首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 62 毫秒
1.
2.
3.
4.
5.
ICP27 is a multifunctional protein that is required for herpes simplex virus 1 mRNA export. ICP27 interacts with the mRNA export receptor TAP/NXF1 and binds RNA through an RGG box motif. Unlike other RGG box proteins, ICP27 does not bind G-quartet structures but instead binds GC-rich sequences that are flexible in structure. To determine the contribution of arginines within the RGG box, we performed in vitro binding assays with N-terminal proteins encoding amino acids 1 to 160 of wild-type ICP27 or arginine-to-lysine substitution mutants. The R138,148,150K triple mutant bound weakly to sequences that were bound by the wild-type protein and single and double mutants. Furthermore, during infection with the R138,148,150K mutant, poly(A)+ RNA and newly transcribed RNA accumulated in the nucleus, indicating that viral RNA export was impaired. To determine if structural changes had occurred, nuclear magnetic resonance (NMR) analysis was performed on N-terminal proteins consisting of amino acids 1 to 160 from wild-type ICP27 and the R138,148,150K mutant. This region of ICP27 was found to be highly flexible, and there were no apparent differences in the spectra seen with wild-type ICP27 and the R138,148,150K mutant. Furthermore, NMR analysis with the wild-type protein bound to GC-rich sequences did not show any discernible folding. We conclude that arginines at positions 138, 148, and 150 within the RGG box of ICP27 are required for binding to GC-rich sequences and that the N-terminal portion of ICP27 is highly flexible in structure, which may account for its preference for binding flexible sequences.The herpes simplex virus 1 (HSV-1) protein ICP27 is a multifunctional regulatory protein that is required for productive viral infection. ICP27 interacts with a number of cellular proteins, and it binds RNA (35). One of the functions that ICP27 performs is to escort viral mRNAs from the nucleus to the cytoplasm for translation (2, 3, 5, 10, 13, 21, 34). ICP27 binds viral RNAs (5, 34) and interacts directly with the cellular mRNA export receptor TAP/NXF1 (2, 21), which is required for the export of HSV-1 mRNAs (20, 21). ICP27 also interacts with the export adaptor proteins Aly/REF (2, 3, 23) and UAP56 (L. A. Johnson, H. Swesey, and R. M. Sandri-Goldin, unpublished results), which form part of the TREX complex that binds to the 5′ end of mRNA through an interaction with CBP80 (26, 32, 41). Aly/REF does not appear to bind viral RNA directly (3), and it is not essential for HSV-1 RNA export based upon small interfering RNA (siRNA) knockdown studies (20), but it contributes to the efficiency of viral RNA export (3, 23). ICP27 also interacts with the SR splicing proteins SRp20 and 9G8 (11, 36), which have been shown to shuttle between the nucleus and the cytoplasm (1). SRp20 and 9G8 have also been shown to facilitate the export of some cellular RNAs (16, 17, 27) by binding RNA and interacting with TAP/NXF1 (14, 16, 18). The knockdown of SRp20 or 9G8 adversely affects HSV-1 replication and specifically results in a nuclear accumulation of newly transcribed RNA during infection (11). Thus, these SR proteins also contribute to the efficiency of viral RNA export. However, the overexpression of SRp20 was unable to rescue the defect in RNA export during infection with an ICP27 mutant that cannot bind RNA (11), suggesting that ICP27 is the major HSV-1 RNA export protein that links viral RNA to TAP/NXF1.ICP27 was shown previously to bind RNA through an RGG box motif located at amino acids 138 to 152 within the 512-amino-acid protein (28, 34). Using electrophoretic mobility shift assays (EMSAs), we showed that the N-terminal portion of ICP27 from amino acids 1 to 160 bound specifically to viral oligonucleotides that are GC rich and that are flexible and relatively unstructured (5). Here we report the importance of three arginine residues within the RGG box for ICP27 binding to GC-rich sequences in vitro and for viral RNA export during infection. We also performed nuclear magnetic resonance (NMR) structural analysis of the N-terminal portion of ICP27 for both the wild-type protein and an ICP27 mutant in which three arginines were replaced with lysines. The NMR data showed that the N-terminal portion of ICP27 is relatively unstructured but compact, and NMR analysis in the presence of oligonucleotide substrates to which the N-terminal portion of ICP27 binds did not show any discernible alterations in this highly flexible structure, nor did the arginine-to-lysine substitutions.  相似文献   

6.
7.
8.
9.
10.
11.
12.
Immediate-early protein ICP0 of herpes simplex virus type 1 (HSV-1) is an E3 ubiquitin ligase of the RING finger class that is required for efficient lytic infection and reactivation from latency. Other alphaherpesviruses also express ICP0-related RING finger proteins, but these have limited homology outside the core RING domain. Existing evidence indicates that ICP0 family members have similar properties, but there has been no systematic comparison of the biochemical activities and biological functions of these proteins. Here, we describe an inducible cell line system that allows expression of the ICP0-related proteins of bovine herpes virus type 1 (BHV-1), equine herpesvirus type 1 (EHV-1), pseudorabies virus (PRV), and varicella-zoster virus (VZV) and their subsequent functional analysis. We report that the RING domains of all the proteins have E3 ubiquitin ligase activity in vitro. The BHV-1, EHV-1, and PRV proteins complement ICP0-null mutant HSV-1 plaque formation and induce derepression of quiescent HSV-1 genomes to levels similar to those achieved by ICP0 itself. VICP0, the ICP0 expressed by VZV, was found to be extremely unstable, which limited its analysis in this system. We compared the abilities of the ICP0-related proteins to disrupt ND10, to induce degradation of PML and Sp100, to affect key components of the interferon signaling pathway, and to interfere with induction of interferon-stimulated genes. We found that the property that correlated most closely with their biological activities was the ability to preclude the recruitment of cellular ND10 proteins to sites closely associated with incoming HSV-1 genomes and early replication compartments.The members of the alphaherpesvirus subfamily are characterized by their ability to establish life-long latent infections in neuronal tissues after the primary infection. Although certain core genes are conserved in all herpesviruses of all subfamilies, there are also genes that are characteristic of particular subfamilies. Among these are the genes that encode the ICP0-related proteins of the alphaherpesviruses, of which the most widely studied is ICP0 of herpes simplex virus type 1 (HSV-1). The interest in ICP0 stems from its biological roles in stimulating lytic infection and reactivation from latency (for reviews, see references 17, 18 and 33). Members of the ICP0 family of proteins are characterized by the presence of a RING finger domain near their N termini, a zinc-stabilized fold that in many other proteins confers E3 ubiquitin ligase activity (43). This has proved to be true of ICP0 (3), and the available evidence indicates that other members of the ICP0 family have similar biochemical functions (13, 61). Although a number of ICP0-related alphaherpesvirus proteins have been studied in a variety of contexts, notably those expressed by bovine herpesvirus 1 (BHV-1), equine herpes virus 1 (EHV-1), pseudorabies virus (PRV), and varicella-zoster virus (VZV), there has been no systematic comparison of their abilities to complement ICP0 null mutant HSV-1 or to induce derepression of quiescent HSV-1 genomes.This paper describes a comparative study of the ICP0-related proteins expressed by the viruses listed above. In terms of nomenclature, the proteins expressed by BHV-1 and EHV-1 have been named BICP0 and EICP0, so although other names have been used for the PRV and VZV proteins (such as EP0 and orf61, respectively), we have adopted the names PICP0 and VICP0 for this study. Previous work found that, like ICP0 itself, all four proteins activate gene expression in reporter assays in a RING finger-dependent manner (4, 5, 8, 29, 38, 41, 45, 51, 54, 59, 64, 75, 76, 78). VICP0 and EICP0 also complement, at least partially, ICP0 null mutant HSV-1 (15, 48, 53, 54). BHV-1, EHV-1, PRV, and VZV mutants in which the ICP0-related genes have been deleted have been isolated and found to have reduced replication efficiencies, as expected by analogy with ICP0 null mutant HSV-1 (2, 7, 11, 12, 30, 46, 74, 77).A prominent property of ICP0 is its localization to and disruption of cellular nuclear substructures known as ND10 or promyelocytic leukemia (PML) nuclear domains. Interactions between ND10 and BICP0, EICP0, PICP0, and VICP0 have also been observed, with various consequences for ND10 integrity (47, 60, 63). Whereas ICP0 achieves ND10 disruption through induction of the degradation of PML and SUMO-modified forms of Sp100 (21, 60), EICP0 appears less efficient than ICP0 in inducing PML degradation (60) while VICP0 is inactive (47). While it is likely that all the ICP0 family members discussed here have RING finger-mediated E3 ubiquitin ligase activity (61), the only other protein for which this has been confirmed is BICP0 (13).The similarities between these members of the ICP0 family of proteins and their apparent differences prompted us to investigate in more detail the properties of these proteins in order to determine which of their properties correlate most closely with biological functions in complementing ICP0 null mutant HSV-1. In addition, there was no existing evidence on whether the related proteins could, like ICP0, induce derepression of gene expression from quiescent HSV-1 genomes. We have taken two approaches to these issues. The first is the use of an inducible cell line system that has been used to study ICP0 itself (24, 26). Although inducible cell line systems have been described for VICP0 and BICP0 (53, 69), much of the work described in the current study is novel. The second approach is in vitro analysis of the E3 ubiquitin ligase activities of the isolated RING finger domains of the proteins. The major findings of the study are the following: (i) that all the proteins studied are active in E3 ubiquitin ligase assays; (ii) that VICP0 is extremely unstable, compromising comparative functional analysis in this system; (iii) that BICP0, EICP0, and PICP0 complement to various degrees the plaque-forming defect of ICP0 null mutant HSV-1; (iv) that these three proteins also efficiently stimulate derepression of gene expression from quiescent HSV-1 genomes; (v) that none of the ICP0 family members impedes interferon (IFN)-induced expression of IFN-stimulated genes (ISGs) or affects the stability of important components of the IFN signaling system (namely STAT1, STAT2, and IRF3); (vi) that BICP0, EICP0, and PICP0 cause some disruption of ND10 integrity and have various effects on PML and Sp100 abundance; and (vii) that the property of the proteins that correlated most closely with their stimulation of ICP0 null mutant HSV-1 infection and derepression of quiescent genomes is their ability to inhibit the recruitment of PML and other ND10 proteins to sites associated with parental HSV-1 genomes and early replication compartments.  相似文献   

13.
14.
Autophagy is an important component of host innate and adaptive immunity to viruses. It is critical for the degradation of intracellular pathogens and for promoting antigen presentation. Herpes simplex virus type 1 (HSV-1) infection induces an autophagy response, but this response is antagonized by the HSV-1 neurovirulence gene product, ICP34.5. This is due, in part, to its interaction with the essential autophagy protein Beclin 1 (Atg6) via the Beclin-binding domain (BBD) of ICP34.5. Using a recombinant virus lacking the BBD, we examined pathogenesis and immune responses using mouse models of infection. The BBD-deficient virus (Δ68H) replicated equivalently to its marker-rescued counterpart (Δ68HR) at early times but was cleared more rapidly than Δ68HR from all tissues at late times following corneal infection. In addition, the infection of the cornea with Δ68H induced less ocular disease than Δ68HR. These results suggested that Δ68H was attenuated due to its failure to control adaptive rather than innate immunity. In support of this idea, Δ68H stimulated a significantly stronger CD4+ T-cell-mediated delayed-type hypersensitivity response and resulted in significantly more production of gamma interferon and interleukin-2 from HSV-specific CD4+ T cells than Δ68HR. Taken together, these data suggest a role for the BBD of ICP34.5 in precluding autophagy-mediated class II antigen presentation, thereby enhancing the virulence and pathogenesis of HSV-1.Autophagy is a conserved cellular pathway that eliminates defective proteins and organelles, prevents abnormal protein aggregate accumulation, and removes intracellular pathogens (11, 22, 32, 56). This process begins with the formation and elongation of a double membrane that fuses to form an autophagosome. The cytoplasmic contents are nonspecifically sequestered inside the autophagosome and then are degraded once the autophagosome fuses with the lysosome. Autophagy is upregulated during starvation, growth factor withdrawal, hypoxia, and infection (10). Following metabolic stress, autophagy can generate metabolic precursors that can be recycled for the de novo synthesis of proteins. The autophagic pathway has important roles in development, immune defense, apoptosis, tumor suppression, and the prevention of neuronal degeneration (reviewed in references 21 and 31).Autophagy is not limited to the degradation of self proteins; it also can engulf and break down invading microorganisms in a process termed xenophagy (26). Xenophagy can limit the replication of pathogens (3, 4, 29, 34, 42), but some infectious agents can exploit autophagy to enhance their replication (2, 18, 38). There are pathogens that actively inhibit autophagy through interaction with the essential autophagy-promoting protein, Beclin 1 (24, 35). Beclin 1 is the mammalian homolog of yeast Atg6 and is required for the formation of the autophagosome membrane through its interaction with VPS34, a class III phosphatidylinositol 3-kinase (19, 27). Autophagy/xenophagy is also an important process for the adaptive immune response to infection through the delivery of antigens for major histocompatibility complex class I and II (MHC-I and -II) presentation (7, 9, 12, 36, 45). The inhibition of autophagy by pathogens therefore would serve to block CD4+ and CD8+ cell responses and allow pathogens to remain underrecognized by the adaptive immune response.The interferon (IFN)-inducible double-strand RNA-inducible protein kinase (PKR) pathway is required for virus- and starvation-induced autophagy (50). The PKR-mediated induction of autophagy requires the phosphorylation of the translation initiation factor eIF2α (50). Herpes simplex virus type 1 (HSV-1) inhibits autophagy through at least two domains and the activities of the late protein ICP34.5, its C-terminal mediation of dephosphorylation of eIF2α, and its N-terminal binding to Beclin 1 (5, 16, 35). HSV-1 strains lacking ICP34.5 show significant attenuation in vivo, increased sensitivity to IFN (33), an inability to counteract the PKR-induced phosphorylation of eIF2α, and the induction of the generalized shutoff of protein synthesis in infected cells. These activities originally were ascribed solely to ICP34.5''s ability to recruit PP1α and redirect its activity to dephosphorylate eIF2α to counteract the general shutoff of protein synthesis mediated by PKR (17). This function is mediated by the C-terminal domain of ICP34.5 that contains homology to the growth arrest and DNA damage 34 (GADD34) gene (6). Independently of the role of ICP34.5 in countering the PKR-induced antiviral state, viruses lacking ICP34.5 also exhibit altered patterns of autophagy. This manifests as increased long-lived protein degradation, the increased formation of autophagosomes, increased autophagic vacuole volume density, and the enhanced xenophagic degradation of virions (35, 50, 51). Such alterations in the autophagy pathway now can be ascribed to ICP34.5''s ability to bind Beclin 1 in addition to its mediation of eIF2α dephosphorylation (35). The deletion of the Beclin 1-binding domain (BBD) of ICP34.5 renders HSV-1 less able to regulate autophagosome formation, and viruses lacking this domain are neuroattenuated (35).To determine the impact of Beclin 1-ICP34.5 interactions on the pathogenesis of HSV-1 during infection, we examined the ability of BBD-deficient virus to replicate, cause disease, and stimulate an immune response in mice. We determined that mice infected with HSV-1 lacking the ICP34.5 BBD were able to clear virus more efficiently than mice infected with wild-type virus. We observed the significantly enhanced stimulation of CD4+ T cells by the virus lacking the BBD compared to that of mice infected with wild-type virus. These data suggest that Beclin 1 binding and the inhibition of autophagy by ICP34.5 are important for HSV-1 pathogenesis through its ability to suppress autophagy and to dampen the activation of CD4+ T cells.  相似文献   

15.
16.
17.
Infection of cells by herpes simplex virus type 1 (HSV-1) triggers host cell shutoff whereby mRNAs are degraded and cellular protein synthesis is diminished. However, virus protein translation continues because the translational apparatus in HSV-infected cells is maintained in an active state. Surprisingly, poly(A)-binding protein 1 (PABP1), a predominantly cytoplasmic protein that is required for efficient translation initiation, is partially relocated to the nucleus during HSV-1 infection. This relocalization occurred in a time-dependent manner with respect to virus infection. Since HSV-1 infection causes cell stress, we examined other cell stress inducers and found that oxidative stress similarly relocated PABP1. An examination of stress-induced kinases revealed similarities in HSV-1 infection and oxidative stress activation of JNK and p38 mitogen-activated protein (MAP) kinases. Importantly, PABP relocalization in infection was found to be independent of the viral protein ICP27. The depletion of PABP1 by small interfering RNA (siRNA) knockdown had no significant effect on viral replication or the expression of selected virus late proteins, suggesting that reduced levels of cytoplasmic PABP1 are tolerated during infection.The lytic replication cycle of herpes simplex virus type 1 (HSV-1) can be divided into three phases, immediate-early (IE), early (E), and late (L), that occur in a coordinated sequential gene expression program. IE proteins can regulate E and L gene expression, which produces proteins involved in DNA replication, capsid production, and virion assembly. HSV infection results in host cell shutoff to facilitate the efficient production of viral proteins. First, mRNA is degraded by the virion-associated vhs protein, and then ICP27, a multifunctional regulator of gene expression, inhibits pre-mRNA splicing. As most viral mRNAs are intronless, this abrogates the production of stable cellular mRNAs that can be exported to the cytoplasm and compete for translation with viral mRNAs (44).HSV mRNAs are capped and polyadenylated and so are translated via a normal cap-dependent mechanism. Translation initiation, during which translationally active ribosomes are assembled, is a tightly regulated process (21). Eukaryotic initiation factor 4F (eIF4F) (composed of eIF4E, eIF4G, and eIF4A) that binds the cap at the 5′ end of the mRNA promotes the recruitment of the 40S ribosomal subunit and associated factors, including eIF2-GTP initiator tRNA. The recognition of the start codon then promotes large ribosomal subunit joining. Poly(A)-binding protein 1 (PABP1), which binds and multimerizes on mRNA poly(A) tails, enhances translation initiation through interactions with the eIF4G component of the eIF4F cap-binding complex (20, 29, 32, 51) to circularize the mRNA in a “closed-loop” conformation (24). Key protein-RNA and protein-protein interactions in the translation initiation complex are strengthened by this PABP1-mediated circularization (12).HSV-1 maintains active viral translation in the face of host translational shutoff. Infection activates protein kinase R (PKR), which phosphorylates eIF2α, resulting in translation inhibition. However, HSV-1 ICP34.5 redirects protein phosphatase 1α to reverse eIF2α phosphorylation, abrogating the block to translation (17, 38). In addition, the HSV-1 US11 protein inhibits PKR and may also block PKR-mediated eIF2α phosphorylation (40, 42). HSV-1 infection also enhances eIF4F assembly in quiescent cells by the phosphorylation and proteasome-mediated degradation of the eIF4E-binding protein (4E-BP), which, when hypophosphorylated, can negatively regulate eIF4F complex formation (54). However, ICP6 may also contribute to eIF4F assembly by binding to eIF4G (55). Finally, ICP6 is required for Mnk-1 phosphorylation of eIF4E, but the mechanisms behind this remain unclear (54). ICP27 has also been implicated in translation regulation during HSV infection (6, 8, 10, 30) and may also activate p38 mitogen-activated protein (MAP) kinase that can phosphorylate eIF4E (16, 59).PABP1 appears to be a common cellular target of RNA and DNA viruses. PABP1 can undergo proteolysis, intracellular relocalization, or modification of its interaction with other translation factors in response to infection. For example, poliovirus induces host cell shutoff by cleaving PABP1, thus disrupting certain PABP1-containing complexes (28, 29). The rotavirus NSP3 protein can displace PABP1 from translation initiation complexes (41). However, NSP3 also interacts with a cellular protein, RoXaN, which is required to relocate PABP1 to the nucleus (13). Similarly, the Kaposi''s sarcoma herpesvirus (KSHV) SOX protein plays a role in relocating PABP1, its cofactor in cellular mRNA decay, to the nucleus (33). Although steady-state levels of PABP1 are highest in the cytoplasm of normal cells, where it has cytoplasmic functions, it is a nucleocytoplasmic shuttling protein (1). However, it is unclear how PABP1 enters or exits the nucleus, as it contains neither a canonical nuclear export nor an import signal.Here we describe the loss of PABP1 from cap-binding complexes and the partial relocation of PABP1 to the nucleus in HSV-1-infected cells in a time-dependent manner. Relocation is specific for PABP1, as other translation factors remained in the cytoplasm. Cells undergo stress during HSV-1 infection, and analysis of a variety of cell stresses revealed that PABP relocalization was also observed upon oxidative stress. Paxillin, a potential PABP1 nuclear chaperone, was phosphorylated, and the paxillin-PABP1 interaction was reduced during virus infection. However, the interaction was weak and cell type dependent, indicating that other effectors of PABP1 relocation in the infected cell must exist. Recently, the HSV-1 ICP27 protein was suggested to alter the PABP1 cellular location (6). However, infections with ICP27-null mutant viruses clearly demonstrated that ICP27 is not required for PABP1 nuclear relocation in the context of infection. Although HSV-1 mRNAs are translated by a normal cap-dependent mechanism known to be enhanced by PABP1, small interfering RNA (siRNA) knockdown of PABP1 indicated that at late times of infection, the translation of certain virus late proteins tolerates very low levels of PABP1.  相似文献   

18.
19.
Several independent lines of evidence indicate that interferon-mediated innate responses are involved in controlling herpes simplex virus type 1 (HSV-1) infection and that the viral immediate-early regulatory protein ICP0 augments HSV-1 replication in interferon-treated cells. However, this is a complex situation in which the experimental outcome is determined by the choice of multiplicity of infection and cell type and by whether cultured cells or animal models are used. It is now known that neither STAT1 nor interferon regulatory factor 3 (IRF-3) play essential roles in the replication defect of ICP0-null mutant HSV-1 in cultured cells. This study set out to investigate the specific role of ICP0 in HSV-1 resistance to the interferon defense. We have used a cell line in which ICP0 expression can be induced at levels similar to those during the early stages of a normal infection to determine whether ICP0 by itself can interfere with interferon or IRF-3-dependent signaling and whether ICP0 enables the virus to circumvent the effects of interferon-stimulated genes (ISGs). We found that the presence of ICP0 was unable to compromise ISG induction by either interferon or double-stranded RNA. On the other hand, ICP0 preexpression reduced but did not eliminate the inhibitory effects of ISGs on HSV-1 infection, with the extent of the relief being highly dependent on multiplicity of infection. The results are discussed in terms of the relationships between ICP0 and intrinsic and innate antiviral resistance mechanisms.The innate immune response mediated through the interferon (IFN) pathway is an important component of antiviral defense mediated by individual cells and whole organisms (10, 28). In turn, many viruses express proteins that counteract the effects of the IFN response (28). In the case of herpes simplex virus type 1 (HSV-1), highly defective HSV-1 mutants activate expression of IFN-stimulated genes (ISGs) through a mechanism that is independent of IFN itself but dependent on IFN regulatory factor 3 (IRF-3) (2, 3, 19, 23, 26). HSV-1 mutants that do not express the immediate-early (IE) regulatory protein ICP0 are more sensitive than the wild-type (wt) virus to IFN pretreatment of cultured cells (13, 20), and ICP0-null mutant HSV-1 is much more pathogenic in mice unable to respond to IFN (12, 15). Furthermore, a number of experimental systems have presented evidence suggesting that a specific function of ICP0 is to interfere with IFN and/or IRF-3-dependent IFN responses (3, 16-18, 21). However, we have reported recently that the replication defect of ICP0-null mutant HSV-1 is not complemented in cultured cells lacking either STAT1 or IRF-3 (9), which raises the question of whether the relative sensitivity of ICP0-null mutant HSV-1 to an IFN-induced antiviral state results from the absence of a specific effect of ICP0 on IFN pathways or is, rather, an indirect consequence of the disabled virus being intrinsically less able to replicate in cells expressing ISGs (9).The investigation of these complex issues is difficult because sensitivity to IFN is highly dependent on multiplicity of infection (MOI) (9) and cell type (20). Therefore, we sought to develop a system in which the specific effects of ICP0 could be examined in the absence of HSV-1 infection and which avoids potential complications arising from the use of viral vectors or plasmid transfection technologies. In an accompanying paper, we describe the construction of a cell line that expresses ICP0 at physiological levels in an inducible manner (7). The cells allow 100% complementation of plaque formation by ICP0-null mutant HSV-1, and induction of ICP0 expression induces efficient reactivation of gene expression from quiescent HSV-1 genomes (7). We have used these cells to investigate whether, by itself, ICP0 is able to impede induction of ISGs in response to IFN (through the normal STAT1 signaling pathway) or to interfere with IRF-3-dependent activation of ISGs induced by double-stranded RNA, the archetypal pathogen-associated molecular pattern (PAMP). We found that preexpression of ICP0 had no deleterious effect on either pathway. On the other hand, preexpression of ICP0 decreased (but did not eliminate) the sensitivity of HSV-1 to an IFN-induced antiviral state. We discuss the relationship between ICP0 and intrinsic and innate cellular defenses to HSV-1 infection.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号