Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 15(4): e0049924, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38470055

RESUMO

Rotavirus (RV) replication takes place in the viroplasms, cytosolic inclusions that allow the synthesis of virus genome segments and their encapsidation in the core shell, followed by the addition of the second layer of the virion. The viroplasms are composed of several viral proteins, including NSP5, which serves as the main building block. Microtubules, lipid droplets, and miRNA-7 are among the host components recruited in viroplasms. We investigated the interaction between RV proteins and host components of the viroplasms by performing a pull-down assay of lysates from RV-infected cells expressing NSP5-BiolD2. Subsequent tandem mass spectrometry identified all eight subunits of the tailless complex polypeptide I ring complex (TRiC), a cellular chaperonin responsible for folding at least 10% of the cytosolic proteins. Our confirmed findings reveal that TRiC is brought into viroplasms and wraps around newly formed double-layered particles. Chemical inhibition of TRiC and silencing of its subunits drastically reduced virus progeny production. Through direct RNA sequencing, we show that TRiC is critical for RV replication by controlling dsRNA genome segment synthesis, particularly negative-sense single-stranded RNA. Importantly, cryo-electron microscopy analysis shows that TRiC inhibition results in defective virus particles lacking genome segments and polymerase complex (VP1/VP3). Moreover, TRiC associates with VP2 and NSP5 but not with VP1. Also, VP2 is shown to be essential for recruiting TRiC in viroplasms and preserving their globular morphology. This study highlights the essential role of TRiC in viroplasm formation and in facilitating virion assembly during the RV life cycle. IMPORTANCE: The replication of rotavirus takes place in cytosolic inclusions termed viroplasms. In these inclusions, the distinct 11 double-stranded RNA genome segments are co-packaged to complete a genome in newly generated virus particles. In this study, we show for the first time that the tailless complex polypeptide I ring complex (TRiC), a cellular chaperonin responsible for the folding of at least 10% of the cytosolic proteins, is a component of viroplasms and is required for the synthesis of the viral negative-sense single-stranded RNA. Specifically, TRiC associates with NSP5 and VP2, the cofactor involved in RNA replication. Our study adds a new component to the current model of rotavirus replication, where TRiC is recruited to viroplasms to assist replication.


Assuntos
Rotavirus , Rotavirus/genética , Compartimentos de Replicação Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Microscopia Crioeletrônica , Replicação Viral/fisiologia , RNA , Peptídeos
2.
Viruses ; 13(7)2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34372610

RESUMO

There is growing evidence that equine papillomavirus type 2 (EcPV2) infection is etiologically associated with the development of genital squamous cell carcinoma (SCC) and precursor lesions in equids. However, the precise mechanisms underlying neoplastic progression remain unknown. To allow the study of EcPV2-induced carcinogenesis, we aimed to establish a primary equine cell culture model of EcPV2 infection. Three-dimensional (3D) raft cultures were generated from equine penile perilesional skin, plaques and SCCs. Using histological, molecular biological and immunohistochemical methods, rafts versus corresponding natural tissue sections were compared with regard to morphology, presence of EcPV2 DNA, presence and location of EcPV2 gene transcripts and expression of epithelial, mesenchymal and tumor/proliferation markers. Raft cultures from perilesional skin harboring only a few EcPV2-positive (EcPV2+) cells accurately recapitulated the differentiation process of normal skin, whilst rafts from EcPV2+ penile plaques were structurally organized but showed early hyperplasia. Rafts from EcPV2+ SCCs exhibited pronounced hyperplasia and marked dysplasia. Raft levels of EcPV2 oncogene transcription (E6/E7) and expression of tumor/proliferation markers p53, Ki67 and MCM7 expression positively correlated with neoplastic progression, again reflecting the natural situation. Three-dimensional raft cultures accurately reflected major features of corresponding ex vivo material, thus constituting a valuable new research model to study EcPV2-induced carcinogenesis.


Assuntos
Técnicas de Cultura de Células/métodos , Hiperplasia/veterinária , Papillomaviridae/genética , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/veterinária , Pênis/citologia , Animais , Carcinogênese , Carcinoma de Células Escamosas/virologia , DNA Viral/genética , Doenças dos Cavalos/virologia , Cavalos , Hiperplasia/virologia , Masculino , Papillomaviridae/classificação , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/virologia , Pênis/virologia
3.
PLoS Pathog ; 17(6): e1009638, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34061891

RESUMO

Adeno-associated virus (AAV) genome replication only occurs in the presence of a co-infecting helper virus such as adenovirus type 5 (AdV5) or herpes simplex virus type 1 (HSV-1). AdV5-supported replication of the AAV genome has been described to occur in a strand-displacement rolling hairpin replication (RHR) mechanism initiated at the AAV 3' inverted terminal repeat (ITR) end. It has been assumed that the same mechanism applies to HSV-1-supported AAV genome replication. Using Southern analysis and nanopore sequencing as a novel, high-throughput approach to study viral genome replication we demonstrate the formation of double-stranded head-to-tail concatemers of AAV genomes in the presence of HSV-1, thus providing evidence for an unequivocal rolling circle replication (RCR) mechanism. This stands in contrast to the textbook model of AAV genome replication when HSV-1 is the helper virus.


Assuntos
Coinfecção , Dependovirus , Simplexvirus , Replicação Viral , Animais , Linhagem Celular , Genoma Viral , Vírus Auxiliares/fisiologia , Herpes Simples , Humanos , Infecções por Parvoviridae
4.
J Virol ; 95(13): e0048621, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33853961

RESUMO

Wild-type adeno-associated virus (AAV) can only replicate in the presence of helper factors, which can be provided by coinfecting helper viruses such as adenoviruses and herpesviruses. The AAV genome consists of a linear, single-stranded DNA (ssDNA), which is converted into different molecular structures within the host cell. Using high-throughput sequencing, we found that herpes simplex virus 1 (HSV-1) coinfection leads to a shift in the type of AAV genome end recombination. In particular, open-end inverted terminal repeat (ITR) recombination was enhanced, whereas open-closed ITR recombination was reduced in the presence of HSV-1. We demonstrate that the HSV-1 protein ICP8 plays an essential role in HSV-1-mediated interference with AAV genome end recombination, indicating that the previously described ICP8-driven mechanism of HSV-1 genome recombination may be underlying the observed changes. We also provide evidence that additional factors, such as products of true late genes, are involved. Although HSV-1 coinfection significantly changed the type of AAV genome end recombination, no significant change in the amount of circular AAV genomes was identified. IMPORTANCE Adeno-associated virus (AAV)-mediated gene therapy represents one of the most promising approaches for the treatment of genetic diseases. Currently, various GMP-compatible production methods can be applied to manufacture clinical-grade vector, including methods that employ helper factors derived from herpes simplex virus 1 (HSV-1). Yet, to date, we do not fully understand how HSV-1 interacts with AAV. We observed that HSV-1 modulates AAV genome ends similarly to the genome recombination events observed during HSV-1 replication and postulate that further improvements of the HSV-1 production platform may enhance packaging of the recombinant AAV particles.


Assuntos
Dependovirus/crescimento & desenvolvimento , Dependovirus/genética , Genoma Viral/genética , Vírus Auxiliares/genética , Herpesvirus Humano 1/genética , Recombinação Genética/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Coinfecção/patologia , Células HEK293 , Células HeLa , Herpes Simples/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Infecções por Parvoviridae/patologia , Sequências Repetidas Terminais/genética , Células Vero , Interferência Viral/genética , Replicação Viral/genética
5.
Int J Mol Sci ; 22(9)2021 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-33923223

RESUMO

Herpes Simplex Virus Type-1 (HSV-1) forms progeny in the nucleus within distinct membrane-less inclusions, the viral replication compartments (VRCs), where viral gene expression, DNA replication, and packaging occur. The way in which the VRCs maintain spatial integrity remains unresolved. Here, we demonstrate that the essential viral transcription factor ICP4 is an intrinsically disordered protein (IDP) capable of driving protein condensation and liquid-liquid phase separation (LLPS) in transfected cells. Particularly, ICP4 forms nuclear liquid-like condensates in a dose- and time-dependent manner. Fluorescence recovery after photobleaching (FRAP) assays revealed rapid exchange rates of EYFP-ICP4 between phase-separated condensates and the surroundings, akin to other viral IDPs that drive LLPS. Likewise, HSV-1 VRCs revealed by EYFP-tagged ICP4 retained their liquid-like nature, suggesting that they are phase-separated condensates. Individual VRCs homotypically fused when reaching close proximity and grew over the course of infection. Together, the results of this study demonstrate that the HSV-1 transcription factor ICP4 has characteristics of a viral IDP, forms condensates in the cell nucleus by LLPS, and can be used as a proxy for HSV-1 VRCs with characteristics of liquid-liquid phase-separated condensates.


Assuntos
Regulação Viral da Expressão Gênica , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Compartimentos de Replicação Viral , Animais , Núcleo Celular/metabolismo , Chlorocebus aethiops , Herpes Simples/genética , Herpes Simples/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Intrinsicamente Desordenadas/genética , Extração Líquido-Líquido , Transição de Fase , Células Vero
6.
J Zoo Wildl Med ; 52(1): 28-37, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33827158

RESUMO

Equine sarcoids (ES) were diagnosed in 12 Somali wild asses (SWA) (Equus africanus somaliensis) from 10 different institutions of the SWA European Endangered Species Programme from 1976 to 2019. Samples of surgically excised masses, biopsies, or necropsy samples were submitted for histologic and virologic analysis. In addition, tissue samples from one onager (Equus hemionus onager), one kulan (Equus hemionus kulan), and two Hartmann's mountain zebras (HMZ) (Equus zebra hartmannae) were examined. Histology confirmed the diagnosis of ES exhibiting the typical microscopic features. Polymerase chain reaction detected bovine papillomavirus type 1 (BPV1) DNA in eight SWA samples and bovine papillomavirus type 2 (BPV2) DNA in one SWA sample. The onager, kulan, and one HMZ sample tested positive for BPV1. The other HMZ tested positive for BPV1 and BPV2. This is the first report of ES in an onager. Surgical excision was the treatment elected by most veterinarians. A follow-up survey of the cases over several years after clinical diagnosis and therapy revealed variable individual outcome with ES recurrence in four cases. Three SWA and the kulan were euthanized due to the severity of the lesions. Nine affected SWA were males with seven having a sarcoid located at the prepuce. Because a genetic disposition is a risk factor for the development of ES in horses, this may also be true for endangered wild equids with few founder animals in their studbook history. Innovative approaches regarding therapy and prevention of ES in wild equids are therefore highly encouraged.


Assuntos
Equidae , Neoplasias Cutâneas/veterinária , Animais , Animais de Zoológico , Papillomavirus Bovino 1/isolamento & purificação , DNA Viral/genética , DNA Viral/isolamento & purificação , Espécies em Perigo de Extinção , Feminino , Predisposição Genética para Doença , Masculino , Neoplasias/veterinária , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia
7.
J Virol Methods ; 289: 114041, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33309756

RESUMO

As the causative agent of Infectious Bovine Rhinotracheitis (IBR) and Infectious Pustular Vulvovaginitis/Balanoposthitis (IPV/IPB), Bovine alphaherpesvirus 1 (BoHV-1) is responsible for high economic losses in the cattle industry worldwide. This study aimed to establish a fast, colorimetric loop-mediated isothermal amplification (LAMP) assay for the detection of viral DNA. Phenol red is used as pH-sensitive readout, relying on a distinct color change from pink to yellow in case of a positive reaction. LAMP reactions with different primers were compared and a newly designed set targeting the gene encoding the tegument protein V67 provided best results, enabling readout within 8-30 min. LAMP showed less cross-reactions with other ruminant alphaherpesviruses than qPCR but was 10-fold less sensitive. However, LAMP still detected down to 14 copies. The test performance was evaluated using 26 well-characterized nasal swabs from cattle with respiratory disease. All samples were correctly identified when using column-extracted DNA. Using a simple DNA precipitation method, only two weak-positive samples turned indeterminate. Combining this DNA precipitation with a makeshift water bath heated by a gastronomic immersion heater allowed successful application of the colorimetric LAMP assay under resource-limited conditions. This technique can therefore help in managing IBR/IPV outbreaks where sophisticated laboratory equipment is unavailable.


Assuntos
Colorimetria , Técnicas de Amplificação de Ácido Nucleico , Animais , Bovinos , DNA Viral/genética , Técnicas de Diagnóstico Molecular , Sensibilidade e Especificidade
8.
J Virol ; 94(8)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32024777

RESUMO

Malignant catarrhal fever (MCF) is a rare but frequently lethal disease of certain cloven-hoofed animals. At least 10 different viruses, all members of the Macavirus genus in the subfamily Gammaherpesvirinae, are known as causative agents of MCF. Among these, ovine herpesvirus 2 (OvHV-2) is the most frequent and economically most important MCF agent. Phenotypically, MCF is characterized by severe lymphocytic arteritis-periarteritis, which leads to the accumulation of activated lymphocytes accompanied by apoptosis and necrosis in a broad range of tissues. However, a viral factor that might be responsible for tissue damage has not yet been identified. We have studied a seemingly intergenic locus on the OvHV-2 genome, which was previously shown to be transcriptionally highly active in MCF-affected tissue. We identified by 5' and 3' rapid amplification of cDNA ends (RACE) a conserved, double-spliced transcript that encoded a 9.9-kDa hydrophobic protein. The newly detected gene, Ov8.25, and its splicing pattern were conserved among OvHV-2 strains of different origins. Upon transient expression of synthetic variants of this gene in various cell types, including bovine lymphocytes, the protein (pOv8.25) was shown to target mitochondria, followed by caspase-dependent apoptosis and necrosis. Notably, a deletion mutant of the same protein lost these abilities. Finally, we detected pOv8.25 in brain-infiltrating lymphocytes of cattle with MCF. Thus, the cell death-causing properties of pOv8.25 in affected cells may be involved in the emergence of typical MCF-associated apoptosis and necrosis. Thus, we have identified a novel OvHV-2 protein, which might contribute to the phenotype of MCF-related lesions.IMPORTANCE Ovine herpesvirus 2 (OvHV-2) circulates among sheep without causing disease. However, upon transmission to cattle, the same virus instigates a frequently lethal disease, malignant catarrhal fever (MCF). While the cause of death and pathogenesis of tissue lesions are still poorly understood, MCF is characterized by the accumulation of lymphocytes in various tissues, associated with vasculitis and cell death. As infectious virus is hardly present in these lesions, the cause of cell death cannot be explained simply by viral replication. The significance of our research is in identifying and characterizing a previously overlooked gene of OvHV-2 (Ov8.25), which is highly expressed in animals with MCF. Its encoded protein targets mitochondria, causing apoptosis and necrosis, thus contributing to an understanding of the source and nature of cell death. As the corresponding genetic locus is also active in the context of MCF due to a different macavirus, we may have detected a common denominator of the disease phenotype.


Assuntos
Apoptose , Gammaherpesvirinae/genética , Gammaherpesvirinae/metabolismo , Mitocôndrias/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Doenças do Gato/virologia , Gatos , Bovinos , Linhagem Celular , Chlorocebus aethiops , Linfócitos , Febre Catarral Maligna/patologia , Febre Catarral Maligna/virologia , Mitocôndrias/patologia , Necrose/virologia , Alinhamento de Sequência , Ovinos , Doenças dos Ovinos/virologia , Células Vero , Proteínas Virais/isolamento & purificação
9.
J Virol ; 94(7)2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-31915278

RESUMO

One step of the life cycle common to all rotaviruses (RV) studied so far is the formation of viroplasms, membrane-less cytosolic inclusions providing a microenvironment for early morphogenesis and RNA replication. Viroplasm-like structures (VLS) are simplified viroplasm models consisting of complexes of nonstructural protein 5 (NSP5) with the RV core shell VP2 or NSP2. We identified and characterized the domains required for NSP5-VP2 interaction and VLS formation. VP2 mutations L124A, V865A, and I878A impaired both NSP5 hyperphosphorylation and NSP5/VP2 VLS formation. Moreover, NSP5-VP2 interaction does not depend on NSP5 hyperphosphorylation. The NSP5 tail region is required for VP2 interaction. Notably, VP2 L124A expression acts as a dominant-negative element by disrupting the formation of either VLS or viroplasms and blocking RNA synthesis. In silico analyses revealed that VP2 L124, V865, and I878 are conserved among RV species A to H. Detailed knowledge of the protein interaction interface required for viroplasm formation may facilitate the design of broad-spectrum antivirals to block RV replication.IMPORTANCE Alternative treatments to combat rotavirus infection are a requirement for susceptible communities where vaccines cannot be applied. This demand is urgent for newborn infants, immunocompromised patients, adults traveling to high-risk regions, and even for the livestock industry. Aside from structural and physiological divergences among RV species studied before now, all replicate within cytosolic inclusions termed viroplasms. These inclusions are composed of viral and cellular proteins and viral RNA. Viroplasm-like structures (VLS), composed of RV protein NSP5 with either NSP2 or VP2, are models for investigating viroplasms. In this study, we identified a conserved amino acid in the VP2 protein, L124, necessary for its interaction with NSP5 and the formation of both VLSs and viroplasms. As RV vaccines cover a narrow range of viral strains, the identification of VP2 L124 residue lays the foundations for the design of drugs that specifically block NSP5-VP2 interaction as a broad-spectrum RV antiviral.


Assuntos
Proteínas do Capsídeo/química , Citosol/virologia , Rotavirus/fisiologia , Proteínas não Estruturais Virais/química , Proteínas Virais/química , Animais , Proteínas do Capsídeo/genética , Chlorocebus aethiops , Simulação por Computador , Genes Dominantes , Cobaias , Células HEK293 , Humanos , Macaca mulatta , Camundongos , Mutação , Fosforilação , Ligação Proteica , Domínios Proteicos , RNA Viral/biossíntese , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Replicação Viral
10.
BMC Vet Res ; 15(1): 356, 2019 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-31640696

RESUMO

BACKGROUND: There is growing evidence that equine papillomavirus type 2 (EcPV2) infection is causally associated with the development of equine genital squamous cell carcinomas (SCCs). Early stages of disease present clinically as plaques or wart-like lesions which can gradually progress to tumoural lesions. Histologically these lesions are inconsistently described as benign hyperplasia, papilloma, penile intraepithelial neoplasia (PIN), carcinoma in situ (CIS) or SCC. Guidelines for histological classification of early SCC precursor lesions are not precisely defined, leading to potential misdiagnosis. The aim of this study was to identify histologic criteria and diagnostic markers allowing for a more accurate diagnosis of EcPV2-associated equine penile lesions. RESULTS: A total of 61 archived equine penile lesions were histologically re-assessed and classified as benign hyperplasia, papilloma, CIS or SCC. From these, 19 representative lesions and adjacent normal skin were comparatively analysed for the presence of EcPV2 DNA and transcripts using PCR and RNA in situ hybridisation (RISH). All lesional samples were positive by EcPV2 PCR and RISH, while adjacent normal skin was negative. RISH analysis yielded signal distribution patterns that allowed distinction of early (hyperplasia, papilloma) from late stage lesions (CIS, SCC). Subsequently, the 19 lesions were further assessed for expression of p53, Ki67, MCM7 and MMP1 by immunohistochemistry (IHC). All four proteins were expressed in both normal and lesional tissue. However, p53 expression was up-regulated in basal keratinocyte layers of papillomas, CIS and SCCs, as well as in upper keratinocyte layers of CIS and SCCs. MCM7 expression was only up-regulated in upper proliferating keratinocyte layers of papillomas, CIS and SCCs. CONCLUSION: This study proposes combining a refined histological protocol for analysis of equine penile lesions with PCR- and/or RISH based EcPV2-screening and p53/MCM7 IHC to more accurately determine the type of lesion. This may help to guide the choice of optimum treatment strategy, especially at early stages of disease.


Assuntos
Doenças dos Cavalos/patologia , Infecções por Papillomavirus/veterinária , Neoplasias Penianas/veterinária , Pênis/patologia , Animais , DNA Viral/análise , Doenças dos Cavalos/virologia , Cavalos , Hibridização In Situ/veterinária , Masculino , Papillomaviridae/classificação , Infecções por Papillomavirus/patologia , Neoplasias Penianas/patologia , Neoplasias Penianas/virologia , Lesões Pré-Cancerosas/patologia , Lesões Pré-Cancerosas/veterinária , Lesões Pré-Cancerosas/virologia
11.
F1000Res ; 8: 727, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31448105

RESUMO

Background: Capsids of herpes simplex virus 1 (HSV-1) are assembled in cell nuclei, released into the perinuclear space by budding at the inner nuclear membrane acquiring tegument and envelope. Alternatively, capsids gain access to the cytoplasm via dilated nuclear pores. They are enveloped by Golgi membranes. Us3 is a non-essential viral kinase that is involved in nucleus-to-cytoplasm translocation, preventing apoptosis and regulation of phospholipid-biosynthesis. Us3-deletion mutants (HSV-1∆Us3) accumulate in the perinuclear space. Nuclear and Golgi membranes proliferate, and homogeneous, proteinaceous structures of unknown identity are deposited in nuclei and cytoplasm. Glycoprotein K (gK), a highly hydrophobic viral protein, is essential for production of infectious progeny virus but, according to the literature, exclusively vital for envelopment of capsids by Golgi membranes. In the absence of Us3, virions remain stuck in the perinuclear space but mature to infectivity without reaching Golgi membranes, suggesting further function of gK than assumed. Methods: We constructed a HSV-1∆Us3 mutant designated CK177∆Us3gK-HA, in which gK was hemagglutinin (HA) epitope-tagged in order to localize gK by immunolabeling using antibodies against HA for light and electron microscopy. Results: CK177∆Us3gK-HA-infected Vero cells showed similar alterations as those reported for other HSV-1∆Us3, including accumulation of virions in the perinuclear space, overproduction of nuclear and Golgi membranes containing electron dense material with staining property of proteins. Immunolabeling using antibodies against HA revealed that gK is overproduced and localized at nuclear membranes, perinuclear virions stuck in the perinuclear space, Golgi membranes and on protein deposits in cytoplasm and nuclei. Conclusions: Us3 is involved in proper assembly of membranes needed for envelopment and incorporation of gK. Without Us3, virions derived by budding at nuclear membranes remain stuck in the perinuclear space but incorporate gK into their envelope to gain infectivity.


Assuntos
Herpesvirus Humano 1 , Animais , Chlorocebus aethiops , Glicoproteínas , Células Vero , Proteínas Virais , Vírion
12.
J Gen Virol ; 100(6): 985-998, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31084699

RESUMO

Equine papillomavirus type 2 (EcPV2) was discovered only recently, but it is found consistently in the context of genital squamous cell carcinomas (SCCs). Since neither cell cultures nor animal models exist, the characterization of this potential disease agent relies on the analysis of patient materials. To analyse the host and viral transcriptome in EcPV2-affected horses, genital tissue samples were collected from horses with EcPV2-positive lesions as well as from healthy EcPV2-negative horses. It was determined by RNA-seq analysis that there were 1957 differentially expressed (DE) host genes between the SCC and control samples. These genes were most abundantly related to DNA replication, cell cycle, extracellular matrix (ECM)-receptor interaction and focal adhesion. By comparison to other cancer studies, MMP1 and IL8 appeared to be potential marker genes for the development of SCCs. Analysis of the viral reads revealed the transcriptional activity of EcPV2 in all SCC samples. While few reads mapped to the structural viral genes, the majority of reads mapped to the non-structural early (E) genes, in particular to E6, E7 and E2/E4. Within these reads a distinct pattern of splicing events, which are essential for the expression of different genes in PV infections, was observed. Additionally, in one sample the integration of EcPV2 DNA into the host genome was detected by DNA-seq and confirmed by PCR. In conclusion, while host MMP1 and IL8 expression and the presence of EcPV2 may be useful markers in genital SCCs, further research on EcPV2-related pathomechanisms may focus on cell cycle-related genes, the viral genes E6, E7 and E2/E4, and integration events.


Assuntos
Carcinoma de Células Escamosas/genética , Regulação Viral da Expressão Gênica/genética , Doenças dos Cavalos/genética , Papillomaviridae/genética , Infecções por Papillomavirus/genética , Splicing de RNA/genética , Transdução de Sinais/genética , Animais , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/virologia , DNA Viral/genética , Genes Virais/genética , Doenças dos Cavalos/virologia , Cavalos/genética , Cavalos/virologia , Interleucina-8/genética , Metaloproteinase 1 da Matriz/genética , Infecções por Papillomavirus/veterinária , Infecções por Papillomavirus/virologia , Reação em Cadeia da Polimerase/métodos , RNA-Seq/métodos , Transcrição Gênica/genética
13.
J Gen Virol ; 100(3): 497-510, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30694168

RESUMO

Bovine herpesvirus 1 (BoHV-1)-encoded UL49.5 (a homologue of herpesvirus glycoprotein N) can combine different functions, regulated by complex formation with viral glycoprotein M (gM). We aimed to identify the mechanisms governing the immunomodulatory activity of BoHV-1 UL49.5. In this study, we addressed the impact of gM/UL49.5-specific regions on heterodimer formation, folding and trafficking from the endoplasmic reticulum (ER) to the trans-Golgi network (TGN) - events previously found to be responsible for abrogation of the UL49.5-mediated inhibition of the transporter associated with antigen processing (TAP). We first established, using viral mutants, that no other viral protein could efficiently compensate for the chaperone function of UL49.5 within the complex. The cytoplasmic tail of gM, containing putative trafficking signals, was dispensable either for ER retention of gM or for the release of the complex. We constructed cell lines with stable co-expression of BoHV-1 gM with chimeric UL49.5 variants, composed of the BoHV-1 N-terminal domain fused to the transmembrane region (TM) from UL49.5 of varicella-zoster virus or TM and the cytoplasmic tail of influenza virus haemagglutinin. Those membrane-anchored N-terminal domains of UL49.5 were sufficient to form a complex, yet gM/UL49.5 folding and ER-TGN trafficking could be affected by the UL49.5 TM sequence. Finally, we found that leucine substitutions in putative glycine zipper motifs within TM helices of gM resulted in strong reduction of complex formation and decreased ability of gM to interfere with UL49.5-mediated major histocompatibility class I downregulation. These findings highlight the importance of gM/UL49.5 transmembrane domains for the biology of this conserved herpesvirus protein complex.


Assuntos
Doenças dos Bovinos/virologia , Retículo Endoplasmático/virologia , Complexo de Golgi/virologia , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 1/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Bovinos , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/química , Herpesvirus Bovino 1/genética , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Ligação Proteica , Domínios Proteicos , Transporte Proteico , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
14.
Artigo em Inglês | MEDLINE | ID: mdl-30533742

RESUMO

We present the full-length genome sequence of a new papillomavirus detected in skin lesions collected from a boa (Boa constrictor). Based on the nucleotide sequence analysis, we propose to designate the newly identified virus as Boa constrictor papillomavirus type 1 (BcPV1), a new species in the genus Dyomupapillomavirus.

15.
Vet Microbiol ; 223: 113-118, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30173736

RESUMO

Papillomavirus-specific DNA was detected in skin lesions collected from an okapi (Okapia johnstoni) in the Zoo Basel. According to the nucleotide sequence analysis, the virus belongs to the genus Deltapapillomavirus. Based on bioinformatics analysis, we propose to designate the newly identified virus as Okapia johnstoni Papillomavirus type 1 (OjPV1). OjPV1 is genetically most closely related to a recently described giraffe (Giraffa camelopardalis) -specific papillomavirus (GcPV1). Of note, the putative oncogenic E5 proteins from OjPV1 and GcPV1 are more conserved than the L1 proteins. This indicates, that the selection pressure on E5 may be more pronounced than that on the otherwise most conserved major capsid protein L1.


Assuntos
Deltapapillomavirus/isolamento & purificação , Girafas/virologia , Infecções por Papillomavirus/veterinária , Animais , Animais de Zoológico , Biologia Computacional , Deltapapillomavirus/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala/veterinária , Especificidade de Hospedeiro , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Filogenia , Análise de Sequência de DNA/veterinária , Pele/patologia , Pele/virologia
16.
Parasitol Res ; 117(6): 1925-1932, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29705877

RESUMO

West Nile virus (WNV) is continuously spreading in Eastern and Southern Europe. However, the extent of vector competence of Aedes japonicus (Theobald, 1901) is controversial. In this work, we elucidated the dynamics of virus growth in this invasive mosquito species. Females of Ae. japonicus were reared from eggs collected in the field in Switzerland and fed on bovine blood spiked with two WNV lineage 1 strains (FIN, Italy; NY99, USA). Fully engorged females were incubated for 14 days under a fluctuating temperature regime of 24 ± 7 °C (average 24 °C), 45-90% relative humidity, which is realistic for a Central European mid-summer day. Infection, dissemination, and transmission rates were assessed from individual mosquitoes by analyzing the abdomen, legs and wings, and saliva for the presence of viral RNA. Saliva was also investigated for the presence of infectious virus particles. Overall, 302 females were exposed to WNV strain FIN and 293 to strain NY99. A higher infection rate was observed for NY99 (57.4%) compared to FIN (30.4%) (p = 0.003). There was no statistical evidence that the dissemination rate (viral RNA in legs and wings) was different between females infected with FIN (57.1%) compared to NY99 (35.5%) (p = 0.16). Viral RNA load of FIN compared to NY99 was significantly higher in the hemocoel (p = 0.031) of exposed females but not at other sites (legs and wings, saliva). This is the first study describing the vector competence parameters for two WNV strains in a European population of Ae. japonicus. The high dissemination and transmission rates for WNV under a realistic temperature regime in Ae. japonicus together with recent findings on its opportunistic feeding behavior (mammals and birds) indicate its potential role in WNV transmission in Central Europe where it is highly abundant.


Assuntos
Aedes/virologia , Mosquitos Vetores/virologia , Febre do Nilo Ocidental/transmissão , Vírus do Nilo Ocidental/crescimento & desenvolvimento , Abdome/virologia , Animais , Bovinos , Linhagem Celular , Chlorocebus aethiops , Comportamento Alimentar , Feminino , Itália , Saliva/virologia , Suíça , Temperatura , Células Vero , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/classificação , Vírus do Nilo Ocidental/isolamento & purificação , Asas de Animais/virologia
17.
Infect Immun ; 86(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29229735

RESUMO

Bacillus subtilis is known as an endospore- and biofilm-forming bacterium with probiotic properties. We have recently developed a method for displaying heterologous proteins on the surface of B. subtilis biofilms by introducing the coding sequences of the protein of interest into the bacterial genome to generate a fusion protein linked to the C terminus of the biofilm matrix protein TasA. Although B. subtilis is a regular component of the gut microflora, we constructed a series of recombinant B. subtilis strains that were tested for their ability to be used to immunize dogs following oral application of the spores. Specifically, we tested recombinant spores of B. subtilis carrying either the fluorescent protein mCherry or else selected antigenic peptides (tropomyosin and paramyosin) from Echinococcus granulosus, a zoonotic intestinal tapeworm of dogs and other carnivores. The application of the recombinant B. subtilis spores led to the colonization of the gut with recombinant B. subtilis but did not cause any adverse effect on the health of the animals. As measured by enzyme-linked immunosorbent assay and immunoblotting, the dogs were able to develop a humoral immune response against mCherry as well as against E. granulosus antigenic peptides. Interestingly, the sera of dogs obtained after immunization with recombinant spores of E. granulosus peptides were able to recognize E. granulosus protoscoleces, which represent the infective form of the head of the tapeworms. These results represent an essential step toward the establishment of B. subtilis as an enteric vaccine agent.


Assuntos
Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/imunologia , Bacillus subtilis/genética , Doenças do Cão/imunologia , Equinococose/veterinária , Echinococcus granulosus/imunologia , Tropomiosina/imunologia , Animais , Antígenos de Helmintos/administração & dosagem , Antígenos de Helmintos/genética , Bacillus subtilis/fisiologia , Biofilmes , Doenças do Cão/parasitologia , Doenças do Cão/prevenção & controle , Cães , Equinococose/imunologia , Equinococose/parasitologia , Equinococose/prevenção & controle , Echinococcus granulosus/genética , Expressão Gênica , Proteínas de Helminto/administração & dosagem , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Imunidade Humoral , Esporos Bacterianos/genética , Esporos Bacterianos/fisiologia , Tropomiosina/administração & dosagem , Tropomiosina/genética , Vacinas/administração & dosagem , Vacinas/genética , Vacinas/imunologia
18.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515305

RESUMO

Adeno-associated virus 2 (AAV2) depends on the simultaneous presence of a helper virus such as herpes simplex virus 1 (HSV-1) for productive replication. At the same time, AAV2 efficiently blocks the replication of HSV-1, which would eventually limit its own replication by diminishing the helper virus reservoir. This discrepancy begs the question of how AAV2 and HSV-1 can coexist in a cell population. Here we show that in coinfected cultures, AAV2 DNA replication takes place almost exclusively in S/G2-phase cells, while HSV-1 DNA replication is restricted to G1 phase. Live microscopy revealed that not only wild-type AAV2 (wtAAV2) replication but also reporter gene expression from both single-stranded and double-stranded (self-complementary) recombinant AAV2 vectors preferentially occurs in S/G2-phase cells, suggesting that the preference for S/G2 phase is independent of the nature of the viral genome. Interestingly, however, a substantial proportion of S/G2-phase cells transduced by the double-stranded but not the single-stranded recombinant AAV2 vectors progressed through mitosis in the absence of the helper virus. We conclude that cell cycle-dependent AAV2 rep expression facilitates cell cycle-dependent AAV2 DNA replication and inhibits HSV-1 DNA replication. This may limit competition for cellular and viral helper factors and, hence, creates a biological niche for either virus to replicate.IMPORTANCE Adeno-associated virus 2 (AAV2) differs from most other viruses, as it requires not only a host cell for replication but also a helper virus such as an adenovirus or a herpesvirus. This situation inevitably leads to competition for cellular resources. AAV2 has been shown to efficiently inhibit the replication of helper viruses. Here we present a new facet of the interaction between AAV2 and one of its helper viruses, herpes simplex virus 1 (HSV-1). We observed that AAV2 rep gene expression is cell cycle dependent and gives rise to distinct time-controlled windows for HSV-1 replication. High Rep protein levels in S/G2 phase support AAV2 replication and inhibit HSV-1 replication. Conversely, low Rep protein levels in G1 phase permit HSV-1 replication but are insufficient for AAV2 replication. This allows both viruses to productively replicate in distinct sets of dividing cells.


Assuntos
Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Dependovirus/crescimento & desenvolvimento , Vírus Auxiliares/crescimento & desenvolvimento , Herpesvirus Humano 1/crescimento & desenvolvimento , Interferência Viral , Proteínas Virais/metabolismo , Replicação Viral , Linhagem Celular , Coinfecção , Expressão Gênica , Humanos , Microscopia , Cultura de Vírus
19.
Int J Mol Sci ; 18(2)2017 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-28212334

RESUMO

Rotaviruses (RVs) are important enteric pathogens of newborn humans and animals, causing diarrhea and in rare cases death, especially in very young individuals. Rotavirus vaccines presently used are modified live vaccines that lack complete biological safety. Previous work from our laboratory suggested that vaccines based on in situ produced, non-infectious rotavirus-like particles (RVLPs) are efficient while being entirely safe. However, using either vaccine, active mucosal immunization cannot induce protective immunity in newborns due to their immature immune system. We therefore hypothesized that offspring from vaccinated dams are passively immunized either by transfer of maternal antibodies during pregnancy or by taking up antibodies from milk. Using a codon optimized polycistronic gene expression cassette packaged into herpesvirus particles, the simultaneous expression of the RV capsid genes led to the intracellular formation of RVLPs in various cell lines. Vaccinated dams developed a strong RV specific IgG antibody response determined in sera and milk of both mother and pups. Moreover, sera of naïve pups nursed by vaccinated dams also had RV specific antibodies suggesting a lactogenic transfer of antibodies. Although full protection of pups was not achieved in this mouse model, our observations are important for the development of improved vaccines against RV in humans as well as in various animal species.


Assuntos
Anticorpos Antivirais/imunologia , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Leite/imunologia , Vacinas contra Rotavirus/genética , Vacinas contra Rotavirus/imunologia , Rotavirus/imunologia , Vacinação , Animais , Anticorpos Antivirais/sangue , Especificidade de Anticorpos , Linhagem Celular Tumoral , Chlorocebus aethiops , Códon , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Gravidez , Infecções por Rotavirus/imunologia , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Transdução Genética , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Células Vero , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/imunologia
20.
BMC Vet Res ; 12(1): 147, 2016 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-27449225

RESUMO

BACKGROUND: Papillomaviruses can cause proliferative skin lesions ranging from benign hyperplasia to squamous cell carcinoma (SCC). However, asymptomatic infection is also possible. Several groups have detected Felis domesticus Papillomavirus type 2 (FdPV2) DNA in association with feline Bowenoid in situ carcinoma (BISC). Therefore, a causative connection has been suggested. However, the knowledge about FdPV2 epidemiology is limited. The aim of this study was to describe the genoprevalence and seroprevalence of FdPV2 in healthy cats. For this purpose an FdPV2-specific quantitative (q)PCR assay was developed and used to analyse Cytobrush samples collected from 100 dermatologically healthy cats. Moreover, an ELISA was established to test the sera obtained from the same cats for antibodies against the major capsid protein (L1) of FdPV2. RESULTS: The genoprevalence of FdPV2 was to 98 %. Surprisingly, the quantities of viral DNA detected in some samples from the healthy cats exceeded the amounts detected in control samples from feline BISC lesions. The seroprevalence was much lower, amounting to 22 %. The concentrations of antibodies against FdPV2 were relatively low in healthy cats, whereas they were very high in control cats with BISC. CONCLUSION: These observations suggest that FdPV2 is highly prevalent, even among healthy cats. However, cats that carry it on their skin mount in most instances no antibody response. It might be hypothesized that FdPV2 is only rarely productively replicating or its replication is only rarely exposed to the immune system.


Assuntos
Doenças do Gato/virologia , Papillomaviridae/genética , Animais , Gatos , Feminino , Masculino , Papillomaviridae/classificação , Papillomaviridae/isolamento & purificação , Estudos Soroepidemiológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...