Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Virol ; 95(17): e0087321, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34133898

ABSTRACT

Nuclear envelope budding in herpesvirus nuclear egress may be negatively regulated, since the pUL31/pUL34 nuclear egress complex heterodimer can induce membrane budding without capsids when expressed ectopically or on artificial membranes in vitro, but not in the infected cell. We have previously described a pUL34 mutant that contained alanine substitutions at R158 and R161 and that showed impaired growth, impaired pUL31/pUL34 interaction, and unregulated budding. Here, we determine the phenotypic contributions of the individual substitutions to these phenotypes. Neither substitution alone was able to reproduce the impaired growth or nuclear egress complex (NEC) interaction phenotypes. Either substitution, however, could fully reproduce the unregulated budding phenotype, suggesting that misregulated budding may not substantially impair virus replication. In addition, the R158A substitution caused relocalization of the NEC to intranuclear punctate structures and recruited lamin A/C to these structures, suggesting that this residue might be important for recruitment of kinases for dispersal of nuclear lamins. IMPORTANCE Herpesvirus nuclear egress is a complex, regulated process coordinated by two virus proteins that are conserved among the herpesviruses that form a heterodimeric nuclear egress complex (NEC). The NEC drives budding of capsids at the inner nuclear membrane and recruits other viral and host cell proteins for disruption of the nuclear lamina, membrane scission, and fusion. The structural basis of individual activities of the NEC, apart from membrane budding, are not clear, nor is the basis of the regulation of membrane budding. Here, we explore the properties of NEC mutants that have an unregulated budding phenotype, determine the significance of that regulation for virus replication, and also characterize a structural requirement for nuclear lamina disruption.


Subject(s)
Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Mutation , Nuclear Lamina/metabolism , Viral Proteins/metabolism , Virus Replication , Active Transport, Cell Nucleus , Animals , Chlorocebus aethiops , Herpes Simplex/genetics , Herpes Simplex/metabolism , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/pathology , Nuclear Envelope/virology , Nuclear Lamina/pathology , Nuclear Lamina/virology , Vero Cells , Viral Proteins/genetics , Virus Release
2.
Viruses ; 10(1)2018 01 13.
Article in English | MEDLINE | ID: mdl-29342872

ABSTRACT

The nuclear phase of herpesvirus replication is regulated through the formation of regulatory multi-component protein complexes. Viral genomic replication is followed by nuclear capsid assembly, DNA encapsidation and nuclear egress. The latter has been studied intensely pointing to the formation of a viral core nuclear egress complex (NEC) that recruits a multimeric assembly of viral and cellular factors for the reorganization of the nuclear envelope. To date, the mechanism of the association of human cytomegalovirus (HCMV) capsids with the NEC, which in turn initiates the specific steps of nuclear capsid budding, remains undefined. Here, we provide electron microscopy-based data demonstrating the association of both nuclear capsids and NEC proteins at nuclear lamina budding sites. Specifically, immunogold labelling of the core NEC constituent pUL53 and NEC-associated viral kinase pUL97 suggested an intranuclear NEC-capsid interaction. Staining patterns with phospho-specific lamin A/C antibodies are compatible with earlier postulates of targeted capsid egress at lamina-depleted areas. Important data were provided by co-immunoprecipitation and in vitro kinase analyses using lysates from HCMV-infected cells, nuclear fractions, or infectious virions. Data strongly suggest that nuclear capsids interact with pUL53 and pUL97. Combined, the findings support a refined concept of HCMV nuclear trafficking and NEC-capsid interaction.


Subject(s)
Capsid/physiology , Cytomegalovirus/enzymology , Cytomegalovirus/physiology , Nuclear Envelope/virology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinases/metabolism , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Cytomegalovirus/ultrastructure , Host-Pathogen Interactions , Humans , Immunohistochemistry , Microscopy, Electron , Nuclear Envelope/ultrastructure , Nuclear Lamina/ultrastructure , Nuclear Lamina/virology , Phosphorylation , Viral Proteins/metabolism , Virus Assembly , Virus Release , Virus Replication
3.
Sci Rep ; 7(1): 7823, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798307

ABSTRACT

Baculovirus nucleocapsids egress from the nucleus primarily via budding at the nuclear membrane. The nuclear lamina underlying the nuclear membrane represents a substantial barrier to nuclear egress. Whether the nuclear lamina undergoes disruption during baculovirus infection remains unknown. In this report, we generated a clonal cell line, Sf9-L, that stably expresses GFP-tagged Drosophila lamin B. GFP autofluorescence colocalized with immunofluorescent anti-lamin B at the nuclear rim of Sf9-L cells, indicating GFP-lamin B was incorporated into the nuclear lamina. Meanwhile, virus was able to replicate normally in Sf9-L cells. Next, we investigated alterations to the nuclear lamina during baculovirus infection in Sf9-L cells. A portion of GFP-lamin B localized diffusely at the nuclear rim, and some GFP-lamin B was redistributed within the nucleus during the late phase of infection, suggesting the nuclear lamina was partially disrupted. Immunoelectron microscopy revealed associations between GFP-lamin B and the edges of the electron-dense stromal mattes of the virogenic stroma, intranuclear microvesicles, and ODV envelopes and nucleocapsids within the nucleus, indicating the release of some GFP-lamin B from the nuclear lamina. Additionally, GFP-lamin B phosphorylation increased upon infection. Based on these data, baculovirus infection induced lamin B phosphorylation and disruption of the nuclear lamina.


Subject(s)
Baculoviridae/physiology , Clone Cells/virology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Lamins/genetics , Lamins/metabolism , Nuclear Lamina/physiology , Animals , Clone Cells/metabolism , Drosophila/genetics , Drosophila/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Immunoelectron , Nuclear Lamina/virology , Phosphorylation , Recombinant Proteins/metabolism , Sf9 Cells , Virus Release , Virus Replication
4.
Curr Opin Cell Biol ; 46: 9-16, 2017 06.
Article in English | MEDLINE | ID: mdl-28086162

ABSTRACT

Herpesviruses, like most DNA viruses, replicate and package their genomes into capsids in the host cell nucleus. Capsids then transit to the cytoplasm in a fascinating process called nuclear egress, which includes several unusual steps: Movement of capsids from the nuclear interior to the periphery, disruption of the nuclear lamina, capsid budding through the inner nuclear membrane, and fusion of enveloped particles with the outer nuclear membrane. Here, we review recent advances and emerging questions relating to herpesvirus nuclear egress, emphasizing controversies regarding mechanisms for capsid trafficking to the nuclear periphery, and implications of recent structures of the two-subunit, viral nuclear egress complex for the process, particularly at the step of budding through the inner nuclear membrane.


Subject(s)
Biological Transport , Cell Nucleus/virology , Herpesviridae/physiology , Nuclear Envelope/virology , Animals , Capsid/chemistry , Capsid/physiology , Cell Nucleus/chemistry , Cytoplasm/virology , Humans , Models, Molecular , Nuclear Envelope/chemistry , Nuclear Lamina/virology
5.
J Virol ; 90(22): 10414-10422, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27630226

ABSTRACT

Herpes simplex virus 1 (HSV-1) remodels nuclear membranes during virus egress. Although the UL31 and UL34 proteins control nucleocapsid transit in infected cells, the molecular interactions required for their function are unclear. Here we report that the γ134.5 gene product of HSV-1 facilitates nucleocapsid release to the cytoplasm through bridging the UL31/UL34 complex, cellular p32, and protein kinase C. Unlike wild-type virus, an HSV mutant devoid of γ134.5 or its amino terminus is crippled for viral growth and release. This is attributable to a defect in virus nuclear egress. In infected cells, wild-type virus recruits protein kinase C to the nuclear membrane and triggers its activation, whereas the γ134.5 mutants fail to exert such an effect. Accordingly, the γ134.5 mutants are unable to induce phosphorylation and reorganization of lamin A/C. When expressed in host cells γ134.5 targets p32 and protein kinase C. Meanwhile, it communicates with the UL31/UL34 complex through UL31. Deletion of the amino terminus from γ134.5 disrupts its activity. These results suggest that disintegration of the nuclear lamina mediated by γ134.5 promotes HSV replication. IMPORTANCE: HSV nuclear egress is a key step that determines the outcome of viral infection. While the nuclear egress complex mediates capsid transit across the nuclear membrane, the regulatory components are not clearly defined in virus-infected cells. We report that the γ134.5 gene product, a virulence factor of HSV-1, facilitates nuclear egress cooperatively with cellular p32, protein kinase C, and the nuclear egress complex. This work highlights a viral mechanism that may contribute to the pathogenesis of HSV infection.


Subject(s)
Herpesvirus 1, Human/metabolism , Lamin Type A/metabolism , Phosphorylation/physiology , Viral Proteins/metabolism , Virus Release/physiology , Animals , Capsid/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/virology , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoplasm/virology , HeLa Cells , Humans , Nuclear Envelope/metabolism , Nuclear Envelope/virology , Nuclear Lamina/metabolism , Nuclear Lamina/virology , Nuclear Proteins/metabolism , Nucleocapsid/metabolism , Protein Kinase C/metabolism , Vero Cells , Virus Assembly/physiology
6.
J Virol ; 90(23): 10738-10751, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27654296

ABSTRACT

Nuclear egress of herpesviruses is accompanied by changes in the architecture of the nuclear membrane and nuclear lamina that are thought to facilitate capsid access to the inner nuclear membrane (INM) and curvature of patches of the INM around the capsid during budding. Here we report the properties of a point mutant of pUL34 (Q163A) that fails to induce gross changes in nuclear architecture or redistribution of lamin A/C. The UL34(Q163A) mutant shows a roughly 100-fold defect in single-step growth, and it forms small plaques. This mutant has a defect in nuclear egress, and furthermore, it fails to disrupt nuclear shape or cause observable displacement of lamin A/C despite retaining the ability to recruit the pUS3 and PKC protein kinases and to mediate phosphorylation of emerin. Extragenic suppressors of the UL34(Q163A) phenotype were isolated, and all of them carry a single mutation of arginine 229 to leucine in UL31. Surprisingly, although this UL31 mutation largely restores virus replication, it does not correct the lamina disruption defect, suggesting that, in Vero cells, changes in nuclear shape and gross displacements of lamin A/C may facilitate but are unnecessary for nuclear egress. IMPORTANCE: Herpesvirus nuclear egress is an essential and conserved process that requires close association of the viral capsid with the inner nuclear membrane and budding of the capsid into that membrane. Access to the nuclear membrane and tight curvature of that membrane are thought to require disruption of the nuclear lamina that underlies the inner nuclear membrane, and consistent with this idea, herpesvirus infection induces biochemical and architectural changes at the nuclear membrane. The significance of the nuclear membrane architectural changes is poorly characterized. The results presented here address that deficiency in our understanding and show that a combination of mutations in two of the viral nuclear egress factors results in a failure to accomplish at least two components of lamina disruption while still allowing relatively efficient viral replication, suggesting that changes in nuclear shape and displacement of lamins are not necessary for herpes simplex virus 1 (HSV-1) nuclear egress.


Subject(s)
Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Point Mutation , Viral Proteins/genetics , Viral Proteins/physiology , Virus Release/genetics , Virus Release/physiology , Amino Acid Substitution , Animals , Cell Nucleus Shape , Chlorocebus aethiops , HEK293 Cells , Herpesvirus 1, Human/pathogenicity , Humans , Lamin Type A/metabolism , Models, Molecular , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Envelope/virology , Nuclear Lamina/metabolism , Nuclear Lamina/ultrastructure , Nuclear Lamina/virology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Protein Conformation , Vero Cells , Viral Proteins/chemistry , Virus Replication
7.
PLoS Pathog ; 12(8): e1005825, 2016 08.
Article in English | MEDLINE | ID: mdl-27556400

ABSTRACT

The nuclear lamina lines the inner nuclear membrane providing a structural framework for the nucleus. Cellular processes, such as nuclear envelope breakdown during mitosis or nuclear export of large ribonucleoprotein complexes, are functionally linked to the disassembly of the nuclear lamina. In general, lamina disassembly is mediated by phosphorylation, but the precise molecular mechanism is still not completely understood. Recently, we suggested a novel mechanism for lamina disassembly during the nuclear egress of herpesviral capsids which involves the cellular isomerase Pin1. In this study, we focused on mechanistic details of herpesviral nuclear replication to demonstrate the general importance of Pin1 for lamina disassembly. In particular, Ser22-specific lamin phosphorylation consistently generates a Pin1-binding motif in cells infected with human and animal alpha-, beta-, and gammaherpesviruses. Using nuclear magnetic resonance spectroscopy, we showed that binding of Pin1 to a synthetic lamin peptide induces its cis/trans isomerization in vitro. A detailed bioinformatic evaluation strongly suggests that this structural conversion induces large-scale secondary structural changes in the lamin N-terminus. Thus, we concluded that a Pin1-induced conformational change of lamins may represent the molecular trigger responsible for lamina disassembly. Consistent with this concept, pharmacological inhibition of Pin1 activity blocked lamina disassembly in herpesvirus-infected fibroblasts and consequently impaired virus replication. In addition, a phospho-mimetic Ser22Glu lamin mutant was still able to form a regular lamina structure and overexpression of a Ser22-phosphorylating kinase did not induce lamina disassembly in Pin1 knockout cells. Intriguingly, this was observed in absence of herpesvirus infection proposing a broader importance of Pin1 for lamina constitution. Thus, our results suggest a functional model of similar events leading to disassembly of the nuclear lamina in response to herpesviral or inherent cellular stimuli. In essence, Pin1 represents a regulatory effector of lamina disassembly that promotes the nuclear pore-independent egress of herpesviral capsids.


Subject(s)
Herpesviridae Infections/virology , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Nuclear Lamina/virology , Virus Replication/physiology , Blotting, Western , Capsid/metabolism , Capsid/virology , Cells, Cultured , Fluorescent Antibody Technique , Herpesviridae , Herpesviridae Infections/metabolism , Humans , Lamins , Magnetic Resonance Spectroscopy , Nuclear Lamina/metabolism , Phosphorylation
8.
J Virol ; 88(18): 10982-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24965476

ABSTRACT

Human cytomegalovirus (HCMV) kinase UL97 is required for efficient nuclear lamina disruption during nuclear egress. However, cellular protein kinase C (PKC) has been implicated in this process in other systems. Comparing the effects of UL97 and cellular kinase inhibitors on HCMV nuclear egress confirms a role for UL97 in lamina disruption and nuclear egress. A pan-PKC inhibitor did not affect lamina disruption but did reduce the number of cytoplasmic capsids more than the number of nuclear capsids.


Subject(s)
Cell Nucleus/virology , Cytomegalovirus Infections/enzymology , Cytomegalovirus/enzymology , Nuclear Lamina/virology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Virus Release/drug effects , Capsid/metabolism , Cell Line , Cell Nucleus/drug effects , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Humans , Nuclear Lamina/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/metabolism , Virus Assembly/drug effects
9.
J Virol ; 87(9): 5019-27, 2013 May.
Article in English | MEDLINE | ID: mdl-23427156

ABSTRACT

Human cytomegalovirus (HCMV) encodes one conventional protein kinase, UL97. During infection, UL97 phosphorylates the retinoblastoma tumor suppressor protein (pRb) on sites ordinarily phosphorylated by cyclin-dependent kinases (CDK), inactivating the ability of pRb to repress host genes required for cell cycle progression to S phase. UL97 is important for viral DNA synthesis in quiescent cells, but this function can be replaced by human papillomavirus type 16 E7, which targets pRb for degradation. However, viruses in which E7 replaces UL97 are still defective for virus production. UL97 is also required for efficient nuclear egress of viral nucleocapsids, which is associated with disruption of the nuclear lamina during infection, and phosphorylation of lamin A/C on serine 22, which antagonizes lamin polymerization. We investigated whether inactivation of pRb might overcome the requirement of UL97 for these roles, as pRb inactivation induces CDK1, and CDK1 phosphorylates lamin A/C on serine 22. We found that lamin A/C serine 22 phosphorylation during HCMV infection correlated with expression of UL97 and was considerably delayed in UL97-null mutants, even when E7 was expressed. E7 failed to restore gaps in the nuclear lamina seen in wild-type but not UL97-null virus infections. In electron microscopy analyses, a UL97-null virus expressing E7 was as impaired as a UL97-null mutant in cytoplasmic accumulation of viral nucleocapsids. Our results demonstrate that pRb inactivation is insufficient to restore efficient viral nuclear egress of HCMV in the absence of UL97 and instead argue further for a direct role of UL97 in this stage of the infectious cycle.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/enzymology , Nuclear Lamina/virology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Retinoblastoma Protein/metabolism , Virus Release , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cell Nucleus/virology , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Humans , Lamin Type A/chemistry , Lamin Type A/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polymerization , Retinoblastoma Protein/genetics
10.
Adv Virus Res ; 80: 69-101, 2011.
Article in English | MEDLINE | ID: mdl-21762822

ABSTRACT

Viral replication is a complex process relying on a network of interacting viral and cellular proteins, in which particularly protein kinases play an important regulatory role. The specific phosphorylation of substrate proteins induces activation, inactivation, or other functional modification and thus determines virus-host cell interregulation. During herpesviral infections, both viral and cellular protein kinases are expressed and provide activities crucial for the efficiency of virus replication. The protein kinase pUL97 encoded by human cytomegalovirus (HCMV) is a multifunctional regulatory enzyme which exerts strong regulatory effects on early and late steps of the viral replication cycle. A number of interacting proteins and substrates of pUL97 have been described, including retinoblastoma (Rb) protein, nuclear lamins and viral pUL69. Recently, it was demonstrated that pUL97 has structural and functional resemblance to cyclin-dependent protein kinases (CDKs) and thus represents a CDK ortholog. pUL97 can phosphorylate and inactivate Rb, resulting in a stimulation of cell cycle progression. In addition, the association of pUL97 activity with nucleocytoplasmic export of viral capsids has been demonstrated by several investigators. We could show that pUL97 is able to phosphorylate nuclear lamins and to contribute to the HCMV-induced reorganization of the nuclear lamina. On the basis of very recent findings, it is becoming increasingly clear that pUL97 is a component of a multiprotein nuclear egress complex (NEC). The NEC contains a small number of egress proteins involved in the recruitment of protein kinases, such as pUL97 and cellular protein kinase C (PKC), to specific sites of the nuclear lamina. Current information about the composition, function, and regulatory complexity of the NEC leads to a mechanistic concept which may set the key features of HCMV nuclear egress in a new light.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Cytomegalovirus/genetics , Gene Expression Regulation, Viral , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Cell Line , Cyclin-Dependent Kinases/genetics , Cytomegalovirus/enzymology , Cytomegalovirus/physiology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Humans , Nuclear Lamina/genetics , Nuclear Lamina/metabolism , Nuclear Lamina/virology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Kinase C/genetics , Protein Kinase C/metabolism , Retinoblastoma/genetics , Retinoblastoma/metabolism , Virus Replication
11.
J Virol ; 85(16): 8263-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21680527

ABSTRACT

African swine fever virus (ASFV), the causative agent of one of the most devastating swine diseases, has been considered exclusively cytoplasmic, even though some authors have shown evidence of an early stage of nuclear replication. In the present study, an increment of lamin A/C phosphorylation was observed in ASFV-infected cells as early as 4 h postinfection, followed by the disassembling of the lamina network close to the sites where the viral genome starts its replication. At later time points, this and other nuclear envelope markers were found in the cytoplasm of the infected cells. The effect of the infection on the cell nucleus was much more severe than previously expected, since a redistribution of other nuclear proteins, such as RNA polymerase II, the splicing speckle SC-35 marker, and the B-23 nucleolar marker, was observed from 4 h postinfection. All this evidence, together with the redistribution, dephosphorylation, and subsequent degradation of RNA polymerase II after ASFV infection, suggests the existence of sophisticated mechanisms to regulate the nuclear machinery during viral infection.


Subject(s)
African Swine Fever Virus/physiology , Cell Nucleus/metabolism , Lamin Type A/metabolism , African Swine Fever Virus/genetics , African Swine Fever Virus/metabolism , Animals , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Chlorocebus aethiops , Cytoplasm/virology , Fluorescent Antibody Technique , In Situ Hybridization , Microscopy, Confocal , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Envelope/virology , Nuclear Lamina/metabolism , Nuclear Lamina/ultrastructure , Nuclear Lamina/virology , Nuclear Proteins/metabolism , Phosphorylation , RNA Polymerase II/metabolism , Vero Cells , Virus Replication
12.
Virology ; 406(1): 127-37, 2010 Oct 10.
Article in English | MEDLINE | ID: mdl-20674954

ABSTRACT

The nuclear lamina is thought to be a steric barrier to the herpesvirus capsid. Disruption of the lamina accompanied by phosphorylation of lamina proteins is a conserved feature of herpesvirus infection. In HSV-1-infected cells, protein kinase C (PKC) alpha and delta isoforms are recruited to the nuclear membrane and PKC delta has been implicated in phosphorylation of emerin and lamin B. We tested two critical hypotheses about the mechanism and significance of lamina disruption. First, we show that chemical inhibition of all PKC isoforms reduced viral growth five-fold and inhibited capsid egress from the nucleus. However, specific inhibition of either conventional PKCs or PKC delta does not inhibit viral growth. Second, we show hyperphosphorylation of emerin by viral and cellular kinases is required for its disassociation from the lamina. These data support hypothesis that phosphorylation of lamina components mediates lamina disruption during HSV nuclear egress.


Subject(s)
Herpesvirus 1, Human/physiology , Host-Pathogen Interactions/physiology , Nuclear Lamina/virology , Protein Kinase C/physiology , Virus Release/physiology , Animals , Base Sequence , Capsid/drug effects , Capsid/physiology , Capsid/ultrastructure , Cell Line , Chlorocebus aethiops , DNA Primers/genetics , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/ultrastructure , Host-Pathogen Interactions/drug effects , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/physiology , Microscopy, Electron, Transmission , Models, Biological , Nuclear Lamina/drug effects , Nuclear Lamina/enzymology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Vero Cells , Virus Assembly/drug effects , Virus Assembly/physiology , Virus Release/drug effects , Virus Replication/drug effects , Virus Replication/physiology
13.
J Biol Chem ; 285(18): 13979-89, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20202933

ABSTRACT

The nucleocytoplasmic egress of viral capsids is a rate-limiting step in the replication of the human cytomegalovirus (HCMV). As reported recently, an HCMV-specific nuclear egress complex is composed of viral and cellular proteins, in particular protein kinases with the capacity to induce destabilization of the nuclear lamina. Viral protein kinase pUL97 and cellular protein kinase C (PKC) play important roles by phosphorylating several types of nuclear lamins. Using pUL97 mutants, we show that the lamin-phosphorylating activity of pUL97 is associated with a reorganization of nuclear lamin A/C. Either pUL97 or PKC has the potential to induce distinct punctate lamina-depleted areas at the periphery of the nuclear envelope, which were detectable in transiently transfected and HCMV-infected cells. Using recombinant HCMV, which produces green fluorescent protein-labeled viral capsids, the direct transition of viral capsids through these areas could be visualized. This process was sensitive to an inhibitor of pUL97/PKC activity. The pUL97-mediated phosphorylation of lamin A/C at Ser(22) generated a novel binding motif for the peptidyl-prolyl cis/trans-isomerase Pin1. In HCMV-infected fibroblasts, the physiological localization of Pin1 was altered, leading to recruitment of Pin1 to viral replication centers and to the nuclear lamina. The local increase in Pin1 peptidyl-prolyl cis/trans-isomerase activity may promote conformational modulation of lamins. Thus, we postulate a novel phosphorylation-triggered mechanism for the reorganization of the nuclear lamina in HCMV-infected cells.


Subject(s)
Capsid/metabolism , Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Nuclear Lamina/metabolism , Viral Proteins/metabolism , Virus Release/physiology , Amino Acid Motifs , Cytomegalovirus Infections/genetics , HeLa Cells , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Nuclear Lamina/genetics , Nuclear Lamina/virology , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Phosphorylation/physiology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Viral Proteins/genetics
14.
Virology ; 392(1): 137-47, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19640559

ABSTRACT

Herpesviruses must cross the inner nuclear membrane and underlying lamina to exit the nucleus. HSV-1 US3 and PKC can phosphorylate lamins and induce their dispersion but do not elicit all of the phosphorylated lamin species produced during infection. UL13 is a serine threonine protein kinase conserved among many herpesviruses. HSV-1 UL13 phosphorylates US3 and thereby controls UL31 and UL34 nuclear rim localization, indicating a role in nuclear egress. Here, we report that HSV-2 UL13 alone induced conformational changes in lamins A and C and redistributed lamin B1 from the nuclear rim to intranuclear granular structures. HSV-2 UL13 directly phosphorylated lamins A, C, and B1 in vitro, and the lamin A1 tail domain. HSV-2 infection recapitulated the lamin alterations seen upon expression of UL13 alone, and other alterations were also observed, indicating that additional viral and/or cellular proteins cooperate with UL13 to alter lamins during HSV-2 infection to allow nuclear egress.


Subject(s)
Herpesvirus 2, Human/enzymology , Herpesvirus 2, Human/pathogenicity , Lamins/metabolism , Nuclear Lamina/metabolism , Nuclear Lamina/virology , Protein Kinases/physiology , Active Transport, Cell Nucleus , Base Sequence , Binding Sites , DNA Primers/genetics , DNA, Viral/genetics , HeLa Cells , Herpesvirus 2, Human/genetics , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Lamin Type A/metabolism , Lamin Type B/chemistry , Lamin Type B/genetics , Lamin Type B/metabolism , Lamins/chemistry , Lamins/genetics , Phosphorylation , Protein Conformation , Protein Kinases/genetics , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
15.
J Gen Virol ; 89(Pt 3): 731-740, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18272765

ABSTRACT

A fundamental step in the efficient production of human cytomegalovirus (HCMV) progeny is viral egress from the nucleus to the cytoplasm of infected cells. In the family Herpesviridae, this process involves alteration of nuclear lamina components by two highly conserved proteins, whose homologues in HCMV are named pUL50 and pUL53. This study showed that HCMV infection induced the mislocalization of nuclear lamins and that pUL50 and pUL53 play a role in this event. At late stages of infection, both lamin A/C and lamin B showed an irregular distribution on the nuclear rim, coincident with areas of pUL53 accumulation. No variations in the total amount of nuclear lamins could be detected, supporting the view that HCMV induces a qualitative, rather than a quantitative, alteration of these cellular components, as has been suggested previously for other herpesviruses. Interestingly, pUL53, in the absence of other viral products, localized diffusely in the nucleus, whilst the co-expression and interaction of pUL53 with its partner, pUL50, restored its nuclear rim localization in distinct patches, thus indicating that pUL50 is sufficient to induce the localization of pUL53 observed during virus infection. Importantly, analysis of the nuclear lamina in the presence of pUL50-pUL53 complexes at the nuclear boundary and in the absence of other viral products showed that the two viral proteins were sufficient to promote alterations of lamins, strongly resembling those observed during HCMV infection. These results suggest that pUL50 and pUL53 may play an important role in the exit of virions from the nucleus by inducing structural modifications of the nuclear lamina.


Subject(s)
Cytomegalovirus/pathogenicity , Nuclear Lamina/metabolism , Nuclear Lamina/virology , Viral Proteins/metabolism , Animals , COS Cells/virology , Cell Nucleus/metabolism , Cell Nucleus/virology , Chlorocebus aethiops , Cytomegalovirus/metabolism , Fibroblasts/virology , Humans , Immunoprecipitation , Lamins/metabolism , Lung/cytology , Virion/metabolism
16.
Virology ; 347(2): 261-76, 2006 Apr 10.
Article in English | MEDLINE | ID: mdl-16427676

ABSTRACT

Cells infected with wild type HSV-1 showed significant lamin A/C and lamin B rearrangement, while UL34-null virus-infected cells exhibited few changes in lamin localization, indicating that UL34 is necessary for lamin disruption. During HSV infection, US3 limited the development of disruptions in the lamina, since cells infected with a US3-null virus developed large perforations in the lamin layer. US3 regulation of lamin disruption does not correlate with the induction of apoptosis. Expression of either UL34 or US3 proteins alone disrupted lamin A/C and lamin B localization. Expression of UL34 and US3 together had little effect on lamin A/C localization, suggesting a regulatory interaction between the two proteins. The data presented in this paper argue for crucial roles for both UL34 and US3 in regulating the state of the nuclear lamina during viral infection.


Subject(s)
Herpesvirus 1, Human/enzymology , Nuclear Lamina/metabolism , Nuclear Lamina/virology , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Protein Serine-Threonine Kinases/genetics , Vero Cells , Viral Proteins/genetics
17.
J Virol ; 80(1): 494-504, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16352573

ABSTRACT

We report that herpes simplex virus type 1 (HSV-1) infection leads to the recruitment of protein kinase C (PKC) to the nuclear rim. In HEp-2 cells, PKC recruitment to the nuclear rim was initiated between 8 h and 12 h postinfection. PKCdelta, a proapoptotic kinase, was completely recruited to the nuclear rim upon infection with HSV-1. PKCalpha was less dramatically relocalized mostly at the nuclear rim upon infection, although some PKCalpha remained in the cytoplasm. PKCzeta-specific immunofluorescence was not significantly relocated to the nuclear rim. The UL34 and UL31 proteins, as well as their association, were each required for PKC recruitment to the nuclear rim. The HSV-1 US3 protein product, a kinase which regulates the phosphorylation state and localization of UL34, was not required for PKC recruitment to the nuclear rim; however, it was required for proper localization along the nuclear rim, as PKC appeared unevenly distributed along the nuclear rim of cells infected with US3 null and kinase-dead mutants. HSV-1 infection induced the phosphorylation of both lamin B and PKC. Elevated lamin B phosphorylation in HSV-1-infected cells was partially reduced by inhibitors of PKC. The data suggest a model in which kinases that normally disassemble the nuclear lamina during apoptosis are recruited to the nuclear membrane through functions requiring UL31 and UL34. We hypothesize that the recruitment of PKC functions to phosphorylate lamin B to help modify the nuclear lamina and promote budding of nucleocapsids at the inner nuclear membrane.


Subject(s)
Herpesvirus 1, Human/enzymology , Nuclear Envelope/enzymology , Nuclear Lamina/metabolism , Nuclear Lamina/virology , Protein Kinase C/metabolism , Animals , Chlorocebus aethiops , Enzyme Activation , HeLa Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Phosphorylation , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/metabolism , Vero Cells , Viral Proteins/metabolism
18.
Virology ; 320(1): 85-98, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-15003865

ABSTRACT

Nuclear matrix is a structural framework of important nuclear processes. We studied the effect of two different types of viral infections on nuclear matrix. HeLa cells were infected with human rhinovirus 1B (HRV 1B) or measles virus (MV), and Nuclear Mitotic Apparatus protein (NuMA) and lamins A/C and B were used as markers for internal nuclear matrix and peripheral nuclear lamina, respectively. We show that NuMA, lamins, and poly(ADP-ribose) polymerase-1 are cleaved during viral infection in a virus family-specific manner suggesting that these viruses activate different sets of proteases. Morphologically, NuMA was excluded from the condensed chromatin, lamins showed a folded distribution, and both proteins finally remained around the nuclear fragments. A general caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (z-VAD-FMK) prevented the nuclear disintegration and the cleavage of the proteins studied. Interestingly, z-VAD-FMK rescued MV-infected but not HRV 1B-infected cells from cell death. These results show for the first time that NuMA and lamins are specific target proteins during virus-induced programmed cell death.


Subject(s)
Caspase Inhibitors , Lamins/metabolism , Measles virus , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Rhinovirus , Spindle Apparatus/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Antigens, Nuclear , Cell Cycle Proteins , Cell Death/drug effects , Cell Nucleus/metabolism , Cysteine Proteinase Inhibitors/pharmacology , HeLa Cells , Humans , Nuclear Lamina/virology , Nuclear Matrix-Associated Proteins , Poly(ADP-ribose) Polymerases/metabolism , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...