Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
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 ; 20(1): e1011956, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38295116

RESUMO

Viral infection is a significant risk factor for fertility issues. Here, we demonstrated that infection by neurotropic alphaherpesviruses, such as pseudorabies virus (PRV), could impair female fertility by disrupting the hypothalamus-pituitary-ovary axis (HPOA), reducing progesterone (P4) levels, and consequently lowering pregnancy rates. Our study revealed that PRV exploited the transient receptor potential mucolipin 1 (TRPML1) and its lipid activator, phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), to facilitate viral entry through lysosomal cholesterol and Ca2+. P4 antagonized this process by inducing lysosomal storage disorders and promoting the proteasomal degradation of TRPML1 via murine double minute 2 (MDM2)-mediated polyubiquitination. Overall, the study identifies a novel mechanism by which PRV hijacks the lysosomal pathway to evade P4-mediated antiviral defense and impair female fertility. This mechanism may be common among alphaherpesviruses and could contribute significantly to their impact on female reproductive health, providing new insights for the development of antiviral therapies.


Assuntos
Herpesvirus Suídeo 1 , Pseudorraiva , Feminino , Camundongos , Animais , Herpesvirus Suídeo 1/fisiologia , Progesterona/farmacologia , Progesterona/metabolismo , Internalização do Vírus , Lisossomos/metabolismo , Antivirais/metabolismo , Pseudorraiva/metabolismo
3.
FASEB J ; 36(3): e22221, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35199383

RESUMO

The DNA damage response (DDR) pathway is critical for maintaining genomic integrity and sustaining organismal development. Viruses can either utilize or circumvent the DDR to facilitate their replication. Pseudorabies virus (PRV) infection was shown to induce apoptosis via stimulating DDR. However, the underlying mechanisms have not been fully explored to date. This study showed that PRV infection robustly activates the ATM and DNA-PK signaling pathways shortly after infection. However, inhibition of ATM, but not DNA-PK, could dampen PRV replication in cells. Importantly, we found that PRV-encoded serine/threonine kinase UL13 interacts with and subsequently phosphorylates H2AX. Furthermore, we found that UL13 deletion largely attenuates PRV neuroinvasiveness and virulence in vivo. In addtion, we showed that UL13 contributes to H2AX phosphorylation upon PRV infection both in vitro and in vivo, but does not affect ATM phosphorylation. Finally, we showed that knockdown of H2AX reduces PRV replication, while this reduction can be further enhanced by deletion of UL13. Taken together, we conclude that PRV-encoded kinase UL13 regulates DNA damage marker γH2AX and UL13-mediated H2AX phosphorylation plays a pivotal role in efficient PRV replication and progeny production.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Histonas/metabolismo , Proteínas Quinases/metabolismo , Pseudorraiva/virologia , Proteínas Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Herpesvirus Suídeo 1/patogenicidade , Herpesvirus Suídeo 1/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Proteínas Quinases/genética , Pseudorraiva/metabolismo , Suínos , Células Vero , Proteínas Virais/genética
4.
Vet Microbiol ; 265: 109327, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34986434

RESUMO

Pseudorabies is caused by pseudorabies virus (PRV), a member of the Herpesvirus family, and has caused tremendous damage to the pig industry. Protein unique lone 16 (pUL16) is a conserved envelope protein in all herpesviruses, that is known to play an important role in several aspects, including virus diffusion in cells and virulence in mice. It has been shown that the pUL16 can interact with the virus proteins UL11, UL49, UL21, gD, and gE. However, the research to date on pUL16 has only focused on etiology, without discussing the possible cellular pathways involved in PRV infection. Leucine-rich PPR motif-containing protein (LRPPRC) is a multifunctional cellular protein that participates in various cellular processes, such as RNA processing, splicing, stabilization, editing, translation, and energy metabolism. This was the first caspase-independent apoptosis protein to be identified. In this study, immune precipitation and mass spectrometry was performed to define the function of the pUL16 in PRV infection to study the possible cellular pathways in which pUL16 may participate. It was found that LRPRRC could interact with PRV pUL16, which may indicate that UL16 is involved in a redox reaction or cellular apoptosis. This is the first study of the interaction between pUL16 and host proteins, which has positive significance to gain a further understanding of the pUL16.


Assuntos
Herpesvirus Suídeo 1 , Proteínas de Neoplasias , Pseudorraiva , Proteínas Virais , Proteínas Virais Reguladoras e Acessórias , Animais , Proliferação de Células , Camundongos , Proteínas de Neoplasias/metabolismo , Pseudorraiva/metabolismo , Pseudorraiva/virologia , Suínos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Virulência
5.
PLoS Pathog ; 17(11): e1010117, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34843605

RESUMO

Plasmacytoid dendritic cells (pDC) are important innate immune cells during the onset of viral infections as they are specialized in the production of massive amounts of antiviral type I interferon (IFN). Alphaherpesviruses such as herpes simplex virus (HSV) or pseudorabies virus (PRV) are double stranded DNA viruses and potent stimulators of pDC. Detailed information on how PRV activates porcine pDC is lacking. Using PRV and porcine primary pDC, we report here that PRV virions, so-called heavy (H-)particles, trigger IFNα production by pDC, whereas light (L-) particles that lack viral DNA and capsid do not. Activation of pDC requires endosomal acidification and, importantly, depends on the PRV gD envelope glycoprotein and O-glycosylations. Intriguingly, both for PRV and HSV-1, we found that L-particles suppress H-particle-mediated activation of pDC, a process which again depends on viral gD. This is the first report describing that gD plays a critical role in alphaherpesvirus-induced pDC activation and that L-particles directly interfere with alphaherpesvirus-induced IFNα production by pDC.


Assuntos
Células Dendríticas/imunologia , Herpes Simples/imunologia , Interferon Tipo I/metabolismo , Pseudorraiva/imunologia , Proteínas do Envelope Viral/metabolismo , Vírion/fisiologia , Animais , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/fisiologia , Masculino , Pseudorraiva/metabolismo , Pseudorraiva/virologia , Suínos , Testículo/imunologia , Testículo/metabolismo , Testículo/virologia , Proteínas do Envelope Viral/genética
6.
PLoS Pathog ; 16(6): e1008597, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32511265

RESUMO

During infection of neurons by alphaherpesviruses including Pseudorabies virus (PRV) and Herpes simplex virus type 1 (HSV-1) viral nucleocapsids assemble in the cell nucleus, become enveloped in the cell body then traffic into and down axons to nerve termini for spread to adjacent epithelia. The viral membrane protein US9p and the membrane glycoprotein heterodimer gE/gI play critical roles in anterograde spread of both HSV-1 and PRV, and several models exist to explain their function. Biochemical studies suggest that PRV US9p associates with the kinesin-3 motor KIF1A in a gE/gI-stimulated manner, and the gE/gI-US9p complex has been proposed to recruit KIF1A to PRV for microtubule-mediated anterograde trafficking into or along the axon. However, as loss of gE/gI-US9p essentially abolishes delivery of alphaherpesviruses to the axon it is difficult to determine the microtubule-dependent trafficking properties and motor-composition of Δ(gE/gI-US9p) particles. Alternatively, studies in HSV-1 have suggested that gE/gI and US9p are required for the appearance of virions in the axon because they act upstream, to help assemble enveloped virions in the cell body. We prepared Δ(gE/gI-US9p) mutant, and control parental PRV particles from differentiated cultured neuronal or porcine kidney epithelial cells and quantitated the efficiency of virion assembly, the properties of microtubule-dependent transport and the ability of viral particles to recruit kinesin motors. We find that loss of gE/gI-US9p has no significant effect upon PRV particle assembly but leads to greatly diminished plus end-directed traffic, and enhanced minus end-directed and bidirectional movement along microtubules. PRV particles prepared from infected differentiated mouse CAD neurons were found to be associated with either kinesin KIF1A or kinesin KIF5C, but not both. Loss of gE/gI-US9p resulted in failure to recruit KIF1A and KF5C, but did not affect dynein binding. Unexpectedly, while KIF5C was expressed in undifferentiated and differentiated CAD neurons it was only found associated with PRV particles prepared from differentiated cells.


Assuntos
Herpesvirus Suídeo 1 , Peptídeos e Proteínas de Sinalização Intracelular , Cinesinas/metabolismo , Lipoproteínas , Microtúbulos/metabolismo , Pseudorraiva , Proteínas do Envelope Viral , Proteínas Virais , Liberação de Vírus , Animais , Transporte Biológico Ativo , Linhagem Celular , Deleção de Genes , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinesinas/genética , Lipoproteínas/genética , Lipoproteínas/metabolismo , Microtúbulos/genética , Microtúbulos/virologia , Pseudorraiva/genética , Pseudorraiva/metabolismo , Pseudorraiva/patologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
7.
Int J Mol Sci ; 21(9)2020 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-32365661

RESUMO

Bcl2-associated athanogene (BAG) 3, which is a chaperone-mediated selective autophagy protein, plays a pivotal role in modulating the life cycle of a wide variety of viruses. Both positive and negative modulations of viruses by BAG3 were reported. However, the effects of BAG3 on pseudorabies virus (PRV) remain unknown. To investigate whether BAG3 could modulate the PRV life cycle during a lytic infection, we first identified PRV protein UL56 (pUL56) as a novel BAG3 interactor by co-immunoprecipitation and co-localization analyses. The overexpression of pUL56 induced a significant degradation of BAG3 at protein level via the lysosome pathway. The C-terminal mutations of 181L/A, 185L/A, or 181L/A-185L/A in pUL56 resulted in a deficiency in pUL56-induced BAG3 degradation. In addition, the pUL56 C-terminal mutants that lost Golgi retention abrogated pUL56-induced BAG3 degradation, which indicates a Golgi retention-dependent manner. Strikingly, BAG3 was not observed to be degraded in either wild-type or UL56-deleted PRV infected cells as compared to mock infected ones, whereas the additional two adjacent BAG3 cleaved products were found in the infected cells in a species-specific manner. Overexpression of BAG3 significantly suppressed PRV proliferation, while knockdown of BAG3 resulted in increased viral yields in HEK293T cells. Thus, these data indicated a negative regulation role of BAG3 during PRV lytic infection. Collectively, our findings revealed a novel molecular mechanism on host protein degradation induced by PRV pUL56. Moreover, we identified BAG3 as a host restricted protein during PRV lytic infection in cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Herpesvirus Suídeo 1/fisiologia , Interações Hospedeiro-Patógeno , Domínios e Motivos de Interação entre Proteínas , Proteínas Estruturais Virais/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Complexo de Golgi/metabolismo , Lisossomos/metabolismo , Modelos Biológicos , Ligação Proteica , Transporte Proteico , Proteólise , Pseudorraiva/metabolismo , Pseudorraiva/virologia , Especificidade da Espécie , Proteínas Estruturais Virais/química
8.
J Virol ; 94(9)2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32075931

RESUMO

Alphaherpesviruses, including pseudorabies virus (PRV), are neuroinvasive pathogens that establish lifelong latency in peripheral ganglia following the initial infection at mucosal surfaces. The establishment of latent infection and subsequent reactivations, during which newly assembled virions are sorted into and transported anterogradely inside axons to the initial mucosal site of infection, rely on axonal bidirectional transport mediated by microtubule-based motors. Previous studies using cultured peripheral nervous system (PNS) neurons have demonstrated that KIF1A, a kinesin-3 motor, mediates the efficient axonal sorting and transport of newly assembled PRV virions. Here we report that KIF1A, unlike other axonal kinesins, is an intrinsically unstable protein prone to proteasomal degradation. Interestingly, PRV infection of neuronal cells leads not only to a nonspecific depletion of KIF1A mRNA but also to an accelerated proteasomal degradation of KIF1A proteins, leading to a near depletion of KIF1A protein late in infection. Using a series of PRV mutants deficient in axonal sorting and anterograde spread, we identified the PRV US9/gE/gI protein complex as a viral factor facilitating the proteasomal degradation of KIF1A proteins. Moreover, by using compartmented neuronal cultures that fluidically and physically separate axons from cell bodies, we found that the proteasomal degradation of KIF1A occurs in axons during infection. We propose that the PRV anterograde sorting complex, gE/gI/US9, recruits KIF1A to viral transport vesicles for axonal sorting and transport and eventually accelerates the proteasomal degradation of KIF1A in axons.IMPORTANCE Pseudorabies virus (PRV) is an alphaherpesvirus related to human pathogens herpes simplex viruses 1 and 2 and varicella-zoster virus. Alphaherpesviruses are neuroinvasive pathogens that establish lifelong latent infections in the host peripheral nervous system (PNS). Following reactivation from latency, infection spreads from the PNS back via axons to the peripheral mucosal tissues, a process mediated by kinesin motors. Here, we unveil and characterize the underlying mechanisms for a PRV-induced, accelerated degradation of KIF1A, a kinesin-3 motor promoting the sorting and transport of PRV virions in axons. We show that PRV infection disrupts the synthesis of KIF1A and simultaneously promotes the degradation of intrinsically unstable KIF1A proteins by proteasomes in axons. Our work implies that the timing of motor reduction after reactivation would be critical because progeny particles would have a limited time window for sorting into and transport in axons for further host-to-host spread.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Cinesinas/metabolismo , Pseudorraiva/metabolismo , Animais , Transporte Axonal/fisiologia , Axônios/virologia , Linhagem Celular , Células Cultivadas , Interações Hospedeiro-Patógeno , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinesinas/fisiologia , Masculino , Microtúbulos/metabolismo , Neurônios/virologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Proteínas do Envelope Viral/genética , Vírion/metabolismo
9.
PLoS Pathog ; 16(1): e1007985, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31995633

RESUMO

Axonal sorting, the controlled passage of specific cargoes from the cell soma into the axon compartment, is critical for establishing and maintaining the polarity of mature neurons. To delineate axonal sorting events, we took advantage of two neuroinvasive alpha-herpesviruses. Human herpes simplex virus 1 (HSV-1) and pseudorabies virus of swine (PRV; suid herpesvirus 1) have evolved as robust cargo of axonal sorting and transport mechanisms. For efficient axonal sorting and subsequent egress from axons and presynaptic termini, progeny capsids depend on three viral membrane proteins (Us7 (gI), Us8 (gE), and Us9), which engage axon-directed kinesin motors. We present evidence that Us7-9 of the veterinary pathogen pseudorabies virus (PRV) form a tripartite complex to recruit Kif1a, a kinesin-3 motor. Based on multi-channel super-resolution and live TIRF microscopy, complex formation and motor recruitment occurs at the trans-Golgi network. Subsequently, progeny virus particles enter axons as enveloped capsids in a transport vesicle. Artificial recruitment of Kif1a using a drug-inducible heterodimerization system was sufficient to rescue axonal sorting and anterograde spread of PRV mutants devoid of Us7-9. Importantly, biophysical evidence suggests that Us9 is able to increase the velocity of Kif1a, a previously undescribed phenomenon. In addition to elucidating mechanisms governing axonal sorting, our results provide further insight into the composition of neuronal transport systems used by alpha-herpesviruses, which will be critical for both inhibiting the spread of infection and the safety of herpesvirus-based oncolytic therapies.


Assuntos
Axônios/virologia , Capsídeo/metabolismo , Herpes Simples/metabolismo , Herpesvirus Humano 1/metabolismo , Herpesvirus Suídeo 1/metabolismo , Cinesinas/metabolismo , Pseudorraiva/metabolismo , Animais , Transporte Axonal , Axônios/metabolismo , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Suídeo 1/genética , Interações Hospedeiro-Patógeno , Humanos , Cinesinas/genética , Ligação Proteica , Pseudorraiva/genética , Pseudorraiva/virologia , Suínos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Rede trans-Golgi/metabolismo , Rede trans-Golgi/virologia
10.
Int J Biol Macromol ; 151: 1181-1193, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-31743714

RESUMO

Interferon-inducible transmembrane proteins (IFITMs) restrict infection by several viruses, such as influenza A virus, West Nile virus and dengue virus. It has not been determined whether porcine IFITMs (pIFITMs) inhibit infection by pseudorabies virus (PRV), an enveloped, double-stranded DNA virus, which is the etiological agent of Aujeszky's disease in pigs. Here, we report that PRV infection elicited pIFITM1 expression in PK15 porcine kidney epithelial cells and 3D4/21 alveolar macrophages. pIFITM2 and pIFITM3 expression was only elevated in PK15 cells during PRV infection. Depletion of pIFITM1 using RNA interference, either in PK15 or in 3D4/21 cells, enhanced PRV infection while overexpression of pIFITM1 had the opposite effect. Knockdown of pIFITM2 and pIFITM3 did not influence PRV infection, suggesting that pIFITM2 and pIFITM3 are independent of PRV infection. PRV-induced pIFITM1 expression was dependent on the cGAS/STING/TBK1/IRF3 innate immune pathway and interferon-alpha receptor-1, suggesting that pIFITM1 is up-regulated by the type I interferon signaling pathway. The anti-PRV role of pIFITM1 was inhibited upon PRV entry. Our data demonstrate that pIFITM1 is a host restriction factor that inhibits PRV entry that may shed light on a strategy for prevention of PRV infection.


Assuntos
Antígenos de Diferenciação/farmacologia , Antivirais/farmacologia , Herpesvirus Suídeo 1/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Linhagem Celular , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Pseudorraiva/genética , Pseudorraiva/metabolismo , Pseudorraiva/virologia , Suínos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
11.
Mol Immunol ; 95: 56-63, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29407577

RESUMO

In a previous study, we demonstrated that porcine cyclic GMP-AMP (cGAMP) synthase (cGAS) catalyzes cGAMP production and is an important DNA sensor for the pseudorabies virus (PRV)-induced activation of interferon ß (IFN-ß). Ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1) has recently been identified as the hydrolase of cGAMP in rodents, but its role in porcine cells is not clear. Our recent study demonstrated that porcine ENPP1 is responsible for the homeostasis of cGAMP and is critical for PRV infection. Porcine ENPP1 mRNA is predominantly expressed in muscle. PRV infection was enhanced by ENPP1 overexpression and attenuated by silencing of ENPP1. During PRV infection, the activation of IFN-ß and NF-κB was reduced in ENPP1 overexpressed cells and promoted in ENPP1 knockdown cells. Investigation of the molecular mechanisms of ENPP1 during PRV infection showed that ENPP1 hydrolyzed cGAMP in PRV-infected or cGAMP-transfected cells and inhibited IRF3 phosphorylation, reducing IFN-ß secretion. These results, combined with those for porcine cGAS, demonstrate that ENPP1 acts coordinately with cGAS to maintain the reservoir of cGAMP and participates in PRV infection.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Diester Fosfórico Hidrolases/fisiologia , Pseudorraiva/metabolismo , Pirofosfatases/fisiologia , Animais , Células Cultivadas , Células HEK293 , Herpesvirus Suídeo 1/fisiologia , Homeostase , Humanos , Suínos
12.
J Food Drug Anal ; 25(4): 908-918, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28987368

RESUMO

Recent investigations have demonstrated that carotenoid extract of Dunaliella salina alga (Alga) contains abundant ß-carotene and has good anti-inflammatory activities. Murine macrophage (RAW264.7 cells) was used to establish as an in vitro model of pseudorabies virus-induced reactive oxygen species (ROS) response. In this study, antioxidant activities of Alga were measured based on 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging, trolox equivalent antioxidant capacity assays, reducing power, and virus-induced ROS formation in RAW264.7 cells. Anti-inflammatory activities of Alga were assessed by its ability to inhibit the production of interleukin-6 and nitric oxide (NO) using enzyme-linked immunosorbent assay, then the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway was investigated by measuring the inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nuclear factor-κB (p50 and p65), JAK, STAT-1/3, and suppressor of cytokine signaling 3 (SOCS3) by Western blotting. In addition, Alga inhibited virus replication by plaque assay. Our results showed that the Alga had high antioxidant activity, significantly reduced the virus-induced accumulation of ROS, and inhibited the levels of nitric oxide and interleukin-6. Further studies revealed that Alga also downregulated the gene and protein expressions of iNOS, COX-2, nuclear factor-κB (p50 and p65), and the JAK/STAT pathway. The inhibitory effects of Alga were similar to pretreatment with specific inhibitors of JAK and STAT-3 in pseudorabies virus -infected RAW264.7 cells. Alga enhanced the expression of SOCS3 to suppress the activity of the JAK/STAT signaling pathway in pseudorabies virus-infected RAW264.7 cells. In addition, Alga has decreased viral replication (p < 0.005) at an early stage. Therefore, our results demonstrate that Alga inhibits ROS, interleukin6, and nitric oxide production via suppression of the JAK/STAT pathways and enhanced the expression of SOCS3 in virus-infected RAW264.7 cells.


Assuntos
Clorófitas/química , Interleucina-6/metabolismo , Janus Quinases/metabolismo , Macrófagos/efeitos dos fármacos , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Herpesvirus Suídeo 1/fisiologia , Interleucina-6/genética , Janus Quinases/genética , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , NF-kappa B/genética , Pseudorraiva/genética , Pseudorraiva/metabolismo , Pseudorraiva/virologia , Células RAW 264.7 , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais
13.
PLoS Pathog ; 13(5): e1006314, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28542478

RESUMO

An early and yet indispensable step in the alphaherpesvirus infection is the engagement of host receptors by the viral envelope glycoprotein D (gD). Of the thus-far identified gD receptors, nectin-1 is likely the most effective in terms of its wide usage by multiple alphaherpesviruses for cell entry. The molecular basis of nectin-1 recognition by the gD protein is therefore an interesting scientific question in the alphaherpesvirus field. Previous studies focused on the herpes simplex virus (HSV) of the Simplexvirus genus, for which both the free gD structure and the gD/nectin-1 complex structure were reported at high resolutions. The structural and functional features of other alphaherpesviral gDs, however, remain poorly characterized. In the current study, we systematically studied the characteristics of nectin-1 binding by the gD of a Varicellovirus genus member, the pseudorabies virus (PRV). We first showed that PRV infects host cells via both human and swine nectin-1, and that its gD exhibits similar binding affinities for nectin-1 of the two species. Furthermore, we demonstrated that removal of the PRV gD membrane-proximal residues could significantly increase its affinity for the receptor binding. The structures of PRV gD in the free and the nectin-1-bound states were then solved, revealing a similar overall 3D fold as well as a homologous nectin-1 binding mode to its HSV counterpart. However, several unique features were observed at the binding interface of PRV gD, enabling the viral ligand to utilize different gD residues (from those of HSV) for nectin-1 engagement. These observed binding characteristics were further verified by the mutagenesis study using the key-residue mutants of nectin-1. The structural and functional data obtained in this study, therefore, provide the basis of receptor recognition by PRV gD.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Nectinas/metabolismo , Pseudorraiva/virologia , Receptores Virais/metabolismo , Doenças dos Suínos/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular , Herpesvirus Suídeo 1/química , Herpesvirus Suídeo 1/genética , Humanos , Nectinas/química , Nectinas/genética , Ligação Proteica , Pseudorraiva/genética , Pseudorraiva/metabolismo , Receptores Virais/química , Receptores Virais/genética , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/metabolismo , Proteínas do Envelope Viral/genética
14.
Bing Du Xue Bao ; 30(4): 436-40, 2014 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-25272600

RESUMO

Tegument protein VP22 is encoded by Pseudorabies Virus (PRV) UL49. To identify the nuclear localization signals of UL49, it is necessary to determine the transport mechanism and biological functions of the VP22 protein. In this study, we identified two nuclear localization signals from UL49, NLS1 (5RKTRVA ADETASGARRR21) and NLS2 (241PGRKGKV247). The functional nuclear localization signal (NLS) of UL49 was identified by constructing truncated or site-specific UL49 mutants. The deletion of both NLS1 and NLS2 abrogated UL49 nuclear accumulation, whereas the deletion of NLS1 or NLS2 did not. Therefore, both NLS1 and NLS2 are critical for the nuclear localization of UL49. And our resuts showed that NLS2 is more important in this regard.


Assuntos
Núcleo Celular/virologia , Herpesvirus Suídeo 1/metabolismo , Sinais de Localização Nuclear , Pseudorraiva/virologia , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo , Animais , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Herpesvirus Suídeo 1/química , Herpesvirus Suídeo 1/genética , Humanos , Transporte Proteico , Pseudorraiva/metabolismo , Proteínas Estruturais Virais/genética
15.
Proc Natl Acad Sci U S A ; 110(37): E3516-25, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23980169

RESUMO

A clinical hallmark of human alphaherpesvirus infections is peripheral pain or itching. Pseudorabies virus (PRV), a broad host range alphaherpesvirus, causes violent pruritus in many different animals, but the mechanism is unknown. Previous in vitro studies have shown that infected, cultured peripheral nervous system (PNS) neurons exhibited aberrant electrical activity after PRV infection due to the action of viral membrane fusion proteins, yet it is unclear if such activity occurs in infected PNS ganglia in living animals and if it correlates with disease symptoms. Using two-photon microscopy, we imaged autonomic ganglia in living mice infected with PRV strains expressing GCaMP3, a genetically encoded calcium indicator, and used the changes in calcium flux to monitor the activity of many neurons simultaneously with single-cell resolution. Infection with virulent PRV caused these PNS neurons to fire synchronously and cyclically in highly correlated patterns among infected neurons. This activity persisted even when we severed the presynaptic axons, showing that infection-induced firing is independent of input from presynaptic brainstem neurons. This activity was not observed after infections with an attenuated PRV recombinant used for circuit tracing or with PRV mutants lacking either viral glycoprotein B, required for membrane fusion, or viral membrane protein Us9, required for sorting virions and viral glycoproteins into axons. We propose that the viral fusion proteins produced by virulent PRV infection induce electrical coupling in unmyelinated axons in vivo. This action would then give rise to the synchronous and cyclical activity in the ganglia and contribute to the characteristic peripheral neuropathy.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Pseudorraiva/metabolismo , Pseudorraiva/virologia , Proteínas Virais/metabolismo , Potenciais de Ação , Animais , Axônios/metabolismo , Axônios/virologia , Sinalização do Cálcio , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/patogenicidade , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipoproteínas/metabolismo , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Nervos Periféricos/metabolismo , Nervos Periféricos/virologia , Doenças do Sistema Nervoso Periférico/etiologia , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/virologia , Fosfoproteínas/metabolismo , Prurido/etiologia , Prurido/metabolismo , Prurido/virologia , Proteínas Recombinantes/metabolismo , Glândula Submandibular/inervação , Glândula Submandibular/virologia , Proteínas do Envelope Viral/metabolismo , Virulência
16.
Cell Host Microbe ; 13(2): 193-203, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23414759

RESUMO

Microtubule transport of herpesvirus capsids from the cell periphery to the nucleus is imperative for viral replication and, in the case of many alphaherpesviruses, transmission into the nervous system. Using the neuroinvasive herpesvirus, pseudorabies virus (PRV), we show that the viral protein 1/2 (VP1/2) tegument protein associates with the dynein/dynactin microtubule motor complex and promotes retrograde microtubule transport of PRV capsids. Functional activation of VP1/2 requires binding to the capsid protein pUL25 or removal of the capsid-binding domain. A proline-rich sequence within VP1/2 is required for the efficient interaction with the dynein/dynactin microtubule motor complex as well as for PRV virulence and retrograde axon transport in vivo. Additionally, in the absence of infection, functionally active VP1/2 is sufficient to move large surrogate cargoes via the dynein/dynactin microtubule motor complex. Thus, VP1/2 tethers PRV capsids to dynein/dynactin to enhance microtubule transport, neuroinvasion, and pathogenesis.


Assuntos
Dineínas/metabolismo , Herpesvirus Suídeo 1/patogenicidade , Células Receptoras Sensoriais/virologia , Proteínas Estruturais Virais/metabolismo , Animais , Axônios/metabolismo , Chlorocebus aethiops , Coinfecção/metabolismo , Coinfecção/virologia , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Herpesvirus Suídeo 1/metabolismo , Humanos , Imunoprecipitação , Masculino , Camundongos , Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Membrana Nuclear/virologia , Prolina/metabolismo , Mapeamento de Interação de Proteínas , Transporte Proteico , Pseudorraiva/metabolismo , Pseudorraiva/patologia , Pseudorraiva/virologia , Ratos , Ratos Long-Evans , Células Receptoras Sensoriais/metabolismo , Células Vero , Ensaio de Placa Viral , Proteínas Estruturais Virais/genética
17.
J Virol ; 86(15): 8002-13, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623768

RESUMO

Glycoprotein H (gH) is an envelope protein conserved in the Herpesviridae. Together with glycoprotein B (gB), the heterodimeric complex of gH and glycoprotein L (gL) mediates penetration and direct viral cell-to-cell spread. In herpes simplex and pseudorabies virus (PrV), coexpression of gH/gL, gB, and gD induces membrane fusion to form polykaryocytes. The recently determined crystal structure of a core fragment of PrV gH revealed marked structural similarity to other gH proteins (M. Backovic et al., Proc. Natl. Acad. Sci. U. S. A. 107:22635-22640, 2010). Within the membrane-proximal part (domain IV), a conserved negatively charged surface loop (flap) is flanked by intramolecular disulfide bonds. Together with an N-linked carbohydrate moiety, this flap covers an underlying patch of hydrophobic residues. To investigate the functional relevance of these structures, nonconservative amino acid substitutions were introduced by site-directed mutagenesis. The mutated proteins were tested for correct expression, fusion activity, and functional complementation of gH-deleted PrV. Several single amino acid changes within the flap and the hydrophobic patch were tolerated, and deletion of the glycosylation site had only minor effects. However, multiple alanine substitutions within the flap or the hydrophobic patch led to significant defects. gH function was also severely affected by disruption of the disulfide bond at the C terminus of the flap and after introduction of cysteine pairs designed to bridge the central part of the flap with the hydrophobic patch. Interestingly, all mutated gH proteins were able to complement gH-deleted PrV, but fusion-deficient gH mutants resulted in a pronounced delay in virus entry.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Fusão de Membrana , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Herpesvirus Suídeo 1/genética , Interações Hidrofóbicas e Hidrofílicas , Mutagênese Sítio-Dirigida , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Pseudorraiva/genética , Pseudorraiva/metabolismo , Proteínas do Envelope Viral/genética
18.
BMC Genomics ; 9: 123, 2008 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-18331636

RESUMO

BACKGROUND: Transcriptomic approaches are relevant for studying virus-host cell dialogues to better understand the physiopathology of infection and the immune response at the cellular level. Pseudorabies virus (PrV), a porcine Alphaherpesvirus, is a good model for such studies in pig. Since PrV displays a strong tropism for mucous epithelial cells, we developed a kinetics study of PrV infection in the porcine PK15 epithelial cell line. To identify as completely as possible, viral and cellular genes regulated during infection, we simultaneously analyzed PrV and cellular transcriptome modifications using two microarrays i.e. a laboratory-made combined SLA/PrV microarray, consisting of probes for all PrV genes and for porcine genes contained in the Swine Leukocyte Antigen (SLA) complex, and the porcine generic Qiagen-NRSP8 oligonucleotide microarray. We confirmed the differential expression of a selected set of genes by qRT-PCR and flow cytometry. RESULTS: An increase in the number of differentially expressed cellular genes and PrV genes especially from 4 h post-infection (pi) was observed concomitantly with the onset of viral progeny while no early global cellular shutoff was recorded. Many cellular genes were down-regulated from 4 h pi and their number increased until 12 h pi. UL41 transcripts encoding the virion host shutoff protein were first detected as differentially expressed at 8 h pi. The viral gene UL49.5 encoding a TAP inhibitor protein was differentially expressed as soon as 2 h pi, indicating that viral evasion via TAP inhibition may start earlier than the cellular gene shutoff. We found that many biological processes are altered during PrV infection. Indeed, several genes involved in the SLA class I antigenic presentation pathway (SLA-Ia, TAP1, TAP2, PSMB8 and PSMB9), were down-regulated, thus contributing to viral immune escape from this pathway and other genes involved in apoptosis, nucleic acid metabolism, cytoskeleton signaling as well as interferon-mediated antiviral response were also modulated during PrV infection. CONCLUSION: Our results show that the gene expression of both PrV and porcine cells can be analyzed simultaneously with microarrays, providing a chronology of PrV gene transcription, which has never been described before, and a global picture of transcription with a direct temporal link between viral and host gene expression.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/virologia , Regulação da Expressão Gênica/imunologia , Herpesvirus Suídeo 1 , Pseudorraiva/metabolismo , Doenças dos Suínos/virologia , Animais , Apoptose/imunologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Complexo Principal de Histocompatibilidade/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Pseudorraiva/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/metabolismo
19.
J Virol ; 81(2): 1048-51, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17079290

RESUMO

The mature virion of the alphaherpesvirus pseudorabies virus (PrV) contains a minimum of 31 structural proteins which are recruited into the virus particle by a network of protein-protein interactions which is only incompletely understood. We show here that deletion of the tegument protein pUL21 resulted in a drastic decrease in the incorporation of the pUL46, pUL49, and pUS3 tegument components into mature virions. Moreover, the attenuated PrV strain Bartha (PrV-Ba), which, among other defects, carries mutations in pUL21, also fails to package pUL46, pUL49, and pUS3 efficiently. By the reconstitution of wild-type pUL21 expression to PrV-Ba and the transfer of mutated PrV-Ba pUL21 into wild-type PrV, we demonstrate that this phenotype is due to the mutated pUL21.


Assuntos
Proteínas do Capsídeo/metabolismo , Herpesvirus Suídeo 1/metabolismo , Pseudorraiva/metabolismo , Proteínas Virais/metabolismo , Vírion/metabolismo , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Herpesvirus Suídeo 1/genética , Espectrometria de Massas , Peptídeos/química , Pseudorraiva/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/química , Montagem de Vírus
20.
J Virol ; 80(14): 7159-68, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16809321

RESUMO

Many different viruses activate the extracellular signal-regulated kinase (ERK)/mitogen-activated protein (MAP) kinase signaling pathway during infection and require ERK activation for the efficient execution of their replication programs. Despite these findings, no virus-encoded proteins have been identified that directly modulate ERK activities. In an effort to determine the function of a conserved alphaherpesvirus structural protein called Us2, we screened a yeast two-hybrid library derived from NIH 3T3 cells and identified ERK as a Us2-interacting protein. Our studies indicate that Us2 binds to ERK in virus-infected cells, mediates the incorporation of ERK into the virion, and inhibits the activation of ERK nuclear substrates. The association of Us2 with ERK leads to the sequestration of ERK at the plasma membrane and to a perinuclear vesicular compartment, thereby keeping ERK out of the nucleus. Us2 can bind to activated ERK, and the data suggest that Us2 does not inhibit ERK enzymatic activity. The treatment of cells with U0126, a specific inhibitor of ERK activation, resulted in a substantial delay in the release of virus from infected cells that was more pronounced with a virus deleted for Us2 than with parental and repaired strains, suggesting that both ERK and Us2 activities are required for efficient virus replication. This study highlights an additional complexity to the activation of ERK by viruses, namely, that localization of active ERK can be altered by virus-encoded proteins.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Herpesvirus Suídeo 1/fisiologia , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Replicação Viral/fisiologia , Animais , Sequência de Bases/genética , Butadienos/farmacologia , Membrana Celular/metabolismo , Membrana Celular/virologia , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/virologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Humanos , Camundongos , Células NIH 3T3 , Nitrilas/farmacologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Pseudorraiva/metabolismo , Deleção de Sequência , Técnicas do Sistema de Duplo-Híbrido , Proteínas do Envelope Viral/genética , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA