remarkably, in capsid protein (VP2) phylogenies, most raccoon viruses fell as evolutionary intermediates between the CPV-2 and CPV-2a strains, suggesting that passage through raccoons assisted in the evolution of CPV-2a. This highlights the potential role of alternative hosts in viral emergence.


Asunto(s)
Transmisión de Enfermedad Infecciosa/veterinaria , Especificidad del Huésped , Pandemias/veterinaria , Infecciones por Parvoviridae/veterinaria , Parvovirus/fisiología , Mapaches/virología , Animales , Evolución Biológica , Proteínas de la Cápside/genética , Gatos , Línea Celular , Perros , Datos de Secuencia Molecular , Infecciones por Parvoviridae/epidemiología , Infecciones por Parvoviridae/transmisión , Infecciones por Parvoviridae/virología , Parvovirus/clasificación , Parvovirus/genética , Parvovirus/aislamiento & purificación , Filogenia , Estados Unidos/epidemiología
19.
Virology ; 571: 1-11, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35421704

RESUMEN

Bats have recently been identified as potential reservoir hosts for mammalian orthoreoviruses (MRVs) throughout Europe and China. Here we present the first evolutionary and biological characterization of bat-borne MRVs in North America, including phylogenomic analysis, in vitro relative infectivity in bat and other mammalian cell cultures, host cell receptor specificity, and epifluorescence microscopy of viral factory formation. Through genetic and phylogenetic comparisons, we show that two divergent MRV serotype 2 (T2) strains - isolated from a silver-haired bat (Lasionycteris noctivagans) and a big brown bat (Eptesicus fuscus) from Pennsylvania, USA - provide an evolutionary link to an MRV strain (T2W) recovered from an 8-week-old infant who died in Winnipeg, Manitoba, Canada in 1997. Although these findings suggest North American bats may represent a previously unrecognized source for the cross-species transmission of MRVs to other animals, including humans, the ecology and epidemiology of MRVs in wildlife remain enigmatic.


Asunto(s)
Quirópteros , Orthoreovirus de los Mamíferos , Animales , Animales Salvajes , Especificidad del Huésped , Humanos , Orthoreovirus de los Mamíferos/genética , Filogenia
20.
Parasitol Res ; 105(3): 775-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19452169

RESUMEN

To determine if double-stranded RNA (dsRNA) viruses exist and are potential virulence factors in Trichomonas gallinae, virus purification via ultracentrifugation was attempted for 12 T. gallinae isolates recovered from wild birds. Following purification, virus-like particles were not observed by transmission electron microscopy, nor were dsRNA segments visualized in agarose gels after electrophoresis of extracted RNA from any of the 12 T. gallinae isolates. However, virus particles and dsRNA segments were detected from a previously determined virus-infected T. vaginalis isolate as a control using identical purification procedures. Subsequent reverse transcription-polymerase chain reaction analysis of the dsRNA of the virus in this isolate revealed a novel sequence of the RNA-dependent RNA polymerase gene of T. vaginalis viruses.


Asunto(s)
Virus ARN/genética , Virus ARN/aislamiento & purificación , ARN Bicatenario/genética , Trichomonas/virología , Animales , Aves/parasitología , Análisis por Conglomerados , Electroforesis en Gel de Agar/métodos , Microscopía Electrónica de Transmisión/métodos , Datos de Secuencia Molecular , Filogenia , Virus ARN/clasificación , ARN Bicatenario/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Homología de Secuencia , Trichomonas/aislamiento & purificación , Ultracentrifugación/métodos , Virión/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA