Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
PLoS Pathog ; 20(4): e1012139, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38578790

RESUMO

Alpha herpesviruses naturally infect the peripheral nervous system, and can spread to the central nervous system, causing severe debilitating or deadly disease. Because alpha herpesviruses spread along synaptic circuits, and infected neurons exhibit altered electrophysiology and increased spontaneous activity, we hypothesized that alpha herpesviruses use activity-dependent synaptic vesicle-like regulated secretory mechanisms for egress and spread from neurons. Using live-cell fluorescence microscopy, we show that Pseudorabies Virus (PRV) particles use the constitutive Rab6 post-Golgi secretory pathway to exit from the cell body of primary neurons, independent of local calcium signaling. Some PRV particles colocalize with Rab6 in the proximal axon, but we did not detect colocalization/co-transport in the distal axon. Thus, the specific secretory mechanisms used for viral egress from axons remains unclear. To address the role of neuronal activity more generally, we used a compartmentalized neuron culture system to measure the egress and spread of PRV from axons, and pharmacological and optogenetics approaches to modulate neuronal activity. Using tetrodotoxin to silence neuronal activity, we observed no inhibition, and using potassium chloride or optogenetics to elevate neuronal activity, we also show no increase in virus spread from axons. We conclude that PRV egress from neurons uses constitutive secretory mechanisms: generally, activity-independent mechanisms in axons, and specifically, the constitutive Rab6 post-Golgi secretory pathway in cell bodies.


Assuntos
Alphaherpesvirinae , Herpesvirus Suídeo 1 , Pseudorraiva , Animais , Corpo Celular/metabolismo , Proteínas do Envelope Viral/metabolismo , Axônios , Alphaherpesvirinae/metabolismo , Neurônios , Herpesvirus Suídeo 1/metabolismo , Pseudorraiva/metabolismo , Exocitose
2.
PLoS Pathog ; 19(2): e1010959, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36749787

RESUMO

Conserved Herpesviridae protein kinases (CHPK) are conserved among all members of the Herpesviridae. Herpesviruses lacking CHPK propagate in cell culture at varying degrees, depending on the virus and cell culture system. CHPK is dispensable for Marek's disease herpesvirus (MDV) replication in cell culture and experimental infection in chickens; however, CHPK-particularly its kinase activity-is essential for horizontal transmission in chickens, also known as natural infection. To address the importance of CHPK during natural infection in chickens, we used liquid chromatography-tandem mass spectrometry (LC-MS/MS) based proteomics of samples collected from live chickens. Comparing modification of viral proteins in feather follicle epithelial (FFE) cells infected with wildtype or a CHPK-null virus, we identified the US10 protein (pUS10) as a potential target for CHPK in vivo. When expression of pUS10 was evaluated in cell culture and in FFE skin cells during in vivo infection, pUS10 was severely reduced or abrogated in cells infected with CHPK mutant or CHPK-null viruses, respectively, indicating a potential role for pUS10 in transmission. To test this hypothesis, US10 was deleted from the MDV genome, and the reconstituted virus was tested for replication, horizontal transmission, and disease induction. Our results showed that removal of US10 had no effect on the ability of MDV to transmit in experimentally infected chickens, but disease induction in naturally infected chickens was significantly reduced. These results show CHPK is necessary for pUS10 expression both in cell culture and in the host, and pUS10 is important for disease induction during natural infection.


Assuntos
Alphaherpesvirinae , Herpesviridae , Doença de Marek , Animais , Proteínas Quinases/metabolismo , Cromatografia Líquida , Galinhas , Espectrometria de Massas em Tandem , Herpesviridae/metabolismo , Alphaherpesvirinae/metabolismo , Proteínas Virais/metabolismo , Vírus Oncogênicos
3.
J Virol ; 97(1): e0157722, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36598202

RESUMO

Duck plague virus (DPV) is a high-morbidity fowl alphaherpesvirus that causes septicemic lesions in various organs. Most DPV genes are conserved among herpesviruses, while a few are specific to fowl herpesviruses, including the LORF3 gene, for which there is currently no literature describing its biological properties and functions. This study first addressed whether the LORF3 protein is expressed by making specific polyclonal antibodies. We could demonstrate that DPV LORF3 is an early gene and encodes a protein involved in virion assembly, mainly localized in the nucleus of DPV-infected DEF cells. To investigate the role of this novel LORF3 protein in DPV pathogenesis, we generated a recombinant virus that lacks expression of the LORF3 protein. Our data revealed that the LORF3 protein is not essential for viral replication but contributes to DPV replication in vitro and in vivo and promotes duck plague disease morbidity and mortality. Interestingly, deletion of the LORF3 protein abolished thymus atrophy in DPV-vaccinated ducks. In conclusion, this study revealed the expression of avian herpesviruses-specific genes and unraveled the role of the early protein LORF3 in the pathogenesis of DPV. IMPORTANCE DPV is a highly lethal alphaherpesvirus that causes duck plague in birds of the order Anseriformes. The virus has caused huge economic losses to the poultry industry due to high morbidity and mortality and the cost of vaccination. DPV encodes 78 open reading frames (ORFs), and these genes are involved in various processes of the viral life cycle. Functional characterization of DPV genes is important for understanding the complex viral life cycle and DPV pathogenesis. Here, we identified a novel protein encoded by LORF3, and our data suggest that the LORF3 protein is involved in the occurrence and development of duck plague.


Assuntos
Alphaherpesvirinae , Infecções por Herpesviridae , Animais , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/patogenicidade , Patos , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Células Cultivadas
4.
J Virol ; 97(5): e0024223, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37154764

RESUMO

pUL51 is a minor tegument protein important for viral assembly and cell-to-cell spread (CCS) but dispensable for replication in cell culture of all Herpesviruses for which its role has been investigated. Here, we show that pUL51 is essential for the growth of Marek's disease virus, an oncogenic alphaherpesvirus of chickens that is strictly cell-associated in cell culture. MDV pUL51 localized to the Golgi apparatus of infected primary skin fibroblasts, as described for other Herpesviruses. However, the protein was also observed at the surface of lipid droplets in infected chicken keratinocytes, hinting at a possible role of this compartment for viral assembly in the unique cell type involved in MDV shedding in vivo. Deletion of the C-terminal half of pUL51 or fusion of GFP to either the N- or C-terminus were sufficient to disable the protein's essential function(s). However, a virus with a TAP domain fused at the C-terminus of pUL51 was capable of replication in cell culture, albeit with viral spread reduced by 35% and no localization to lipid droplets. In vivo, we observed that although the replication of this virus was moderately impacted, its pathogenesis was strongly impaired. This study describes for the first time the essential role of pUL51 in the biology of a herpesvirus, its association to lipid droplets in a relevant cell type, and its unsuspected role in the pathogenesis of a herpesvirus in its natural host. IMPORTANCE Viruses usually spread from cell to cell through two mechanisms: cell-released virus and/or cell-to-cell spread (CCS). The molecular determinants of CCS and their importance in the biology of viruses during infection of their natural host are unclear. Marek's disease virus (MDV) is a deadly and highly contagious herpesvirus of chickens that produces no cell-free particles in vitro, and therefore, spreads only through CCS in cell culture. Here, we show that viral protein pUL51, an important factor for CCS of Herpesviruses, is essential for MDV growth in vitro. We demonstrate that the fusion of a large tag at the C-terminus of the protein is sufficient to moderately impair viral replication in vivo and almost completely abolish pathogenesis while only slightly reducing viral growth in vitro. This study thus uncovers a role for pUL51 associated with virulence, linked to its C-terminal half, and possibly independent of its essential functions in CCS.


Assuntos
Alphaherpesvirinae , Herpesviridae , Herpesvirus Galináceo 2 , Doença de Marek , Animais , Galinhas , Herpesvirus Galináceo 2/genética , Herpesviridae/metabolismo , Alphaherpesvirinae/metabolismo , Replicação Viral
5.
J Virol ; 96(6): e0217821, 2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35045266

RESUMO

The assembly and egress of alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), within neurons is poorly understood. A key unresolved question is the structure of the viral particle that moves by anterograde transport along the axon, and two alternative mechanisms have been described. In the "married" model, capsids acquire their envelopes in the cell body and then traffic along axons as enveloped virions within a bounding organelle. In the "separate" model, nonenveloped capsids travel from the cell body into and along the axon, eventually encountering their envelopment organelles at a distal site, such as the nerve cell terminal. Here, we describe an "envelopment trap" to test these models using the dominant negative terminal endosomal sorting complex required for transport (ESCRT) component VPS4-EQ. Green fluorescent protein (GFP)-tagged VPS4-EQ was used to arrest HSV-1 or PRV capsid envelopment, inhibit downstream trafficking, and GFP-label envelopment intermediates. We found that GFP-VPS4-EQ inhibited trafficking of HSV-1 capsids into and along the neurites and axons of mouse CAD cells and rat embryonic primary cortical neurons, consistent with egress via the married pathway. In contrast, transport of HSV-1 capsids was unaffected in the neurites of human SK-N-SH neuroblastoma cells, consistent with the separate mechanism. Unexpectedly, PRV (generally thought to utilize the married pathway) also appeared to employ the separate mechanism in SK-N-SH cells. We propose that apparent differences in the methods of HSV-1 and PRV egress are more likely a reflection of the host neuron in which transport is studied rather than true biological differences between the viruses themselves. IMPORTANCE Alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), are pathogens of the nervous system. They replicate in the nerve cell body and then travel great distances along axons to reach nerve termini and spread to adjacent epithelial cells; however, key aspects of how these viruses travel along axons remain controversial. Here, we test two alternative mechanisms for transport, the married and separate models, by blocking envelope assembly, a critical step in viral egress. When we arrest formation of the viral envelope using a mutated component of the cellular ESCRT apparatus, we find that entry of viral particles into axons is blocked in some types of neurons but not others. This approach allows us to determine whether envelope assembly occurs prior to entry of viruses into axons or afterwards and, thus, to distinguish between the alternative models for viral transport.


Assuntos
Alphaherpesvirinae , Complexos Endossomais de Distribuição Requeridos para Transporte , Herpesvirus Humano 1 , Herpesvirus Suídeo 1 , Neurônios , Alphaherpesvirinae/metabolismo , Animais , Axônios/virologia , Linhagem Celular Tumoral , Células Cultivadas , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/fisiologia , Humanos , Camundongos , Neurônios/virologia , Ratos , Montagem de Vírus/fisiologia , Internalização do Vírus
6.
J Virol ; 96(4): e0151021, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34935440

RESUMO

Recent studies have demonstrated that the signaling activity of the cytosolic pathogen sensor retinoic acid-inducible gene-I (RIG-I) is modulated by a variety of posttranslational modifications (PTMs) to fine-tune the antiviral type I interferon (IFN) response. Whereas K63-linked ubiquitination of the RIG-I caspase activation and recruitment domains (CARDs) catalyzed by TRIM25 or other E3 ligases activates RIG-I, phosphorylation of RIG-I at S8 and T170 represses RIG-I signal transduction by preventing the TRIM25-RIG-I interaction and subsequent RIG-I ubiquitination. While strategies to suppress RIG-I signaling by interfering with its K63-polyubiquitin-dependent activation have been identified for several viruses, evasion mechanisms that directly promote RIG-I phosphorylation to escape antiviral immunity are unknown. Here, we show that the serine/threonine (Ser/Thr) kinase US3 of herpes simplex virus 1 (HSV-1) binds to RIG-I and phosphorylates RIG-I specifically at S8. US3-mediated phosphorylation suppressed TRIM25-mediated RIG-I ubiquitination, RIG-I-MAVS binding, and type I IFN induction. We constructed a mutant HSV-1 encoding a catalytically-inactive US3 protein (K220A) and found that, in contrast to the parental virus, the US3 mutant HSV-1 was unable to phosphorylate RIG-I at S8 and elicited higher levels of type I IFNs, IFN-stimulated genes (ISGs), and proinflammatory cytokines in a RIG-I-dependent manner. Finally, we show that this RIG-I evasion mechanism is conserved among the alphaherpesvirus US3 kinase family. Collectively, our study reveals a novel immune evasion mechanism of herpesviruses in which their US3 kinases phosphorylate the sensor RIG-I to keep it in the signaling-repressed state. IMPORTANCE Herpes simplex virus 1 (HSV-1) establishes lifelong latency in the majority of the human population worldwide. HSV-1 occasionally reactivates to produce infectious virus and to facilitate dissemination. While often remaining subclinical, both primary infection and reactivation occasionally cause debilitating eye diseases, which can lead to blindness, as well as life-threatening encephalitis and newborn infections. To identify new therapeutic targets for HSV-1-induced diseases, it is important to understand the HSV-1-host interactions that may influence infection outcome and disease. Our work uncovered direct phosphorylation of the pathogen sensor RIG-I by alphaherpesvirus-encoded kinases as a novel viral immune escape strategy and also underscores the importance of RNA sensors in surveilling DNA virus infection.


Assuntos
Proteína DEAD-box 58/metabolismo , Herpesvirus Humano 1/imunologia , Evasão da Resposta Imune , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Imunológicos/metabolismo , Proteínas Virais/metabolismo , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/fisiologia , Sequência de Aminoácidos , Proteína DEAD-box 58/química , Células HEK293 , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Receptores Imunológicos/química , Proteínas Virais/genética
7.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31748397

RESUMO

Fibropapillomatosis (FP) is a tumor disease associated with a herpesvirus (chelonid herpesvirus 5 [ChHV5]) that affects mainly green turtles globally. Understanding the epidemiology of FP has been hampered by a lack of robust serological assays to monitor exposure to ChHV5. This is due in part to an inability to efficiently culture the virus in vitro for neutralization assays. Here, we expressed two glycoproteins (FUS4 and FUS8) from ChHV5 using baculovirus. These proteins were immobilized on enzyme-linked immunosorbent assay plates in their native form and assayed for reactivity to two types of antibodies, full-length 7S IgY and 5.7S IgY, which has a truncated Fc region. Turtles from Florida were uniformly seropositive to ChHV5 regardless of tumor status. In contrast, in turtles from Hawaii, we detected strong antibody reactivity mainly in tumored animals, with a lower antibody response being seen in nontumored animals, including those from areas where FP is enzootic. Turtles from Hawaii actively shedding ChHV5 were more seropositive than nonshedders. In trying to account for differences in the serological responses to ChHV5 between green turtles from Hawaii and green turtles from Florida, we rejected the cross-reactivity of antibodies to other herpesviruses, differences in viral epitopes, or differences in procedure as likely explanations. Rather, behavioral or other differences between green turtles from Hawaii and green turtles from Florida might have led to the emergence of biologically different viral strains. While the strains from turtles in Florida apparently spread independently of tumors, the transmission of the Hawaiian subtype relies heavily on tumor formation.IMPORTANCE Fibropapillomatosis (FP) is a tumor disease associated with chelonid herpesvirus 5 (ChHV5) that is an important cause of mortality in threatened green turtles globally. FP is expanding in Florida and the Caribbean but declining in Hawaii. We show that Hawaiian turtles mount antibodies to ChHV5 mainly in response to tumors, which are the only sites of viral replication, whereas tumored and nontumored Floridian turtles are uniformly seropositive. Tumor viruses that depend on tumors for replication and spread are rare, with the only example being the retrovirus causing walleye dermal sarcoma in fish. The Hawaiian strain of ChHV5 may be the first DNA virus with such an unusual life history. Our findings, along with the fundamental differences in the life histories between Floridian turtles and Hawaiian turtles, may partly explain the differential dynamics of FP between the two regions.


Assuntos
Alphaherpesvirinae/imunologia , Formação de Anticorpos/imunologia , Tartarugas/imunologia , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Animais , Vírus de DNA , Florida , Glicoproteínas/imunologia , Havaí , Herpesviridae/genética , Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Papiloma/virologia , Filogenia , Neoplasias Cutâneas/virologia , Infecções Tumorais por Vírus/virologia , Tartarugas/virologia
8.
J Virol ; 94(5)2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31801854

RESUMO

The Herpesviridae encode many conserved genes, including the conserved herpesvirus protein kinase (CHPK) that has multifunctional properties. In most cases, herpesviruses lacking CHPK can propagate in cell culture to various degrees, depending on the virus and cell culture system. However, in the natural animal model system of Marek's disease alphaherpesvirus (MDV) in chickens, CHPK is absolutely required for interindividual spread from chicken to chicken. The lack of biological reagents for chicken and MDV has limited our understanding of this important gene during interindividual spread. Here, we engineered epitope-tagged proteins in the context of virus infection in order to detect CHPK in the host. Using immunofluorescence assays and Western blotting during infection in cell culture and in chickens, we determined that the invariant lysine 170 (K170) of MDV CHPK is required for interindividual spread and autophosphorylation of CHPK and that mutation to methionine (M170) results in instability of the CHPK protein. Using these newly generated viruses allowed us to examine the expression of CHPK in infected chickens, and these results showed that mutant CHPK localization and late viral protein expression were severely affected in feather follicles wherein MDV is shed, providing important information on the requirement of CHPK for interindividual spread.IMPORTANCE Marek's disease in chickens is caused by Gallid alphaherpesvirus 2, better known as Marek's disease alphaherpesvirus (MDV). Current vaccines only reduce tumor formation but do not block interindividual spread from chicken to chicken. Understanding MDV interindividual spread provides important information for the development of potential therapies to protect against Marek's disease while also providing a reliable natural host in order to study herpesvirus replication and pathogenesis in animals. Here, we studied the conserved Herpesviridae protein kinase (CHPK) in cell culture and during infection in chickens. We determined that MDV CHPK is not required for cell-to-cell spread, for disease induction, and for oncogenicity. However, it is required for interindividual spread, and mutation of the invariant lysine (K170) results in stability issues and aberrant expression in chickens. This study is important because it addresses the critical role CHPK orthologs play in the natural host.


Assuntos
Alphaherpesvirinae/metabolismo , Galinhas/virologia , Herpesviridae/metabolismo , Doença de Marek/virologia , Proteínas Quinases/metabolismo , Proteínas Virais/metabolismo , Animais , Epitopos , Herpesvirus Galináceo 2 , Doença de Marek/transmissão , Modelos Moleculares , Doenças das Aves Domésticas/virologia , Proteínas Quinases/química , Proteínas Quinases/genética , Pele/patologia , Pele/virologia , Proteínas Virais/química , Proteínas Virais/genética
9.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31748393

RESUMO

Viruses may hijack glycolysis, glutaminolysis, or fatty acid ß-oxidation of host cells to provide the energy and macromolecules required for efficient viral replication. Marek's disease virus (MDV) causes a deadly lymphoproliferative disease in chickens and modulates metabolism of host cells. Metabolic analysis of MDV-infected chicken embryonic fibroblasts (CEFs) identified elevated levels of metabolites involved in glutamine catabolism, such as glutamic acid, alanine, glycine, pyrimidine, and creatine. In addition, our results demonstrate that glutamine uptake is elevated by MDV-infected cells in vitro Although glutamine, but not glucose, deprivation significantly reduced cell viability in MDV-infected cells, both glutamine and glucose were required for virus replication and spread. In the presence of minimum glutamine requirements based on optimal cell viability, virus replication was partially rescued by the addition of the tricarboxylic acid (TCA) cycle intermediate, α-ketoglutarate, suggesting that exogenous glutamine is an essential carbon source for the TCA cycle to generate energy and macromolecules required for virus replication. Surprisingly, the inhibition of carnitine palmitoyltransferase 1a (CPT1a), which is elevated in MDV-infected cells, by chemical (etomoxir) or physiological (malonyl-CoA) inhibitors, did not reduce MDV replication, indicating that MDV replication does not require fatty acid ß-oxidation. Taken together, our results demonstrate that MDV infection activates anaplerotic substrate from glucose to glutamine to provide energy and macromolecules required for MDV replication, and optimal MDV replication occurs when the cells do not depend on mitochondrial ß-oxidation.IMPORTANCE Viruses can manipulate host cellular metabolism to provide energy and essential biosynthetic requirements for efficient replication. Marek's disease virus (MDV), an avian alphaherpesvirus, causes a deadly lymphoma in chickens and hijacks host cell metabolism. This study provides evidence for the importance of glycolysis and glutaminolysis, but not fatty acid ß-oxidation, as an essential energy source for the replication and spread of MDV. Moreover, it suggests that in MDV infection, as in many tumor cells, glutamine is used for generation of energetic and biosynthetic requirements of the MDV infection, while glucose is used biosynthetically.


Assuntos
Glucose/metabolismo , Glutamina/metabolismo , Mardivirus/fisiologia , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/fisiologia , Animais , Embrião de Galinha , Galinhas/virologia , Glucose/fisiologia , Glutamina/fisiologia , Glicólise/fisiologia , Herpesvirus Galináceo 2/metabolismo , Herpesvirus Galináceo 2/fisiologia , Mardivirus/metabolismo , Doença de Marek/metabolismo , Doença de Marek/virologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia
10.
PLoS Pathog ; 15(1): e1007559, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30682178

RESUMO

Type I interferon response plays a prominent role against viral infection, which is frequently disrupted by viruses. Here, we report Bcl-2 associated transcription factor 1 (Bclaf1) is degraded during the alphaherpesvirus Pseudorabies virus (PRV) and Herpes simplex virus type 1 (HSV-1) infections through the viral protein US3. We further reveal that Bclaf1 functions critically in type I interferon signaling. Knockdown or knockout of Bclaf1 in cells significantly impairs interferon-α (IFNα) -mediated gene transcription and viral inhibition against US3 deficient PRV and HSV-1. Mechanistically, Bclaf1 maintains a mechanism allowing STAT1 and STAT2 to be efficiently phosphorylated in response to IFNα, and more importantly, facilitates IFN-stimulated gene factor 3 (ISGF3) binding with IFN-stimulated response elements (ISRE) for efficient gene transcription by directly interacting with ISRE and STAT2. Our studies establish the importance of Bclaf1 in IFNα-induced antiviral immunity and in the control of viral infections.


Assuntos
Interferons/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Virais/metabolismo , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/patogenicidade , Animais , Antivirais/farmacologia , Linhagem Celular , China , Herpesvirus Humano 1/metabolismo , Herpesvirus Suídeo 1/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon Tipo I/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade alfa/metabolismo , Interferon-alfa/metabolismo , Interferons/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Repressoras/fisiologia , Elementos de Resposta , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais/imunologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas Virais/genética , Viroses/genética
11.
J Gen Virol ; 101(3): 284-289, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31958050

RESUMO

Infections with human herpesviruses share several molecular characteristics, but the diversified medical outcomes are distinct to viral subfamilies and species. Notably, both clinical and molecular correlates of infection are a challenging field and distinct patterns of virus-host interaction have rarely been defined; this study therefore focuses on the search for virus-specific molecular indicators. As previous studies have demonstrated the impact of herpesvirus infections on changes in host signalling pathways, we illustrate virus-modulated expression levels of individual cellular protein kinases. Current data reveal (i) α-, ß- and γ-herpesvirus-specific patterns of kinase modulation as well as (ii) differential levels of up-/downregulated kinase expression and phosphorylation, which collectively suggest (iii) defined signalling patterns specific for the various viruses (VSS) that may prove useful for defining molecular indicators. Combined, the study confirms the correlation between herpesviral replication and modulation of signalling kinases, possibly exploitable for the in vitro characterization of viral infections.


Assuntos
Alphaherpesvirinae/metabolismo , Betaherpesvirinae/metabolismo , Fibroblastos/metabolismo , Gammaherpesvirinae/metabolismo , Infecções por Herpesviridae/metabolismo , Linfócitos/metabolismo , Proteínas Quinases/metabolismo , Replicação Viral/fisiologia , Células Cultivadas , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno , Humanos , Fosforilação , Transdução de Sinais/fisiologia , Regulação para Cima
12.
Rev Med Virol ; 28(3): e1972, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29573302

RESUMO

Herpesviruses (Herpesvirales) and tailed bacteriophages (Caudovirales) package their dsDNA genomes through an evolutionarily conserved mechanism. Much is known about the biochemistry and structural biology of phage portal proteins and the DNA encapsidation (viral genome cleavage and packaging) process. Although not at the same level of detail, studies on HSV-1, CMV, VZV, and HHV-8 have revealed important information on the function and structure of herpesvirus portal proteins. During dsDNA phage and herpesviral genome replication, concatamers of viral dsDNA are cleaved into single length units by a virus-encoded terminase and packaged into preformed procapsids through a channel located at a single capsid vertex (portal). Oligomeric portals are formed by the interaction of identical portal protein monomers. Comparing portal protein primary aa sequences between phage and herpesviruses reveals little to no sequence similarity. In contrast, the secondary and tertiary structures of known portals are remarkable. In all cases, function is highly conserved in that portals are essential for DNA packaging and also play a role in releasing viral genomic DNA during infection. Preclinical studies have described small molecules that target the HSV-1 and VZV portals and prevent viral replication by inhibiting encapsidation. This review summarizes what is known concerning the structure and function of herpesvirus portal proteins primarily based on their conserved bacteriophage counterparts and the potential to develop novel portal-specific DNA encapsidation inhibitors.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Herpesviridae/metabolismo , Alphaherpesvirinae/classificação , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/ultraestrutura , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Proteínas do Capsídeo/genética , Herpesviridae/classificação , Herpesviridae/genética , Herpesviridae/ultraestrutura , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/virologia , Humanos , Modelos Moleculares , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Relação Estrutura-Atividade
13.
Virol J ; 15(1): 12, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29334975

RESUMO

BACKGROUND: Duck enteritis virus (DEV) belongs to the subfamily Alphaherpesvirinae, and information on the DEV UL41 gene is limited. METHODS: The DEV UL41 gene was cloned into the pET32a(+) vector and expressed in a prokaryotic expression system. Antiserum was raised against a bacterially expressed UL41-His fusion protein for further experiments. Transcription was quantified and UL41 protein expression levels were determined in DEV-infected cells at different time points by RT-qPCR and western blotting, respectively. DEV virions were purified by sucrose gradient centrifugation and analyzed by mass spectrometry to identify protein content. We confirmed the DEV UL41 gene kinetic class using a pharmacological test. IFA was used to analyze the intracellular localization of pUL41. RESULTS: The recombinant expression plasmid, pET-32a(+)-UL41, which highly expresses a 76.0 kDa fusion protein, was constructed and expressed in E. coli BL21 (DE3) after induction with 0.2 mM IPTG at 30 °C for 10 h, generating a specific mouse anti-UL41 protein polyclonal antibody. RT-qPCR and western blot analyses revealed that the UL41 transcript number peaked at 36 hpi, and peak protein expression occurred at 48 hpi. The pharmacological test showed that UL41 was a γ2 gene. Mass spectrometry analysis showed that pUL41 was a virion component. IFA results revealed that pUL41 was localized throughout DEV-infected cells but only localized to the cytoplasm of transfected cells. DEV pUL47 translocated pUL41 to the nuclei of DEF cells; this translocation was dependent on predicted pUL47 NLS signals (40-50 aa and 768-777 aa). CONCLUSIONS: DEV UL41 is a γ2 gene that encodes a virion structural protein, pUL41 localizes throughout DEV-infected cells but only localizes to the cytoplasm of transfected cells. pUL41 cannot autonomously localize to the nucleus, as this nuclear localization is dependent on predicted DEV pUL47 NLS signals (40-50 aa and 768-777 aa).


Assuntos
Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Patos/virologia , Infecções por Herpesviridae/veterinária , Doenças das Aves Domésticas/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica , Vetores Genéticos/genética , Transporte Proteico , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transcrição Gênica
14.
Genet Mol Res ; 13(2): 4505-15, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-25036355

RESUMO

Glycoprotein C is one of the duck plague virus (DPV) glycoproteins and is encoded by the DPV UL44 gene. DPV glycoprotein C (DPV-gC) comprises 431 amino acids with a putative molecular mass of 47.35 kDa. Sequence analysis indicated that the protein possesses typical characteristics of type-I membrane glycoproteins, containing an N-terminal signal sequence, an external domain, a C-terminal membrane anchor region, and a short cytoplasmic domain. Comparisons of 22 alphaherpesvirus-gC protein sequences revealed eight conservative Cys-residue sites, which may play a crucial role in the biological functions and structural stabilization of the DPV-gC protein. Estimates of potential antigenic epitopes and secondary structure identified four B cell dominant epitopes, which are located at amino acids 68-71, 87-91, 369-352, and 372-374. A model for the structure of DPV-gC was derived by associating its predicted secondary and three-dimensional structures. In conclusion, these results will provide a basis for further functional studies of DPV-gC, establishing novel clinical diagnoses of DPV, and in the development of a new DPV vaccine.


Assuntos
Alphaherpesvirinae/metabolismo , Biologia Computacional/métodos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Mapeamento de Epitopos , Genes Virais , Modelos Moleculares , Filogenia , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
15.
mBio ; 15(8): e0144524, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38953638

RESUMO

Neurotropic alphaherpesviruses, including herpes simplex virus type 1 and pseudorabies virus, establish a lifelong presence within the peripheral nervous system of their mammalian hosts. Upon entering cells, two conserved tegument proteins, pUL36 and pUL37, traffic DNA-containing capsids to nuclei. These proteins support long-distance retrograde axonal transport and invasion of the nervous system in vivo. To better understand how pUL36 and pUL37 function, recombinant viral particles carrying BioID2 fused to these proteins were produced to biotinylate cellular proteins in their proximity (<10 nm) during infection. Eighty-six high-confidence host proteins were identified by mass spectrometry and subsequently targeted by CRISPR-Cas9 gene editing to assess their contributions to early infection. Proteins were identified that both supported and antagonized infection in immortalized human epithelial cells. The latter included zyxin, a protein that localizes to focal adhesions and regulates actin cytoskeletal dynamics. Zyxin knockout cells were hyper-permissive to infection and could be rescued with even modest expression of GFP-zyxin. These results provide a resource for studies of the virus-cell interface and identify zyxin as a novel deterrent to alphaherpesvirus infection.IMPORTANCENeuroinvasive alphaherpesviruses are highly prevalent with many members found across mammals [e.g., herpes simplex virus type 1 (HSV-1) in humans and pseudorabies virus in pigs]. HSV-1 causes a range of clinical manifestations from cold sores to blindness and encephalitis. There are no vaccines or curative therapies available for HSV-1. A fundamental feature of these viruses is their establishment of lifelong infection of the nervous system in their respective hosts. This outcome is possible due to a potent neuroinvasive property that is coordinated by two proteins: pUL36 and pUL37. In this study, we explore the cellular protein network in proximity to pUL36 and pUL37 during infection and examine the impact of knocking down the expression of these proteins upon infection.


Assuntos
Biotina , Humanos , Biotina/metabolismo , Zixina/metabolismo , Zixina/genética , Animais , Linhagem Celular , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/fisiologia , Interações Hospedeiro-Patógeno , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Sistemas CRISPR-Cas , Células Epiteliais/virologia , Células Epiteliais/metabolismo
16.
Virology ; 597: 110159, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38943781

RESUMO

Therapies targeting virus-host interactions are seen as promising strategies for treating gallid alphaherpesvirus 1 (ILTV) infection. Our study revealed a biphasic activation of two MAPK cascade pathways, MEK/ERK and p38 MAPK, as a notably activated host molecular event in response to ILTV infection. It exhibits antiviral functions at different stages of infection. Initially, the MEK/ERK pathway is activated upon viral invasion, leading to a broad suppression of metabolic pathways crucial for ILTV replication, thereby inhibiting viral replication from the early stage of ILTV infection. As the viral replication progresses, the p38 MAPK pathway activates its downstream transcription factor, STAT1, further hindering viral replication. Interestingly, ILTV overcomes this biphasic antiviral barrier by hijacking host p38-AKT axis, which protects infected cells from the apoptosis induced by infection and establishes an intracellular equilibrium conducive to extensive ILTV replication. These insights could provide potential therapeutic targets for ILTV infection.


Assuntos
Infecções por Herpesviridae , Sistema de Sinalização das MAP Quinases , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Infecções por Herpesviridae/virologia , Infecções por Herpesviridae/metabolismo , Alphaherpesvirinae/fisiologia , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Interações Hospedeiro-Patógeno , Linhagem Celular , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/genética
17.
PLoS Pathog ; 7(10): e1002282, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22022263

RESUMO

Alphaherpesviruses are widespread in the human population, and include herpes simplex virus 1 (HSV-1) and 2, and varicella zoster virus (VZV). These viral pathogens cause epithelial lesions, and then infect the nervous system to cause lifelong latency, reactivation, and spread. A related veterinary herpesvirus, pseudorabies (PRV), causes similar disease in livestock that result in significant economic losses. Vaccines developed for VZV and PRV serve as useful models for the development of an HSV-1 vaccine. We present full genome sequence comparisons of the PRV vaccine strain Bartha, and two virulent PRV isolates, Kaplan and Becker. These genome sequences were determined by high-throughput sequencing and assembly, and present new insights into the attenuation of a mammalian alphaherpesvirus vaccine strain. We find many previously unknown coding differences between PRV Bartha and the virulent strains, including changes to the fusion proteins gH and gB, and over forty other viral proteins. Inter-strain variation in PRV protein sequences is much closer to levels previously observed for HSV-1 than for the highly stable VZV proteome. Almost 20% of the PRV genome contains tandem short sequence repeats (SSRs), a class of nucleic acids motifs whose length-variation has been associated with changes in DNA binding site efficiency, transcriptional regulation, and protein interactions. We find SSRs throughout the herpesvirus family, and provide the first global characterization of SSRs in viruses, both within and between strains. We find SSR length variation between different isolates of PRV and HSV-1, which may provide a new mechanism for phenotypic variation between strains. Finally, we detected a small number of polymorphic bases within each plaque-purified PRV strain, and we characterize the effect of passage and plaque-purification on these polymorphisms. These data add to growing evidence that even plaque-purified stocks of stable DNA viruses exhibit limited sequence heterogeneity, which likely seeds future strain evolution.


Assuntos
Alphaherpesvirinae/genética , Variação Genética , Herpesvirus Suídeo 1/genética , Repetições de Microssatélites , Vacinas contra Pseudorraiva/genética , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/patogenicidade , Sequência de Aminoácidos , Animais , Sequência de Bases , Genoma Viral , Herpesvirus Humano 1/genética , Herpesvirus Humano 3/genética , Vacinas contra Herpesvirus/genética , Vacinas contra Herpesvirus/imunologia , Humanos , Dados de Sequência Molecular , Polimorfismo Genético , Alinhamento de Sequência , Análise de Sequência de DNA , Proteínas Virais/genética
18.
J Virol ; 85(1): 568-81, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20962082

RESUMO

The protein kinase found in the short region of alphaherpesviruses, termed US3 in herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV) and ORF66 in varicella-zoster virus (VZV), affects several viral and host cell processes, and its specific targets remain an area of active investigation. Reports suggesting that HSV-1 US3 substrates overlap with those of cellular protein kinase A (PKA) prompted the use of an antibody specific for phosphorylated PKA substrates to identify US3/ORF66 targets. HSV-1, VZV, and PRV induced very different substrate profiles that were US3/ORF66 kinase dependent. The predominant VZV-phosphorylated 125-kDa species was identified as matrin 3, one of the major nuclear matrix proteins. Matrin 3 was also phosphorylated by HSV-1 and PRV in a US3 kinase-dependent manner and by VZV ORF66 kinase at a novel residue (KRRRT150EE). Since VZV-directed T150 phosphorylation was not blocked by PKA inhibitors and was not induced by PKA activation, and since PKA predominantly targeted matrin 3 S188, it was concluded that phosphorylation by VZV was PKA independent. However, purified VZV ORF66 kinase did not phosphorylate matrin 3 in vitro, suggesting that additional cellular factors were required. In VZV-infected cells in the absence of the ORF66 kinase, matrin 3 displayed intranuclear changes, while matrin 3 showed a pronounced cytoplasmic distribution in late-stage cells infected with US3-negative HSV-1 or PRV. This work identifies phosphorylation of the nuclear matrix protein matrin 3 as a new conserved target of this kinase group.


Assuntos
Alphaherpesvirinae/enzimologia , Regulação da Expressão Gênica , Proteínas Associadas à Matriz Nuclear/metabolismo , Fases de Leitura Aberta/fisiologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/metabolismo , Alphaherpesvirinae/classificação , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Linhagem Celular , Fibroblastos/virologia , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Herpesvirus Suídeo 1/enzimologia , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/metabolismo , Herpesvirus Humano 3/enzimologia , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/metabolismo , Humanos , Rim/citologia , Rim/virologia , Proteínas Associadas à Matriz Nuclear/genética , Fosforilação , Proteínas Quinases/genética , Proteínas de Ligação a RNA/genética
19.
Proc Natl Acad Sci U S A ; 106(21): 8707-12, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19435845

RESUMO

The US3 protein is a viral serine/threonine kinase that is conserved among all members of the Alphaherpesvirinae. The US3 protein of different alphaherpesviruses causes dramatic alterations in the actin cytoskeleton, such as the disassembly of actin stress fibers and formation of cell projections, which have been associated with increased intercellular virus spread. Here, we find that inhibiting group A p21-activated kinases (PAKs), which are key regulators in Cdc42/Rac1 Rho GTPase signaling pathways, impairs US3-mediated actin alterations. By using PAK1(-/-) and PAK2(-/-) mouse embryo fibroblasts (MEFs), we show that US3-mediated stress fiber disassembly requires PAK2, whereas US3-mediated cell projection formation mainly is mediated by PAK1, also indicating that PAK1 and PAK2 can have different biological effects on the organization of the actin cytoskeleton. In addition, US3 was found to bind and phosphorylate group A PAKs. Lack of group A PAKs in MEFs was correlated with inefficient virus spread. Thus, US3 induces its effect on the actin cytoskeleton via group A PAKs.


Assuntos
Actinas/metabolismo , Alphaherpesvirinae/metabolismo , Citoesqueleto/metabolismo , Proteínas Virais/metabolismo , Quinases Ativadas por p21/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Alphaherpesvirinae/genética , Animais , Células Cultivadas , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Virais/genética , Quinases Ativadas por p21/deficiência , Quinases Ativadas por p21/genética
20.
Viruses ; 13(8)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34452486

RESUMO

The alphaherpesviruses are pathogens of the mammalian nervous system. Initial infection is commonly at mucosal epithelia, followed by spread to, and establishment of latency in, the peripheral nervous system. During productive infection, viral gene expression, replication of the dsDNA genome, capsid assembly and genome packaging take place in the infected cell nucleus, after which mature nucleocapsids emerge into the cytoplasm. Capsids must then travel to their site of envelopment at cytoplasmic organelles, and enveloped virions need to reach the cell surface for release and spread. Transport at each of these steps requires movement of alphaherpesvirus particles through a crowded and viscous cytoplasm, and for distances ranging from several microns in epithelial cells, to millimeters or even meters during egress from neurons. To solve this challenging problem alphaherpesviruses, and their assembly intermediates, exploit microtubule- and actin-dependent cellular motors. This review focuses upon the mechanisms used by alphaherpesviruses to recruit kinesin, myosin and dynein motors during assembly and egress.


Assuntos
Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Dineínas/metabolismo , Cinesinas/metabolismo , Miosinas/metabolismo , Montagem de Vírus , Liberação de Vírus , Actinas/metabolismo , Alphaherpesvirinae/patogenicidade , Capsídeo/metabolismo , Dineínas/genética , Interações entre Hospedeiro e Microrganismos , Humanos , Cinesinas/genética , Microtúbulos/metabolismo , Miosinas/genética , Vírion/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA