201
|
YB-1 functions as a porter to lead influenza virus ribonucleoprotein complexes to microtubules. J Virol 2012; 86:11086-95. [PMID: 22855482 DOI: 10.1128/jvi.00453-12] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
De novo-synthesized RNAs are under the regulation of multiple posttranscriptional processes by a variety of RNA-binding proteins. The influenza virus genome consists of single-stranded RNAs and exists as viral ribonucleoprotein (vRNP) complexes. After the replication of vRNP in the nucleus, it is exported to the cytoplasm and then reaches the budding site beneath the cell surface in a process mediated by Rab11a-positive recycling endosomes along microtubules. However, the regulatory mechanisms of the postreplicational processes of vRNP are largely unknown. Here we identified, as a novel vRNP-interacting protein, Y-box-binding protein 1 (YB-1), a cellular protein that is involved in regulation of cellular transcription and translation. YB-1 translocated to the nucleus from the cytoplasm and accumulated in PML nuclear bodies in response to influenza virus infection. vRNP assembled into the exporting complexes with YB-1 at PML nuclear bodies. After nuclear export, using YB-1 knockdown cells and in vitro reconstituted systems, YB-1 was shown to be required for the interaction of vRNP exported from the nucleus with microtubules around the microtubule-organizing center (MTOC), where Rab11a-positive recycling endosomes were located. Further, we also found that YB-1 overexpression stimulates the production of progeny virions in an Rab11a-dependent manner. Taking these findings together, we propose that YB-1 is a porter that leads vRNP to microtubules from the nucleus and puts it into the vesicular trafficking system.
Collapse
|
202
|
McBride AA, Sakakibara N, Stepp WH, Jang MK. Hitchhiking on host chromatin: how papillomaviruses persist. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1819:820-5. [PMID: 22306660 PMCID: PMC3357461 DOI: 10.1016/j.bbagrm.2012.01.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 01/07/2012] [Accepted: 01/10/2012] [Indexed: 10/14/2022]
Abstract
Persistent viruses need mechanisms to protect their genomes from cellular defenses and to ensure that they are efficiently propagated to daughter host cells. One mechanism by which papillomaviruses achieve this is through the association of viral genomes with host chromatin, mediated by the viral E2 tethering protein. Association of viral DNA with regions of active host chromatin ensures that the virus remains transcriptionally active and is not relegated to repressed heterochromatin. In addition, viral genomes are tethered to specific regions of host mitotic chromosomes to efficiently partition their DNA to daughter cells. Vegetative viral DNA replication also initiates at specific regions of host chromatin, where the viral E1 and E2 proteins initiate a DNA damage response that recruits cellular DNA damage and repair proteins to viral replication foci for efficient viral DNA synthesis. Thus, these small viruses have capitalized on interactions with chromatin to efficiently target their genomes to beneficial regions of the host nucleus. This article is part of a Special Issue entitled: Chromatin in time and space.
Collapse
Affiliation(s)
| | | | - Wesley H. Stepp
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Moon Kyoo Jang
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, USA
| |
Collapse
|
203
|
Okino Y, Inayoshi Y, Kojima Y, Kidani S, Kaneoka H, Honkawa A, Higuchi H, Nishijima KI, Miyake K, Iijima S. Moloney murine leukemia virus integrase and reverse transcriptase interact with PML proteins. J Biochem 2012; 152:161-9. [PMID: 22685230 DOI: 10.1093/jb/mvs063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Pull-down assay and co-immunoprecipitation of cell extracts in which the integrase or reverse transcriptase of Moloney murine leukemia virus was transiently expressed showed that both enzymes interacted with PML proteins. In infected cells, interaction between the integrase and PML was also observed. Transient expression of PIASy and SUMO proteins facilitated SUMOylation of the integrase but had no apparent effects on the interaction with PML. A FLAG-tagged integrase co-localized with PML protein possibly in the PML body. Knockdown of PML by small interfering RNA resulted in reduced viral cDNA levels and integration efficiency. This suggested that PML proteins activated reverse transcription.
Collapse
Affiliation(s)
- Yuuki Okino
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Chikusa-ku, Nagoya, 464-8603, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
3D reconstruction of VZV infected cell nuclei and PML nuclear cages by serial section array scanning electron microscopy and electron tomography. PLoS Pathog 2012; 8:e1002740. [PMID: 22685402 PMCID: PMC3369938 DOI: 10.1371/journal.ppat.1002740] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 04/25/2012] [Indexed: 12/21/2022] Open
Abstract
Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella (chickenpox) and herpes zoster (shingles). Like all herpesviruses, the VZV DNA genome is replicated in the nucleus and packaged into nucleocapsids that must egress across the nuclear membrane for incorporation into virus particles in the cytoplasm. Our recent work showed that VZV nucleocapsids are sequestered in nuclear cages formed from promyelocytic leukemia protein (PML) in vitro and in human dorsal root ganglia and skin xenografts in vivo. We sought a method to determine the three-dimensional (3D) distribution of nucleocapsids in the nuclei of herpesvirus-infected cells as well as the 3D shape, volume and ultrastructure of these unique PML subnuclear domains. Here we report the development of a novel 3D imaging and reconstruction strategy that we term Serial Section Array-Scanning Electron Microscopy (SSA-SEM) and its application to the analysis of VZV-infected cells and these nuclear PML cages. We show that SSA-SEM permits large volume imaging and 3D reconstruction at a resolution sufficient to localize, count and distinguish different types of VZV nucleocapsids and to visualize complete PML cages. This method allowed a quantitative determination of how many nucleocapsids can be sequestered within individual PML cages (sequestration capacity), what proportion of nucleocapsids are entrapped in single nuclei (sequestration efficiency) and revealed the ultrastructural detail of the PML cages. More than 98% of all nucleocapsids in reconstructed nuclear volumes were contained in PML cages and single PML cages sequestered up to 2,780 nucleocapsids, which were shown by electron tomography to be embedded and cross-linked by an filamentous electron-dense meshwork within these unique subnuclear domains. This SSA-SEM analysis extends our recent characterization of PML cages and provides a proof of concept for this new strategy to investigate events during virion assembly at the single cell level. Varicella-zoster virus (VZV), the cause of varicella and zoster, is a human herpesvirus that replicates in the host cell nucleus where viral genomes are packaged into virion nucleocapsids. We have recently identified antiviral PML (promyelocytic leukemia) nuclear cages that sequester VZV nucleocapsids and inhibit formation of infectious particles. Here we developed a novel three-dimensional (3D) imaging and reconstruction strategy, termed Serial Section Array-Scanning Electron Microscopy (SSA-SEM) that together with electron tomography made it possible to derive 3D reconstructions of complete herpesvirus infected host cell nuclei and of PML cages with ultrastructural precision for the first time. We determined the 3D distribution of several thousand nucleocapsids within reconstructed volumes of single host cell nuclei and in PML cages as well as their sequestration efficiency and sequestration capacity: more than 98% of nucleocapsids were entrapped within PML cages and individual PML cages could sequester nearly 3,000 nucleocapsids which were cross-linked by an irregular electron-dense meshwork within the PML cages. This 3D analysis provides a proof of concept for using SSA-SEM to investigate virion assembly at the whole cell level and further elucidates our observation that PML cages are antiviral nuclear domains which block VZV nucleocapsid egress from the infected cell nucleus.
Collapse
|
205
|
Patsalo V, Yondola MA, Luan B, Shoshani I, Kisker C, Green DF, Raleigh DP, Hearing P. Biophysical and functional analyses suggest that adenovirus E4-ORF3 protein requires higher-order multimerization to function against promyelocytic leukemia protein nuclear bodies. J Biol Chem 2012; 287:22573-83. [PMID: 22573317 DOI: 10.1074/jbc.m112.344234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The early region 4 open reading frame 3 protein (E4-ORF3; UniProt ID P04489) is the most highly conserved of all adenovirus-encoded gene products at the amino acid level. A conserved attribute of the E4-ORF3 proteins of different human adenoviruses is the ability to disrupt PML nuclear bodies from their normally punctate appearance into heterogeneous filamentous structures. This E4-ORF3 activity correlates with the inhibition of PML-mediated antiviral activity. The mechanism of E4-ORF3-mediated reorganization of PML nuclear bodies is unknown. Biophysical analysis of the purified WT E4-ORF3 protein revealed an ordered secondary/tertiary structure and the ability to form heterogeneous higher-order multimers in solution. Importantly, a nonfunctional E4-ORF3 mutant protein, L103A, forms a stable dimer with WT secondary structure content. Because the L103A mutant is incapable of PML reorganization, this result suggests that higher-order multimerization of E4-ORF3 may be required for the activity of the protein. In support of this hypothesis, we demonstrate that the E4-ORF3 L103A mutant protein acts as a dominant-negative effector when coexpressed with the WT E4-ORF3 in mammalian cells. It prevents WT E4-ORF3-mediated PML track formation presumably by binding to the WT protein and inhibiting the formation of higher-order multimers. In vitro protein binding studies support this conclusion as demonstrated by copurification of coexpressed WT and L103A proteins in Escherichia coli and coimmunoprecipitation of WT·L103A E4-ORF3 complexes in mammalian cells. These results provide new insight into the properties of the Ad E4-ORF3 protein and suggest that higher-order protein multimerization is essential for E4-ORF3 activity.
Collapse
Affiliation(s)
- Vadim Patsalo
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York 11794, USA
| | | | | | | | | | | | | | | |
Collapse
|
206
|
Chung I, Osterwald S, Deeg KI, Rippe K. PML body meets telomere: the beginning of an ALTernate ending? Nucleus 2012; 3:263-75. [PMID: 22572954 PMCID: PMC3414403 DOI: 10.4161/nucl.20326] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The unlimited proliferation potential of cancer cells requires the maintenance of their telomeres. This is frequently accomplished by reactivation of telomerase. However, in a significant fraction of tumors an alternative lengthening of telomeres (ALT) mechanism is active. The molecular mechanism of the ALT pathway remains elusive. In particular, the role of characteristic complexes of promyelocytic leukemia nuclear bodies (PML-NBs) with telomeres, the ALT-associated PML-NBs (APBs), is currently under investigation. Here, we review recent findings on the assembly, structure and functions of APBs. It is discussed how genomic aberrations in ALT-positive cancer cells could result in the formation of APBs and in ALT activity. We conclude that they are important functional intermediates in what is considered the canonical ALT pathway and discuss deregulations of cellular pathways that contribute to the emergence of the ALT phenotype.
Collapse
Affiliation(s)
- Inn Chung
- Research Group Genome Organization & Function, Deutsches Krebsforschungszentrum (DKFZ) and BioQuant, Heidelberg, Germany
| | | | | | | |
Collapse
|
207
|
Jiang M, Imperiale MJ. Design stars: how small DNA viruses remodel the host nucleus. Future Virol 2012; 7:445-459. [PMID: 22754587 DOI: 10.2217/fvl.12.38] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Numerous host components are encountered by viruses during the infection process. While some of these host structures are left unchanged, others may go through dramatic remodeling processes. In this review, we summarize these host changes that occur during small DNA virus infections, with a focus on host nuclear components and pathways. Although these viruses differ significantly in their genome structures and infectious pathways, there are common nuclear targets that are altered by various viral factors. Accumulating evidence suggests that these nuclear remodeling processes are often essential for productive viral infections and/or viral-induced transformation. Understanding the complex interactions between viruses and these host structures and pathways will help to build a more integrated network of how the virus completes its life cycle and point toward the design of novel therapeutic regimens that either prevent harmful viral infections or employ viruses as nontraditional treatment options or molecular tools.
Collapse
Affiliation(s)
- Mengxi Jiang
- Department of Microbiology & Immunology, & Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
208
|
The potential link between PML NBs and ICP0 in regulating lytic and latent infection of HSV-1. Protein Cell 2012; 3:372-82. [PMID: 22544561 DOI: 10.1007/s13238-012-2021-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 01/19/2012] [Indexed: 01/28/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a common human pathogen causing cold sores and even more serious diseases. It can establish a latent stage in sensory ganglia after primary epithelial infections, and reactivate in response to stress or sunlight. Previous studies have demonstrated that viral immediate-early protein ICP0 plays a key role in regulating the balance between lytic and latent infection. Recently, It has been determined that promyelocytic leukemia (PML) nuclear bodies (NBs), small nuclear sub-structures, contribute to the repression of HSV-1 infection in the absence of functional ICP0. In this review, we discuss the fundamentals of the interaction between ICP0 and PML NBs, suggesting a potential link between PML NBs and ICP0 in regulating lytic and latent infection of HSV-1.
Collapse
|
209
|
Erickson KD, Bouchet-Marquis C, Heiser K, Szomolanyi-Tsuda E, Mishra R, Lamothe B, Hoenger A, Garcea RL. Virion assembly factories in the nucleus of polyomavirus-infected cells. PLoS Pathog 2012; 8:e1002630. [PMID: 22496654 PMCID: PMC3320610 DOI: 10.1371/journal.ppat.1002630] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 02/22/2012] [Indexed: 11/19/2022] Open
Abstract
Most DNA viruses replicate in the cell nucleus, although the specific sites of virion assembly are as yet poorly defined. Electron microscopy on freeze-substituted, plastic-embedded sections of murine polyomavirus (PyV)-infected 3T3 mouse fibroblasts or mouse embryonic fibroblasts (MEFs) revealed tubular structures in the nucleus adjacent to clusters of assembled virions, with virions apparently “shed” or “budding” from their ends. Promyelocytic leukemia nuclear bodies (PML-NBs) have been suggested as possible sites for viral replication of polyomaviruses (BKV and SV40), herpes simplex virus (HSV), and adenovirus (Ad). Immunohistochemistry and FISH demonstrated co-localization of the viral T-antigen (Tag), PyV DNA, and the host DNA repair protein MRE11, adjacent to the PML-NBs. In PML−/− MEFs the co-localization of MRE11, Tag, and PyV DNA remained unchanged, suggesting that the PML protein itself was not responsible for their association. Furthermore, PyV-infected PML−/− MEFs and PML−/− mice replicated wild-type levels of infectious virus. Therefore, although the PML protein may identify sites of PyV replication, neither the observed “virus factories” nor virus assembly were dependent on PML. The ultrastructure of the tubes suggests a new model for the encapsidation of small DNA viruses. Polyomaviruses are infectious pathogens of mammals and birds that have been linked to the development of cancers in their hosts. Members of the polyomavirus family are associated with human disease, such as JCV and BKV, and over the past few years, several more human polyomaviruses (WUV, KIV and MCV) have been discovered in immune-suppressed individuals. We are studying the way in which these viruses assemble in cells in order to identify critical points where anti-viral therapies could target these viruses. Using a structural, biochemical and cell biological approach, we set out to define sites of virus assembly and virus intermediates. We identified virus-specific structures that we termed “virus factories”. We believe that these sites serve as an assembly line for the production of new viruses. Our study provides new evidence for the presence and composition of virus assembly factories, and identifies a host protein that may be important for infection by polyomaviruses.
Collapse
Affiliation(s)
- Kimberly D. Erickson
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
- The Biofrontiers Institute, University of Colorado, Boulder, Colorado, United States of America
| | - Cedric Bouchet-Marquis
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Katie Heiser
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
- The Biofrontiers Institute, University of Colorado, Boulder, Colorado, United States of America
| | - Eva Szomolanyi-Tsuda
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Rabinarayan Mishra
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Benjamin Lamothe
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Andreas Hoenger
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Robert L. Garcea
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
- The Biofrontiers Institute, University of Colorado, Boulder, Colorado, United States of America
- * E-mail:
| |
Collapse
|
210
|
Wilson VG. Sumoylation at the host-pathogen interface. Biomolecules 2012; 2:203-27. [PMID: 23795346 PMCID: PMC3685863 DOI: 10.3390/biom2020203] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 03/21/2012] [Accepted: 03/27/2012] [Indexed: 12/11/2022] Open
Abstract
Many viral proteins have been shown to be sumoylated with corresponding regulatory effects on their protein function, indicating that this host cell modification process is widely exploited by viral pathogens to control viral activity. In addition to using sumoylation to regulate their own proteins, several viral pathogens have been shown to modulate overall host sumoylation levels. Given the large number of cellular targets for SUMO addition and the breadth of critical cellular processes that are regulated via sumoylation, viral modulation of overall sumoylation presumably alters the cellular environment to ensure that it is favorable for viral reproduction and/or persistence. Like some viruses, certain bacterial plant pathogens also target the sumoylation system, usually decreasing sumoylation to disrupt host anti-pathogen responses. The recent demonstration that Listeria monocytogenes also disrupts host sumoylation, and that this is required for efficient infection, extends the plant pathogen observations to a human pathogen and suggests that pathogen modulation of host sumoylation may be more widespread than previously appreciated. This review will focus on recent aspects of how pathogens modulate the host sumoylation system and how this benefits the pathogen.
Collapse
Affiliation(s)
- Van G Wilson
- Department of Microbial & Molecular Pathogenesis, College of Medicine, Texas A&M Health Science Center, 8447 HWY 47, Bryan, TX 77807-1359
| |
Collapse
|
211
|
Functions of the Epstein-Barr virus EBNA1 protein in viral reactivation and lytic infection. J Virol 2012; 86:6146-58. [PMID: 22491455 DOI: 10.1128/jvi.00013-12] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
EBNA1 is the only nuclear Epstein-Barr virus (EBV) protein expressed in both latent and lytic modes of infection. While EBNA1 is known to play several important roles in latent infection, the reason for its continued expression in lytic infection is unknown. Here we identified two roles for EBNA1 in the reactivation of latent EBV to the lytic cycle in epithelial cells. First, EBNA1 depletion in latently infected cells was shown to positively contribute to spontaneous EBV reactivation, showing that EBNA1 has a role in suppressing reactivation. Second, when the lytic cycle was induced, EBNA1 depletion decreased lytic gene expression and DNA amplification, showing that it positively contributed to lytic infection. Since we have previously shown that EBNA1 disrupts promyelocytic leukemia (PML) nuclear bodies, we investigated whether this function could account for the effects of EBNA1 on lytic infection by repeating the experiments with cells lacking PML proteins. In the absence of PML, EBNA1 did not promote lytic infection, indicating that the EBNA1-mediated PML disruption is responsible for promoting lytic infection. In keeping with this conclusion, PML silencing was found to be sufficient to induce the EBV lytic cycle. Finally, by generating cells with single PML isoforms, we showed that individual PML isoforms were sufficient to suppress EBV lytic reactivation, although PML isoform IV (PML IV) was ineffective because it was most efficiently degraded by EBNA1. Our results provide the first function for EBNA1 in lytic infection and show that EBNA1 interactions with PML IV lead to a loss of PML nuclear bodies (NBs) that promotes lytic infection.
Collapse
|
212
|
Full F, Reuter N, Zielke K, Stamminger T, Ensser A. Herpesvirus saimiri antagonizes nuclear domain 10-instituted intrinsic immunity via an ORF3-mediated selective degradation of cellular protein Sp100. J Virol 2012; 86:3541-53. [PMID: 22278248 PMCID: PMC3302493 DOI: 10.1128/jvi.06992-11] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 01/09/2012] [Indexed: 01/28/2023] Open
Abstract
In recent studies, the nuclear domain 10 (ND10) components PML, Sp100, human Daxx (hDaxx), and ATRX were identified to be cellular restriction factors that are able to inhibit the replication of several herpesviruses. The antiviral function of ND10, however, is antagonized by viral effector proteins by a variety of strategies, including degradation of PML or relocalization of ND10 proteins. In this study, we analyzed the interplay between infection with herpesvirus saimiri (HVS), the prototypic rhadinovirus, and cellular defense by ND10. In contrast to other herpesviruses, we found that HVS specifically degraded the cellular ND10 component Sp100, whereas other factors like PML or hDaxx remained intact. We could further identify the ORF3 tegument protein of HVS, which shares homology with the cellular formylglycinamide ribotide amidotransferase (FGARAT) enzyme, to be the viral factor that induces the proteasomal degradation of Sp100. Interestingly, recent studies showed that the ORF3-homologous proteins ORF75c of murine gammaherpesvirus 68 and BNRF-1 of Epstein-Barr virus modulate the ND10 proteins PML and ATRX, respectively, suggesting that the ND10 targets of viral FGARAT-homologous proteins diversified during evolution. Furthermore, a virus with the ORF3 deletion was efficiently complemented in Sp100-depleted cells, indicating that Sp100 is able to inhibit HVS in the absence of antagonistic mechanisms. In contrast, we observed that PML, which was neither degraded nor redistributed after HVS infection, strongly restricted both wild-type HVS and virus with the ORF3 deletion. Thus, HVS may lack a factor that efficiently counteracts the repressive function of PML, which may foster latency as the outcome of infection.
Collapse
Affiliation(s)
- Florian Full
- Institut für Klinische und Molekulare Virologie, Universitätsklinikum, Friedrich Alexander Universität, Erlangen, Germany
| | | | | | | | | |
Collapse
|
213
|
Ranneberg-Nilsen T, Rollag H, Slettebakk R, Backe PH, Olsen Ø, Luna L, Bjørås M. The chromatin remodeling factor SMARCB1 forms a complex with human cytomegalovirus proteins UL114 and UL44. PLoS One 2012; 7:e34119. [PMID: 22479537 PMCID: PMC3313996 DOI: 10.1371/journal.pone.0034119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 02/22/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Human cytomegalovirus (HCMV) uracil DNA glycosylase, UL114, is required for efficient viral DNA replication. Presumably, UL114 functions as a structural partner to other factors of the DNA-replication machinery and not as a DNA repair protein. UL114 binds UL44 (HCMV processivity factor) and UL54 (HCMV-DNA-polymerase). In the present study we have searched for cellular partners of UL114. METHODOLOGY/PRINCIPAL FINDINGS In a yeast two-hybrid screen SMARCB1, a factor of the SWI/SNF chromatin remodeling complex, was found to be an interacting partner of UL114. This interaction was confirmed in vitro by co-immunoprecipitation and pull-down. Immunofluorescence microscopy revealed that SMARCB1 along with BRG-1, BAF170 and BAF155, which are the core SWI/SNF components required for efficient chromatin remodeling, were present in virus replication foci 24-48 hours post infection (hpi). Furthermore a direct interaction was also demonstrated for SMARCB1 and UL44. CONCLUSIONS/SIGNIFICANCE The core SWI/SNF factors required for efficient chromatin remodeling are present in the HCMV replication foci throughout infection. The proteins UL44 and UL114 interact with SMARCB1 and may participate in the recruitment of the SWI/SNF complex to the chromatinized virus DNA. Thus, the presence of the SWI/SNF chromatin remodeling complex in replication foci and its association with UL114 and with UL44 might imply its involvement in different DNA transactions.
Collapse
Affiliation(s)
- Toril Ranneberg-Nilsen
- Department of Microbiology, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Centre for Molecular Biology and Neuroscience, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Halvor Rollag
- Department of Microbiology, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Ragnhild Slettebakk
- Department of Microbiology, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Paul Hoff Backe
- Department of Microbiology, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Department of Medical Biochemistry, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Centre for Molecular Biology and Neuroscience, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Øyvind Olsen
- Department of Microbiology, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Department of Medical Biochemistry, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Centre for Molecular Biology and Neuroscience, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Luisa Luna
- Department of Microbiology, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Centre for Molecular Biology and Neuroscience, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Magnar Bjørås
- Department of Microbiology, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Department of Medical Biochemistry, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
- Centre for Molecular Biology and Neuroscience, University of Oslo and Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| |
Collapse
|
214
|
Sathish N, Wang X, Yuan Y. Tegument Proteins of Kaposi's Sarcoma-Associated Herpesvirus and Related Gamma-Herpesviruses. Front Microbiol 2012; 3:98. [PMID: 22435068 PMCID: PMC3304090 DOI: 10.3389/fmicb.2012.00098] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 02/28/2012] [Indexed: 12/12/2022] Open
Abstract
A herpesvirus virion is composed of a viral genomic DNA-containing capsid surrounded by a viral envelope with glycoprotein spikes on its surface. Located between the capsid and the outer viral envelope is the virion tegument layer. Though the majority of the virion proteins are located in the tegument, this layer is less studied and was thought to be an amorphous structure. Over the last decade, a number of studies have indicated the presence of organized tegument structures across the spectrum of herpesviruses, implicating tegument components in critical steps governing the viral life cycle. In the case of Kaposi’s sarcoma-associated herpesvirus (KSHV), the etiological agent of Kaposi’s sarcoma, several functions exerted by tegument proteins at different stages of the viral life cycle, inclusive of primary de novo infection and virion assembly, have been identified over the last several years. In this review, KSHV tegument components are cataloged and the occurrence of organized tegument structures in KSHV, built through interactions amongst the different virion proteins, is discussed in depth. The significant functional roles of the KSHV tegument proteins at different stages of the viral life cycle are elaborated under separate headings. Definitive functional roles exerted by tegument proteins of related gamma-herpesviruses are also discussed. Since tegument proteins play key roles during viral assembly, viral entry, and represent an important interface for virus–host interactions, further research in this area should provide detailed insights into the functional capacity of the KSHV tegument, resulting in a better understanding of the viral life cycle.
Collapse
Affiliation(s)
- Narayanan Sathish
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal Bhopal, Madhya Pradesh, India
| | | | | |
Collapse
|
215
|
Abstract
Viral persistence is the rule following infection with all herpesviruses. The β-herpesvirus, human cytomegalovirus (HCMV), persists through chronic and latent states of infection. Both of these states of infection contribute to HCMV persistence and to the high HCMV seroprevalence worldwide. The chronic infection is poorly defined molecularly, but clinically manifests as low-level virus shedding over extended periods of time and often in the absence of symptoms. Latency requires long-term maintenance of viral genomes in a reversibly quiescent state in the immunocompetent host. In this review, we focus on recent advances in the biology of HCMV persistence, particularly with respect to the latent mode of persistence. Latently infected individuals harbour HCMV genomes in haematopoietic cells and maintain large subsets of HCMV-specific T-cells. In the last few years, impressive advances have been made in understanding virus-host interactions important to HCMV infection, many of which will profoundly impact HCMV persistence. We discuss these advances and their known or potential impact on viral latency. As herpesviruses are met with similar challenges in achieving latency and often employ conserved strategies to persist, we discuss current and future directions of HCMV persistence in the context of the greater body of knowledge regarding α- and γ-herpesviruses persistence.
Collapse
Affiliation(s)
- Felicia Goodrum
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | | | | |
Collapse
|
216
|
Willmes C, Adam C, Alb M, Völkert L, Houben R, Becker JC, Schrama D. Type I and II IFNs inhibit Merkel cell carcinoma via modulation of the Merkel cell polyomavirus T antigens. Cancer Res 2012; 72:2120-8. [PMID: 22389452 DOI: 10.1158/0008-5472.can-11-2651] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Merkel cell carcinoma (MCC) is a rare and highly aggressive skin cancer associated with the Merkel cell polyomavirus (MCV). As MCC cell lines show oncogene addiction to the MCV T antigens, pharmacologic interference of the large T antigen (LTA) may represent an effective therapeutic approach for this deadly cancer. In this study, we investigated the effects of IFNs on MCC cell lines, especially on MCV-positive (MCV(+)) lines. Type I IFNs (i.e., Multiferon, a mix of different IFN-α subtypes, and IFN-β) strongly inhibited the cellular viability. Cell-cycle analysis showed increased sub-G fractions for these cells upon IFN treatment indicating apoptotic cell death; these effects were less pronounced for IFN-γ. Notably, this inhibitory effect of type I IFNs on MCV(+) MCC cell lines was associated with a reduced expression of the MCV LTA as well as an increased expression of promyelocytic leukemia (PML) protein, which is known to interfere with the function of the LTA. In addition, the intratumoral application of Multiferon resulted in a regression of MCV(+) but not MCV(-) MCCs in vivo. Together, our findings show that type I IFNs have a strong antitumor effect, which is at least in part explained by modulation of the virally encoded LTA.
Collapse
Affiliation(s)
- Christoph Willmes
- Department of Dermatology, University Hospital of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
217
|
Abstract
The tripartite motif (TRIM)-containing proteins are involved in many cellular functions such as cell signaling, apoptosis, cell differentiation, and immune modulation. TRIM5 proteins, including TRIM5α and TRIM-Cyp, are known to possess antiretroviral activity against many different retroviruses. Besides being retroviral restriction factors, TRIM5 proteins participate in other cellular functions that have recently emerged in the study of TRIM5α. In this review, we discuss properties of TRIM5α such as cytoplasmic body formation, protein turnover, and trafficking. Also, we discuss recent insights into innate immune modulation mediated by TRIM5α, highlighting the various functions TRIM5α has in cellular processes.
Collapse
Affiliation(s)
- Zana Lukic
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Edward M. Campbell
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL 60153, USA
| |
Collapse
|
218
|
Sandri-Goldin RM. The many roles of the highly interactive HSV protein ICP27, a key regulator of infection. Future Microbiol 2012; 6:1261-77. [PMID: 22082288 DOI: 10.2217/fmb.11.119] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Human herpes viruses cause an array of illnesses ranging from cancers for Epstein?Barr virus and Kaposi?s sarcoma-associated herpes virus, to painful skin lesions, and more rarely, keratitis and encephalitis for HSV. All herpes viruses encode a multifunctional protein, typified by HSV ICP27, which plays essential roles in viral infection. ICP27 functions in all stages of mRNA biogenesis from transcription, RNA processing and export through to translation. ICP27 has also been implicated in nuclear protein quality control, cell cycle control, activation of stress signaling pathways and prevention of apoptosis. ICP27 interacts with many proteins and it binds RNA. This article focuses on how ICP27 performs its many roles and highlights similarities with its homologs, which could be targets for antiviral intervention.
Collapse
Affiliation(s)
- Rozanne M Sandri-Goldin
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
219
|
Glauser DL, Gillet L, Stevenson PG. Virion endocytosis is a major target for murid herpesvirus-4 neutralization. J Gen Virol 2012; 93:1316-1327. [PMID: 22377583 PMCID: PMC3755512 DOI: 10.1099/vir.0.040790-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpesviruses consistently transmit from immunocompetent carriers, implying that their neutralization is hard to achieve. Murid herpesvirus-4 (MuHV-4) exploits host IgG Fc receptors to bypass blocks to cell binding, and pH-dependent protein conformation changes to unveil its fusion machinery only after endocytosis. Nevertheless, neutralization remains possible by targeting the virion glycoprotein H (gH)–gL heterodimer, and the neutralizing antibody responses of MuHV-4 carriers are improved by boosting with recombinant gH–gL. We analysed here how gH–gL-directed neutralization works. The MuHV-4 gH–gL binds to heparan sulfate. However, most gH–gL-specific neutralizing antibodies did not block this interaction; neither did they act directly on fusion. Instead, they blocked virion endocytosis and transport to the late endosomes, where membrane fusion normally occurs. The poor endocytosis of gH–gL-neutralized virions was recapitulated precisely by virions genetically lacking gL. Therefore, driving virion uptake appears to be an important function of gH–gL that provides a major target for antibody-mediated neutralization.
Collapse
Affiliation(s)
- Daniel L. Glauser
- Division of Virology, Department of Pathology, University of Cambridge, UK
| | - Laurent Gillet
- Immunology–Vaccinology, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | | |
Collapse
|
220
|
Transcriptional activation of the adenoviral genome is mediated by capsid protein VI. PLoS Pathog 2012; 8:e1002549. [PMID: 22427750 PMCID: PMC3303589 DOI: 10.1371/journal.ppat.1002549] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 01/10/2012] [Indexed: 12/12/2022] Open
Abstract
Gene expression of DNA viruses requires nuclear import of the viral genome. Human
Adenoviruses (Ads), like most DNA viruses, encode factors within early
transcription units promoting their own gene expression and counteracting
cellular antiviral defense mechanisms. The cellular transcriptional repressor
Daxx prevents viral gene expression through the assembly of repressive chromatin
remodeling complexes targeting incoming viral genomes. However, it has remained
unclear how initial transcriptional activation of the adenoviral genome is
achieved. Here we show that Daxx mediated repression of the immediate early Ad
E1A promoter is efficiently counteracted by the capsid protein VI. This requires
a conserved PPxY motif in protein VI. Capsid proteins from other DNA viruses
were also shown to activate the Ad E1A promoter independent of Ad gene
expression and support virus replication. Our results show how Ad entry is
connected to transcriptional activation of their genome in the nucleus. Our data
further suggest a common principle for genome activation of DNA viruses by
counteracting Daxx related repressive mechanisms through virion proteins. To initiate infection, DNA viruses deliver their genome to the nucleus and
express viral genes required for genome replication. Efficient transport is
achieved by packing the viral genome as a condensed, transcriptionally inactive
nucleo-protein complex. However, for most DNA viruses, including Adenoviruses
(Ads), it remains unclear how the viral genome is decondensed and how
transcription is initiated inside the nucleus. Cells control unwanted gene
expression by chromatin modification mediated through transcriptionally
repressive complexes. A key factor in repressive complex assemblies is the
transcriptional repressor Daxx. The Ad structural capsid protein VI is required
for endosomal escape and nuclear transport. Here we show that protein VI also
activates the Ad E1A promoter to initiate Ad gene expression. This is achieved
through the removal of Daxx repression from the E1A promoter, which requires a
conserved ubiquitin ligase interacting motif (PPxY-motif) in protein VI. We
further show that capsid proteins from other unrelated DNA viruses also activate
the Ad E1A promoter and support Ad replication by counteracting Daxx repression,
functionally replacing protein VI. Our data suggest that reversal of Daxx
repression by virion proteins is a widespread mechanism among DNA viruses that
is not restricted to a single virus family.
Collapse
|
221
|
Shishido-Hara Y, Ichinose S, Uchihara T. JC virus intranuclear inclusions associated with PML-NBs: analysis by electron microscopy and structured illumination microscopy. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1095-1106. [PMID: 22266251 DOI: 10.1016/j.ajpath.2011.11.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 11/03/2011] [Accepted: 11/29/2011] [Indexed: 01/08/2023]
Abstract
Progressive multifocal leukoencephalopathy is a fatal demyelinating disorder caused by JC virus infection. JC virus was recently found to target promyelocytic leukemia nuclear bodies (PML-NBs), punctuate domains in the nuclei. Thus, the virus progenies cluster in dots as intranuclear inclusions (ie, as dot-shaped inclusions). In the present study, both the viral major and minor capsid proteins were expressed from polycistronic expression vectors with a powerful promoter, and formation into virus-like particles (VLPs) was examined by electron microscopy. When the upstream regulatory sequence including the agnogene (nt 275 to 490) was present, capsid protein expression was suppressed, but numerous VLPs were efficiently formed with restricted accumulation to PML-NBs. VLPs were uniform, and the cells were severely degraded. In contrast, when the 5' terminus of the agnogene (nt 275 to 409; 135 bp) was deleted, capsid protein expression was markedly enhanced, but VLPs were more randomly produced in the nucleus outside of PML-NBs. VLPs were pleomorphic, and cell degradation was minimal. JC virus association with PML-NBs was confirmed in human brain tissues by structured illumination microscopy. PML-NBs were shaped in spherical shells, with viral capsid proteins circumscribing the surface. These findings indicate that PML-NBs are intranuclear locations for pathogenic JC virus proliferation. Either the agnogene or its product likely supports efficient progeny production at PML-NBs, leading to subsequent degeneration of host glial cells.
Collapse
Affiliation(s)
| | - Shizuko Ichinose
- Research Center for Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshiki Uchihara
- Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
222
|
Frappier L. The Epstein-Barr Virus EBNA1 Protein. SCIENTIFICA 2012; 2012:438204. [PMID: 24278697 PMCID: PMC3820569 DOI: 10.6064/2012/438204] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 11/28/2012] [Indexed: 05/06/2023]
Abstract
Epstein-Barr virus (EBV) is a widespread human herpes virus that immortalizes cells as part of its latent infection and is a causative agent in the development of several types of lymphomas and carcinomas. Replication and stable persistence of the EBV genomes in latent infection require the viral EBNA1 protein, which binds specific DNA sequences in the viral DNA. While the roles of EBNA1 were initially thought to be limited to effects on the viral genomes, more recently EBNA1 has been found to have multiple effects on cellular proteins and pathways that may also be important for viral persistence. In addition, a role for EBNA1 in lytic infection has been recently identified. The multiple roles of EBNA1 in EBV infection are the subject of this paper.
Collapse
Affiliation(s)
- Lori Frappier
- Department of Molecular Genetics, University of Toronto, 1 Kings College Circle, Toronto, ON, Canada M5S 1A8
- *Lori Frappier:
| |
Collapse
|
223
|
Batty EC, Jensen K, Freemont PS. PML nuclear bodies and other TRIM-defined subcellular compartments. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 770:39-58. [PMID: 23630999 DOI: 10.1007/978-1-4614-5398-7_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Tripartite motif (TRIM) proteins are defined by their possession of a RING, B-box and predicted coiled coil (RBCC) domain. The coiled-coil region facilitates the oligomerisation of TRIMs and contributes to the formation of high molecular weight complexes that show interesting subcellular compartmentalisations and structures. TRIM protein compartments include both nuclear and cytoplasmic filaments and aggregates (bodies), as well as diffuse subcellular distributions. TRIM 19, otherwise known as promyelocytic leukaemia (PML) protein forms nuclear aggregates termed PML nuclear bodies (PML NBs), at which a number of functionally diverse proteins transiently or covalently associate. PML NBs are therefore implicated in a wide variety of cellular functions such as transcriptional regulation, viral response, apoptosis and nuclear protein storage.
Collapse
Affiliation(s)
- Elizabeth C Batty
- Macromolecular Structure and Function Group, Division of Molecular Biosciences, Imperial College London, South Kensington, London, UK
| | | | | |
Collapse
|
224
|
TRIM Proteins and the Innate Immune Response to Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 770:93-104. [DOI: 10.1007/978-1-4614-5398-7_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
225
|
Adenovirus E4orf3 targets transcriptional intermediary factor 1γ for proteasome-dependent degradation during infection. J Virol 2011; 86:3167-79. [PMID: 22205733 DOI: 10.1128/jvi.06583-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The ability of adenovirus early region proteins, E1B-55K and E4orf6, to usurp control of cellular ubiquitin ligases and target proteins for proteasome-dependent degradation during infection is well established. Here we show that the E4 gene product, E4orf3 can, independently of E1B-55K and E4orf6, target the transcriptional corepressor transcriptional intermediary factor 1γ (TIF1γ) for proteasome-mediated degradation during infection. Initial mass spectrometric studies identified TIF1 family members-TIF1α, TIF1β, and TIF1γ-as E1B-55K-binding proteins in both transformed and infected cells, but analyses revealed that, akin to TIF1α, TIF1γ is reorganized in an E4orf3-dependent manner to promyelocytic leukemia protein-containing nuclear tracks during infection. The use of a number of different adenovirus early region mutants identified the specific and sole requirement for E4orf3 in mediating TIF1γ degradation. Further analyses revealed that TIF1γ is targeted for degradation by a number of divergent human adenoviruses, suggesting that the ability of E4orf3 to regulate TIF1γ expression is evolutionarily conserved. We also determined that E4orf3 does not utilize the Cullin-based ubiquitin ligases, CRL2 and CRL5, or the TIF1α ubiquitin ligase in order to promote TIF1γ degradation. Further studies suggested that TIF1γ possesses antiviral activity and limits adenovirus early and late gene product expression during infection. Indeed, TIF1γ knockdown accelerates the adenovirus-mediated degradation of MRE11, while TIF1γ overexpression delays the adenovirus-mediated degradation of MRE11. Taken together, these studies have identified novel adenovirus targets and have established a new role for the E4orf3 protein during infection.
Collapse
|
226
|
Everett RD. The use of fluorescence microscopy to study the association between herpesviruses and intrinsic resistance factors. Viruses 2011; 3:2412-24. [PMID: 22355446 PMCID: PMC3280513 DOI: 10.3390/v3122412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 12/01/2011] [Accepted: 12/01/2011] [Indexed: 01/09/2023] Open
Abstract
Intrinsic antiviral resistance is a branch of antiviral defence that involves constitutively expressed cellular proteins that act within individual infected cells. In recent years it has been discovered that components of cellular nuclear structures known as ND10 or PML nuclear bodies contribute to intrinsic resistance against a variety of viruses, notably of the herpesvirus family. Several ND10 components are rapidly recruited to sites that are closely associated with herpes simplex virus type 1 (HSV-1) genomes during the earliest stages of infection, and this property correlates with the efficiency of ND10 mediated restriction of HSV-1 replication. Similar but distinct recruitment of certain DNA damage response proteins also occurs during infection. These recruitment events are inhibited in a normal wild type HSV-1 infection by the viral regulatory protein ICP0. HSV‑1 mutants that do not express ICP0 are highly susceptible to repression through intrinsic resistance factors, but they replicate more efficiently in cells depleted of certain ND10 proteins or in which ND10 component recruitment is inefficient. This article presents the background to this recruitment phenomenon and summaries how it is conveniently studied by fluorescence microscopy.
Collapse
Affiliation(s)
- Roger D Everett
- The Medical Research Council (MRC), University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, Scotland, UK.
| |
Collapse
|
227
|
Verrier ER, Langevin C, Benmansour A, Boudinot P. Early antiviral response and virus-induced genes in fish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:1204-1214. [PMID: 21414349 DOI: 10.1016/j.dci.2011.03.012] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 12/21/2010] [Accepted: 03/06/2011] [Indexed: 05/30/2023]
Abstract
In fish as in mammals, virus infections induce changes in the expression of many host genes. Studies conducted during the last fifteen years revealed a major contribution of the interferon system in fish antiviral response. This review describes the screening methods applied to compare the impact of virus infections on the transcriptome in different fish species. These approaches identified a "core" set of genes that are strongly induced in most viral infections. The "core" interferon-induced genes (ISGs) are generally conserved in vertebrates, some of them inhibiting a wide range of viruses in mammals. A selection of ISGs -PKR, vig-1/viperin, Mx, ISG15 and finTRIMs - is further analyzed here to illustrate the diversity and complexity of the mechanisms involved in establishing an antiviral state. Most of the ISG-based pathways remain to be directly determined in fish. Fish ISGs are often duplicated and the functional specialization of multigenic families will be of particular interest for future studies.
Collapse
Affiliation(s)
- Eloi R Verrier
- INRA, Fish Infection and Immunity, Molecular Virology and Immunology, Domaine de Vilvert, 78352 Jouy en Josas, France
| | | | | | | |
Collapse
|
228
|
Vink EI, Yondola MA, Wu K, Hearing P. Adenovirus E4-ORF3-dependent relocalization of TIF1α and TIF1γ relies on access to the Coiled-Coil motif. Virology 2011; 422:317-25. [PMID: 22123502 DOI: 10.1016/j.virol.2011.10.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 08/19/2011] [Accepted: 10/10/2011] [Indexed: 10/14/2022]
Abstract
The adenovirus E4-ORF3 protein promotes viral replication by relocalizing cellular proteins into nuclear track structures, interfering with potential anti-viral activities. E4-ORF3 targets transcriptional intermediary factor 1 alpha (TIF1α), but not homologous TIF1β. Here, we introduce TIF1γ as a novel E4-ORF3-interacting partner. E4-ORF3 relocalizes endogenous TIF1γ in virus-infected cells in vivo and binds to TIF1γ in vitro. We used the homologous nature, yet differing binding capabilities, of these proteins to study how E4-ORF3 targets proteins for track localization. We mapped the ability of E4-ORF3 to interact with specific TIF1 subdomains, demonstrating that E4-ORF3 interacts with the Coiled-Coil domains of TIF1α, TIF1β, and TIF1γ, and that the C-terminal half of TIF1β interferes with this interaction. The results of E4-ORF3-directed TIF1 protein relocalization assays performed in vivo were verified using coimmunoprecipitation assays in vitro. These results suggest that E4-ORF3 targets proteins for relocalization through a loosely homologous sequence dependent on accessibility.
Collapse
Affiliation(s)
- Elizabeth I Vink
- Department of Molecular Genetics and Microbiology, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | | |
Collapse
|
229
|
Lunardi A, Gaboli M, Giorgio M, Rivi R, Bygrave A, Antoniou M, Drabek D, Dzierzak E, Fagioli M, Salmena L, Botto M, Cordon-Cardo C, Luzzatto L, Pelicci PG, Grosveld F, Pandolfi PP. A Role for PML in Innate Immunity. Genes Cancer 2011; 2:10-9. [PMID: 21779477 DOI: 10.1177/1947601911402682] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Accepted: 02/09/2011] [Indexed: 12/13/2022] Open
Abstract
The promyelocytic leukemia gene (PML) of acute promyelocytic leukemia is an established tumor suppressor gene with critical functions in growth suppression, induction of apoptosis, and cellular senescence. Interestingly, although less studied, PML seems to play a key role also in immune response to viral infection. Herein, we report that Pml(-/-) mice spontaneously develop an atypical invasive and lethal granulomatous lesion known as botryomycosis (BTM). In Pml(-/-) mice, BTM is the result of impaired function of macrophages, whereby they fail to become activated and are thus unable to clear pathogenic microorganisms. Accordingly, Pml(-/-) mice are resistant to lipopolysaccharide (LPS)-induced septic shock as a result of an ineffective production of cytokines and chemokines, suggesting a role for PML in the innate immune Toll-like receptor (TLR)/NF-κB prosurvival pathway. These results not only shed light on a new fundamental function of PML in innate immunity, but they also point to a proto-oncogenic role for PML in certain cellular and pathological contexts.
Collapse
Affiliation(s)
- Andrea Lunardi
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Tsai K, Thikmyanova N, Wojcechowskyj JA, Delecluse HJ, Lieberman PM. EBV tegument protein BNRF1 disrupts DAXX-ATRX to activate viral early gene transcription. PLoS Pathog 2011; 7:e1002376. [PMID: 22102817 PMCID: PMC3213115 DOI: 10.1371/journal.ppat.1002376] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 09/28/2011] [Indexed: 12/12/2022] Open
Abstract
Productive infection by herpesviruses involve the disabling of host-cell intrinsic defenses by viral encoded tegument proteins. Epstein-Barr Virus (EBV) typically establishes a non-productive, latent infection and it remains unclear how it confronts the host-cell intrinsic defenses that restrict viral gene expression. Here, we show that the EBV major tegument protein BNRF1 targets host-cell intrinsic defense proteins and promotes viral early gene activation. Specifically, we demonstrate that BNRF1 interacts with the host nuclear protein Daxx at PML nuclear bodies (PML-NBs) and disrupts the formation of the Daxx-ATRX chromatin remodeling complex. We mapped the Daxx interaction domain on BNRF1, and show that this domain is important for supporting EBV primary infection. Through reverse transcription PCR and infection assays, we show that BNRF1 supports viral gene expression upon early infection, and that this function is dependent on the Daxx-interaction domain. Lastly, we show that knockdown of Daxx and ATRX induces reactivation of EBV from latently infected lymphoblastoid cell lines (LCLs), suggesting that Daxx and ATRX play a role in the regulation of viral chromatin. Taken together, our data demonstrate an important role of BNRF1 in supporting EBV early infection by interacting with Daxx and ATRX; and suggest that tegument disruption of PML-NB-associated antiviral resistances is a universal requirement for herpesvirus infection in the nucleus.
Collapse
Affiliation(s)
- Kevin Tsai
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- Cell and Molecular Biology Program, The University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Jason A. Wojcechowskyj
- Cell and Molecular Biology Program, The University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Paul M. Lieberman
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
231
|
Abstract
Since posttranslational modification (PTM) by the small ubiquitin-related modifiers (SUMOs) was discovered over a decade ago, a huge number of cellular proteins have been found to be reversibly modified, resulting in alteration of differential cellular pathways. Although the molecular consequences of SUMO attachment are difficult to predict, the underlying principle of SUMOylation is altering inter- and/or intramolecular interactions of the modified substrate, changing localization, stability, and/or activity. Unsurprisingly, many different pathogens have evolved to exploit the cellular SUMO modification system due to its functional flexibility and far-reaching functional downstream consequences. Although the extensive knowledge gained so far is impressive, a definitive conclusion about the role of SUMO modification during virus infection in general remains elusive and is still restricted to a few, yet promising concepts. Based on the available data, this review aims, first, to provide a detailed overview of the current state of knowledge and, second, to evaluate the currently known common principles/molecular mechanisms of how human pathogenic microbes, especially viruses and their regulatory proteins, exploit the host cell SUMO modification system.
Collapse
|
232
|
Kim YE, Lee JH, Kim ET, Shin HJ, Gu SY, Seol HS, Ling PD, Lee CH, Ahn JH. Human cytomegalovirus infection causes degradation of Sp100 proteins that suppress viral gene expression. J Virol 2011; 85:11928-37. [PMID: 21880768 PMCID: PMC3209270 DOI: 10.1128/jvi.00758-11] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 08/18/2011] [Indexed: 01/02/2023] Open
Abstract
The interferon-inducible Sp100 proteins are thought to play roles in the chromatin pathway and in transcriptional regulation. Sp100A, the smallest isoform, is one of the major components of PML nuclear bodies (NBs) that exhibit intrinsic antiviral activity against several viruses. Since PML NBs are disrupted by the immediate-early 1 (IE1) protein during human cytomegalovirus (HCMV) infection, the modulation of Sp100 protein expression or activity during infection has been suggested. Here, we show that Sp100 proteins are lost largely in the late stages of HCMV infection. This event required viral gene expression and involved posttranscriptional control. The mutant virus with deletion of the sequence for IE1 (CR208) did not have Sp100 loss. In CR208 infection, PML depletion by RNA interference abrogated the accumulation of SUMO-modified Sp100A and of certain high-molecular-weight Sp100 isoforms but did not significantly affect unmodified Sp100A, suggesting that the IE1-induced disruption of PML NBs is not sufficient for the complete loss of Sp100 proteins. Sp100A loss was found to require proteasome activity. Depletion of all Sp100 proteins by RNA silencing enhanced HCMV replication and major IE (MIE) gene expression. Sp100 knockdown enhanced the acetylation level of histones associated with the MIE promoter, demonstrating that the repressive effect of Sp100 proteins may involve, at least in part, the epigenetic control of the MIE promoter. Sp100A was found to interact directly with IE1 through the N-terminal dimerization domain. These findings indicate that the IE1-dependent loss of Sp100 proteins during HCMV infection may represent an important requirement for efficient viral growth.
Collapse
Affiliation(s)
- Young-Eui Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon
| | - Jin-Hyoung Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon
| | - Eui Tae Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon
| | - Hye Jin Shin
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon
| | - Su Yeon Gu
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon
| | - Hyang Sook Seol
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon
| | - Paul D. Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Chan Hee Lee
- Division of Life Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Jin-Hyun Ahn
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon
| |
Collapse
|
233
|
Bauer DC, Willadsen K, Buske FA, Lê Cao KA, Bailey TL, Dellaire G, Bodén M. Sorting the nuclear proteome. ACTA ACUST UNITED AC 2011; 27:i7-14. [PMID: 21685104 PMCID: PMC3117375 DOI: 10.1093/bioinformatics/btr217] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Motivation: Quantitative experimental analyses of the nuclear interior reveal a morphologically structured yet dynamic mix of membraneless compartments. Major nuclear events depend on the functional integrity and timely assembly of these intra-nuclear compartments. Yet, unknown drivers of protein mobility ensure that they are in the right place at the time when they are needed. Results: This study investigates determinants of associations between eight intra-nuclear compartments and their proteins in heterogeneous genome-wide data. We develop a model based on a range of candidate determinants, capable of mapping the intra-nuclear organization of proteins. The model integrates protein interactions, protein domains, post-translational modification sites and protein sequence data. The predictions of our model are accurate with a mean AUC (over all compartments) of 0.71. We present a complete map of the association of 3567 mouse nuclear proteins with intra-nuclear compartments. Each decision is explained in terms of essential interactions and domains, and qualified with a false discovery assessment. Using this resource, we uncover the collective role of transcription factors in each of the compartments. We create diagrams illustrating the outcomes of a Gene Ontology enrichment analysis. Associated with an extensive range of transcription factors, the analysis suggests that PML bodies coordinate regulatory immune responses. Contact:m.boden@uq.edu.au Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Denis C Bauer
- Queensland Brain Institute, School of Chemistry and Molecular Biosciences, Queensland Facility for Advanced Bioinformatics, The University of Queensland, St Lucia, Australia
| | | | | | | | | | | | | |
Collapse
|
234
|
Contributions of the Epstein-Barr virus EBNA1 protein to gastric carcinoma. J Virol 2011; 86:60-8. [PMID: 22013060 DOI: 10.1128/jvi.05623-11] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Approximately 10% of gastric carcinomas (GC) are comprised of cells latently infected with Epstein-Barr virus (EBV); however, the mechanism by which EBV contributes to the development of this malignancy is unclear. We have investigated the cellular effects of the only EBV nuclear protein expressed in GC, EBNA1, focusing on promyelocytic leukemia (PML) nuclear bodies (NBs), which play important roles in apoptosis, p53 activation, and tumor suppression. AGS GC cells infected with EBV were found to contain fewer PML NBs and less PML protein than the parental EBV-negative AGS cells, and these levels were restored by silencing EBNA1. Conversely, EBNA1 expression was sufficient to induce the loss of PML NBs and proteins in AGS cells. Consistent with PML functions, EBNA1 expression decreased p53 activation and apoptosis in response to DNA damage and resulted in increased cell survival. In addition, EBNA1 mutants unable to bind CK2 kinase or ubiquitin-specific protease 7 had decreased ability to induce PML loss and to interfere with p53 activation. PML levels in EBV-positive and EBV-negative GC biopsy specimens were then compared by immunohistochemistry. Consistent with the results in the AGS cells, EBV-positive tumors had significantly lower PML levels than EBV-negative tumors. The results indicate that EBV infection of GC cells leads to loss of PML NBs through the action of EBNA1, resulting in impaired responses to DNA damage and promotion of cell survival. Therefore, PML disruption by EBNA1 is one mechanism by which EBV may contribute to the development of gastric cancer.
Collapse
|
235
|
Physical and functional interaction between PML and TBX2 in the establishment of cellular senescence. EMBO J 2011; 31:95-109. [PMID: 22002537 DOI: 10.1038/emboj.2011.370] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 09/19/2011] [Indexed: 11/09/2022] Open
Abstract
Cellular senescence acts as a potent barrier for tumour initiation and progression. Previous studies showed that the PML tumour suppressor promotes senescence, although the precise mechanisms remain to be elucidated. Combining gene expression profiling with chromatin-binding analyses and promoter reporter studies, we identify TBX2, a T-box transcription factor frequently overexpressed in cancer, as a novel and direct PML-repressible E2F-target gene in senescence but not quiescence. Recruitment of PML to the TBX2 promoter is dependent on a functional p130/E2F4 repressor complex ultimately implementing a transcriptionally inactive chromatin environment at the TBX2 promoter. TBX2 repression actively contributes to senescence induction as cells depleted for TBX2 trigger PML pro-senescence function(s) and enter senescence. Reciprocally, elevated TBX2 levels antagonize PML pro-senescence function through direct protein-protein interaction. Collectively, our findings indicate that PML and TBX2 act in an autoregulatory loop to control the effective execution of the senescence program.
Collapse
|
236
|
Promyelocytic leukemia isoform IV confers resistance to encephalomyocarditis virus via the sequestration of 3D polymerase in nuclear bodies. J Virol 2011; 85:13164-73. [PMID: 21994459 DOI: 10.1128/jvi.05808-11] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Promyelocytic leukemia (PML) protein is the organizer of nuclear matrix-associated nuclear bodies (NBs), and its conjugation to the small ubiquitin-like modifier (SUMO) is required for the formation of these structures. Several alternatively spliced PML transcripts from a single PML gene lead to the production of seven PML isoforms (PML isoform I [PMLI] to VII [PMLVII]), which all share a N-terminal region that includes the RBCC (RING, B boxes, and a α-helical coiled-coil) motif but differ in the C-terminal region. This diversity of PML isoforms determines the specific functions of each isoform. There is increasing evidence implicating PML in host antiviral defense and suggesting various strategies involving PML to counteract viral production. We reported that mouse embryonic fibroblasts derived from PML knockout mice are more sensitive than wild-type cells to infection with encephalomyocarditis virus (EMCV). Here, we show that stable expression of PMLIV or PMLIVa inhibited viral replication and protein synthesis, leading to a substantial reduction of EMCV multiplication. This protective effect required PMLIV SUMOylation and was not observed with other nuclear PML isoforms (I, II, III, V, and VI) or with the cytoplasmic PMLVII. We demonstrated that only PMLIV interacted with EMCV 3D polymerase (3Dpol) and sequestered it within PML NBs. The C-terminal region specific to PMLIV was required for both interaction with 3Dpol and the antiviral properties. Also, depletion of PMLIV by RNA interference significantly boosted EMCV production in interferon-treated cells. These findings indicate the mechanism by which PML confers resistance to EMCV. They also reveal a new pathway mediating the antiviral activity of interferon against EMCV.
Collapse
|
237
|
Sides MD, Block GJ, Chadwick RW, Shan B, Flemington EK, Lasky JA. Epstein - Barr virus Latent Membrane Protein 1 suppresses reporter activity through modulation of promyelocytic leukemia protein-nuclear bodies. Virol J 2011; 8:461. [PMID: 21975125 PMCID: PMC3204298 DOI: 10.1186/1743-422x-8-461] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Accepted: 10/05/2011] [Indexed: 12/20/2022] Open
Abstract
The Epstein-Barr virus (EBV) encoded Latent Membrane Protein 1 (LMP1) has been shown to increase the expression of promyelocytic leukemia protein (PML) and the immunofluorescent intensity of promyelocytic leukemia nuclear bodies (PML NBs). PML NBs have been implicated in the modulation of transcription and the association of reporter plasmids with PML NBs has been implicated in repression of reporter activity. Additionally, repression of various reporters in the presence of LMP1 has been noted. This study demonstrates that LMP1 suppresses expression of reporter activity in a dose responsive manner and corresponds with the LMP1 induced increase in PML NB intensity. Disruption of PML NBs with arsenic trioxide or a PML siRNA restores reporter activity. These data offer an explanation for previously conflicting data on LMP1 signaling and calls attention to the possibility of false-positives and false-negatives when using reporter assays as a research tool in cells expressing LMP1.
Collapse
Affiliation(s)
- Mark D Sides
- Department of Medicine, Section of Pulmonary Disease and Critical Care, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | | | | | | | |
Collapse
|
238
|
Li C, Sun SY, Khuri FR, Li R. Pleiotropic functions of EAPII/TTRAP/TDP2: cancer development, chemoresistance and beyond. Cell Cycle 2011; 10:3274-83. [PMID: 21926483 DOI: 10.4161/cc.10.19.17763] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
EAPII (also called TTRAP, TDP2), a protein identified a decade ago, has recently been shown to function as an oncogenic factor. This protein was also proven to be the first 5'- tyrosyl-DNA phosphodiesterase. EAPII has been demonstrated to have promiscuous protein associations, broad responsiveness to various extracellular signals, and pleiotropic functions in the development of human diseases including cancer and neurodegenerative disease. Emerging data suggest that EAPII is a multi-functional protein: EAPII repairs enzyme (topoisomerase)-mediated DNA damage by removing phosphotyrosine from DNA adducts; EAPII is involved in multiple signal transduction pathways such as TNF-TNFR, TGFβ and MAPK, and EAPII is responsive to immune defense, inflammatory response, virus infection and DNA toxins (chemo or radiation therapy). This review focuses on the current understanding of EAPII biology and its potential relations to many aspects of cancer development, including chromosome instability, tumorigenesis, tumor metastasis and chemoresistance, suggesting it as a potential target for intervention in cancer and other human diseases.
Collapse
Affiliation(s)
- Chunyang Li
- Winship Cancer Institute, Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
239
|
DNA mismatch repair proteins are required for efficient herpes simplex virus 1 replication. J Virol 2011; 85:12241-53. [PMID: 21957315 DOI: 10.1128/jvi.05487-11] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) is a double-stranded DNA virus that replicates in the nucleus of its human host cell and is known to interact with many cellular DNA repair proteins. In this study, we examined the role of cellular mismatch repair (MMR) proteins in the virus life cycle. Both MSH2 and MLH1 are required for efficient replication of HSV-1 in normal human cells and are localized to viral replication compartments. In addition, a previously reported interaction between MSH6 and ICP8 was confirmed by coimmunoprecipitation and extended to show that UL12 is also present in this complex. We also report for the first time that MLH1 associates with ND10 nuclear bodies and that like other ND10 proteins, MLH1 is recruited to the incoming genome. Knockdown of MLH1 inhibits immediate-early viral gene expression. MSH2, on the other hand, which is generally thought to play a role in mismatch repair at a step prior to that of MLH1, is not recruited to incoming genomes and appears to act at a later step in the viral life cycle. Silencing of MSH2 appears to inhibit early gene expression. Thus, both MLH1 and MSH2 are required but appear to participate in distinct events in the virus life cycle. The observation that MLH1 plays an earlier role in HSV-1 infection than does MSH2 is surprising and may indicate a novel function for MLH1 distinct from its known MSH2-dependent role in mismatch repair.
Collapse
|
240
|
Liang Q, Deng H, Li X, Wu X, Tang Q, Chang TH, Peng H, Rauscher FJ, Ozato K, Zhu F. Tripartite motif-containing protein 28 is a small ubiquitin-related modifier E3 ligase and negative regulator of IFN regulatory factor 7. THE JOURNAL OF IMMUNOLOGY 2011; 187:4754-63. [PMID: 21940674 DOI: 10.4049/jimmunol.1101704] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IFN regulatory factor 7 (IRF7) is a potent transcription factor of type I IFNs and IFN-stimulated genes and is known as the master regulator of type I IFN-dependent immune responses. Because excessive responses could harm the host, IRF7 itself is delicately regulated at the transcriptional, translational, and posttranslational levels. Modification of IRF7 by small ubiquitin-related modifiers (SUMOs) has been shown to regulate IFN expression and antiviral responses negatively, but the specific E3 ligase needed for IRF7 SUMOylation has remained unknown. As reported in this article, we have identified the tripartite motif-containing protein 28 (TRIM28) as a binding partner of IRF7. We have demonstrated that TRIM28 also interacts with the SUMO E2 enzyme and increases SUMOylation of IRF7 both in vivo and in vitro, suggesting it acts as a SUMO E3 ligase of IRF7. Unlike the common SUMO E3 ligase, protein inhibitor of activated STAT1, the E3 activity of TRIM28 is specific to IRF7, because it has little effect on IRF7's close relative IRF3. TRIM28 is therefore, so far as we know, the first IRF7-specific SUMO E3 reported. TRIM28-mediated SUMOylation of IRF7 is increased during viral infection, and SUMOylation of transcription factors usually results in transcriptional repression. Overexpression of TRIM28 therefore inhibits IRF7 transactivation activity, whereas knockdown of TRIM28 has the opposite effect and potentiates IFN production and antiviral responses. Collectively, our results suggest that TRIM28 is a specific SUMO E3 ligase and negative regulator of IRF7.
Collapse
Affiliation(s)
- Qiming Liang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Boutell C, Cuchet-Lourenço D, Vanni E, Orr A, Glass M, McFarlane S, Everett RD. A viral ubiquitin ligase has substrate preferential SUMO targeted ubiquitin ligase activity that counteracts intrinsic antiviral defence. PLoS Pathog 2011; 7:e1002245. [PMID: 21949651 PMCID: PMC3174244 DOI: 10.1371/journal.ppat.1002245] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 07/27/2011] [Indexed: 12/20/2022] Open
Abstract
Intrinsic antiviral resistance represents the first line of intracellular defence against virus infection. During herpes simplex virus type-1 (HSV-1) infection this response can lead to the repression of viral gene expression but is counteracted by the viral ubiquitin ligase ICP0. Here we address the mechanisms by which ICP0 overcomes this antiviral response. We report that ICP0 induces the widespread proteasome-dependent degradation of SUMO-conjugated proteins during infection and has properties related to those of cellular SUMO-targeted ubiquitin ligases (STUbLs). Mutation of putative SUMO interaction motifs within ICP0 not only affects its ability to degrade SUMO conjugates, but also its capacity to stimulate HSV-1 lytic infection and reactivation from quiescence. We demonstrate that in the absence of this viral countermeasure the SUMO conjugation pathway plays an important role in mediating intrinsic antiviral resistance and the repression of HSV-1 infection. Using PML as a model substrate, we found that whilst ICP0 preferentially targets SUMO-modified isoforms of PML for degradation, it also induces the degradation of PML isoform I in a SUMO modification-independent manner. PML was degraded by ICP0 more rapidly than the bulk of SUMO-modified proteins in general, implying that the identity of a SUMO-modified protein, as well as the presence of SUMO modification, is involved in ICP0 targeting. We conclude that ICP0 has dual targeting mechanisms involving both SUMO- and substrate-dependent targeting specificities in order to counteract intrinsic antiviral resistance to HSV-1 infection. Viruses must evade several antiviral defences in order to establish a productive infection. These include antibody- and cell-mediated acquired immunity and interferon-regulated innate immunity. Recently, a third arm of antiviral defence has been discovered, so called intrinsic immunity. This aspect of antiviral resistance represents the first line of intracellular defence against virus infection and is mediated by pre-existing cellular factors that attempt to repress viral replication during the initial stages of infection. Like acquired and innate immunity, viruses have evolved mechanisms that overcome intrinsic defence. Here we show that in response to herpes simplex virus type-1 (HSV-1) infection an important aspect of intrinsic immunity is regulated by the small ubiquitin-like modifier (SUMO) conjugation pathway. In response to this defence, the virus induces rapid degradation of specific SUMO-conjugated proteins, followed by widespread loss of SUMO-conjugated species in general. Inactivation of the SUMO pathway inhibits the cell’s ability to efficiently repress viral replication in the absence of this viral countermeasure. Our data identifies an important regulatory pathway that mediates intrinsic resistance to HSV-1 infection and describes the biochemical mechanism that the virus utilizes in order to counteract this antiviral defence.
Collapse
Affiliation(s)
- Chris Boutell
- MRC-University of Glasgow Centre for Virus Research (CVR), Glasgow, Scotland, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
242
|
Tavalai N, Adler M, Scherer M, Riedl Y, Stamminger T. Evidence for a dual antiviral role of the major nuclear domain 10 component Sp100 during the immediate-early and late phases of the human cytomegalovirus replication cycle. J Virol 2011; 85:9447-58. [PMID: 21734036 PMCID: PMC3165758 DOI: 10.1128/jvi.00870-11] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 06/27/2011] [Indexed: 12/20/2022] Open
Abstract
In recent studies, the nuclear domain 10 (ND10) components PML and hDaxx were identified as cellular restriction factors that inhibit the initiation of human cytomegalovirus (HCMV) replication. The antiviral function of ND10, however, is antagonized by the IE1 protein, which induces ND10 disruption. Here we show that IE1 not only de-SUMOylates PML immediately upon infection but also directly targets Sp100. IE1 expression alone was sufficient to downregulate endogenous Sp100 independently of the presence of PML. Moreover, cotransfection experiments revealed that IE1 negatively interferes with the SUMOylation of all Sp100 isoforms. The modulation of Sp100 at immediate-early (IE) times of infection, indeed, seemed to have an in vivo relevance for HCMV replication, since knockdown of Sp100 resulted in more cells initiating the viral gene expression program. In addition, we observed that Sp100 was degraded in a proteasome-dependent manner at late times postinfection, suggesting that Sp100 may play an additional antiviral role during the late phase. Infection experiments conducted with Sp100 knockdown human foreskin fibroblasts (HFFs) confirmed this hypothesis: depletion of Sp100 resulted in augmented release of progeny virus particles compared to that from control cells. Consistent with this observation, we noted increased amounts of viral late gene products in the absence of Sp100. Importantly, this elevated late gene expression was not dependent on enhanced viral IE gene expression. Taken together, our data provide evidence that Sp100 is the first ND10-related factor identified that not only possesses the potential to restrict the initial stage of infection but also inhibits HCMV replication during the late phase.
Collapse
Affiliation(s)
- Nina Tavalai
- Institute for Clinical and Molecular Virology, University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Martina Adler
- Institute for Clinical and Molecular Virology, University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Myriam Scherer
- Institute for Clinical and Molecular Virology, University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Yvonne Riedl
- Institute for Clinical and Molecular Virology, University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Thomas Stamminger
- Institute for Clinical and Molecular Virology, University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| |
Collapse
|
243
|
Abstract
Rabies is among the longest known and most dangerous and feared infectious diseases for humans and animals and still is responsible for tenth of thousands of human deaths per year. The rabies virus (RABV) is a rather atypical member of the Rhabdoviridae family as it has completely adapted during evolution to warm-blooded hosts and is directly transmitted between them, whereas most other rhabdoviruses are transmitted by insect vectors. The virus is also unique with respect to its extremely broad host species range and a very narrow host organ range, namely its strict neurotropism. It is becoming increasingly clear that the host innate immune system, particularly the type I interferon system, and the viral counteractions profoundly shape this virus-host relationship. In the past few years, exciting new insight was obtained on how viruses are sensed by innate immune receptors, how the downstream signaling networks for activation of interferon are working, and how viruses can interfere with the system. While RABV 5'-triphosphate RNAs were identified as the major pathogen-associated molecular pattern sensed by cytoplasmic RIG-I-like receptors (RLR), the RABV phosphoprotein (P) has emerged as a potent multifunctional antagonist able to counteract the signaling cascades leading to transcriptional activation of interferon genes as well as interferon signaling pathways, thereby limiting expression of antiviral and immune-stimulatory genes.
Collapse
Affiliation(s)
- Martina Rieder
- Max von Pettenkofer Institute and Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | | |
Collapse
|
244
|
Wang L, Oliver SL, Sommer M, Rajamani J, Reichelt M, Arvin AM. Disruption of PML nuclear bodies is mediated by ORF61 SUMO-interacting motifs and required for varicella-zoster virus pathogenesis in skin. PLoS Pathog 2011; 7:e1002157. [PMID: 21901090 PMCID: PMC3161977 DOI: 10.1371/journal.ppat.1002157] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 05/23/2011] [Indexed: 12/15/2022] Open
Abstract
Promyelocytic leukemia protein (PML) has antiviral functions and many viruses encode gene products that disrupt PML nuclear bodies (PML NBs). However, evidence of the relevance of PML NB modification for viral pathogenesis is limited and little is known about viral gene functions required for PML NB disruption in infected cells in vivo. Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes cutaneous lesions during primary and recurrent infection. Here we show that VZV disrupts PML NBs in infected cells in human skin xenografts in SCID mice and that the disruption is achieved by open reading frame 61 (ORF61) protein via its SUMO-interacting motifs (SIMs). Three conserved SIMs mediated ORF61 binding to SUMO1 and were required for ORF61 association with and disruption of PML NBs. Mutation of the ORF61 SIMs in the VZV genome showed that these motifs were necessary for PML NB dispersal in VZV-infected cells in vitro. In vivo, PML NBs were highly abundant, especially in basal layer cells of uninfected skin, whereas their frequency was significantly decreased in VZV-infected cells. In contrast, mutation of the ORF61 SIMs reduced ORF61 association with PML NBs, most PML NBs remained intact and importantly, viral replication in skin was severely impaired. The ORF61 SIM mutant virus failed to cause the typical VZV lesions that penetrate across the basement membrane into the dermis and viral spread in the epidermis was limited. These experiments indicate that VZV pathogenesis in skin depends upon the ORF61-mediated disruption of PML NBs and that the ORF61 SUMO-binding function is necessary for this effect. More broadly, our study elucidates the importance of PML NBs for the innate control of a viral pathogen during infection of differentiated cells within their tissue microenvironment in vivo and the requirement for a viral protein with SUMO-binding capacity to counteract this intrinsic barrier. PML nuclear bodies (PML NBs) are spherical nuclear structures that are present in most human and animal cells. These bodies contribute to anti-viral defense and therefore many viruses have developed strategies to disrupt them. This interaction has been demonstrated for a number of viruses in cultured cells but little is known about these processes in differentiated cells within human tissues. Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes chicken pox and shingle lesions in skin. Here we show that VZV disrupts PML NBs in epidermal and dermal cells in skin tissues implanted subcutaneously in immunodeficient mice. We found that PML NB dispersal is mediated by VZV ORF61 protein and is required for VZV cell to cell spread and lesion formation in skin. The ability of ORF61 to disrupt PML NBs depends on its capacity to bind to SUMO1 protein, which is conjugated to PML and other proteins within PML NBs. To our knowledge, our study provides the first evidence of PML NB modification through the SUMO-binding function of a viral protein, VZV ORF61, and the importance of this molecular mechanism for virus-induced PML NB disruption in differentiated cells infected within their tissue microenvironment in vivo.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Line, Tumor
- Cell Proliferation
- Cloning, Molecular
- Genes, Viral
- Herpesvirus 3, Human/genetics
- Herpesvirus 3, Human/pathogenicity
- Herpesvirus 3, Human/physiology
- Humans
- Intranuclear Inclusion Bodies/metabolism
- Intranuclear Inclusion Bodies/virology
- Leukemia, Promyelocytic, Acute
- Mice
- Mice, SCID
- Models, Animal
- Mutagenesis
- Plasmids/genetics
- Protein Interaction Domains and Motifs/genetics
- SUMO-1 Protein/genetics
- SUMO-1 Protein/metabolism
- Skin/virology
- Up-Regulation
- Viral Proteins/genetics
- Viral Proteins/metabolism
- Virus Replication
Collapse
Affiliation(s)
- Li Wang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, United States of America.
| | | | | | | | | | | |
Collapse
|
245
|
Abstract
The concept that viral sensing systems, via their ability to drive pro-inflammatory cytokine and interferon production, contribute to the development of autoimmune and autoinflammatory disease is supported by a wide range of clinical and experimental observations. Recently, the tripartite motif-containing proteins (TRIMs) have emerged as having key roles in antiviral immunity - either as viral restriction factors or as regulators of pathways downstream of viral RNA and DNA sensors, and the inflammasome. Given their involvement in these pathways, we propose that TRIM proteins contribute to the development and pathology of autoimmune and autoinflammatory conditions, thus making them potential novel targets for therapeutic manipulation.
Collapse
|
246
|
Everett RD. Study of early events during herpes simplex virus type 1 infection by confocal microscopy. Methods 2011; 55:144-52. [PMID: 21855636 DOI: 10.1016/j.ymeth.2011.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/01/2011] [Indexed: 11/18/2022] Open
Abstract
Laser scanning confocal microscopy is a powerful technique that can be applied to study the localisation and behaviour of proteins and nucleic acids in many experimental situations. It is a particularly useful technique for the study of virus infections because of the changes that occur in the distribution and amounts of both viral and cellular proteins as infection develops. These changes reflect key stages and important regulatory events that govern the efficiency of infection. Using herpes simplex virus type 1 infected cells as an experimental model, this article provides guidance for users new to confocal microscopy on basic principles and techniques. The emphasis is on recognising, diagnosing and avoiding potential artifacts, and the workflow of the production of high quality, technically correct images.
Collapse
Affiliation(s)
- Roger D Everett
- MRC-University of Glasgow, Centre for Virus Research, 8, Church Street, Glasgow G11 5JR, UK.
| |
Collapse
|
247
|
Zydek M, Uecker R, Tavalai N, Stamminger T, Hagemeier C, Wiebusch L. General blockade of human cytomegalovirus immediate-early mRNA expression in the S/G2 phase by a nuclear, Daxx- and PML-independent mechanism. J Gen Virol 2011; 92:2757-2769. [PMID: 21832009 DOI: 10.1099/vir.0.034173-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The onset of human cytomegalovirus (HCMV) lytic replication is strictly controlled by the host cell division cycle. Although viral entry of S/G2-phase cells is unperturbed expression of major immediate-early (MIE) genes IE1 and IE2 is tightly blocked in these cells. Besides the finding that cyclin-dependent kinase (CDK) activity is required for IE1/IE2 repression little is known about the nature of this cell cycle-dependent block. Here, we show that the block occurs after nuclear entry of viral DNA and prevents the accumulation of IE1/IE2 mRNAs, suggesting an inhibition of transcription. Remarkably, the presence of cis-regulatory regions of the MIE locus is neither sufficient nor necessary for IE1/IE2 repression in the S/G2 phase. Furthermore, the block of viral mRNA expression also affects other immediate-early transcribed regions, i.e. the US3 and UL36-38 gene loci. This suggests a mechanism of repression that acts in a general and not a gene-specific fashion. Such a nuclear, genome-wide repression of HCMV is typically mediated by the intrinsic immune defence at nuclear domain 10 (ND10) structures. However, we found that neither Daxx nor PML, the main players of ND10-based immunity, are required for the block to viral gene expression in the S/G2 phase. In addition, the viral tegument protein pp71 (pUL82), a major antagonist of the intrinsic immunity at pre-immediate-early times of infection, proved to be functional in S-phase cells. This suggests the existence of a yet undiscovered, CDK-dependent mechanism exerting higher-level control over immediate-early mRNA expression in HCMV-infected cells.
Collapse
Affiliation(s)
- Martin Zydek
- Labor für Pädiatrische Molekularbiologie, Charité-Universitätsmedizin Berlin, Ziegelstr. 5-9, 10117 Berlin, Germany
| | - Ralf Uecker
- Labor für Pädiatrische Molekularbiologie, Charité-Universitätsmedizin Berlin, Ziegelstr. 5-9, 10117 Berlin, Germany
| | - Nina Tavalai
- Institut für Klinische und Molekulare Virologie der Universität Erlangen-Nürnberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Thomas Stamminger
- Institut für Klinische und Molekulare Virologie der Universität Erlangen-Nürnberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Christian Hagemeier
- Labor für Pädiatrische Molekularbiologie, Charité-Universitätsmedizin Berlin, Ziegelstr. 5-9, 10117 Berlin, Germany
| | - Lüder Wiebusch
- Labor für Pädiatrische Molekularbiologie, Charité-Universitätsmedizin Berlin, Ziegelstr. 5-9, 10117 Berlin, Germany
| |
Collapse
|
248
|
Gustin JK, Moses AV, Früh K, Douglas JL. Viral takeover of the host ubiquitin system. Front Microbiol 2011; 2:161. [PMID: 21847386 PMCID: PMC3147166 DOI: 10.3389/fmicb.2011.00161] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 07/14/2011] [Indexed: 01/29/2023] Open
Abstract
Like the other more well-characterized post-translational modifications (phosphorylation, methylation, acetylation, acylation, etc.), the attachment of the 76 amino acid ubiquitin (Ub) protein to substrates has been shown to govern countless cellular processes. As obligate intracellular parasites, viruses have evolved the capability to commandeer many host processes in order to maximize their own survival, whether it be to increase viral production or to ensure the long-term survival of latently infected host cells. The first evidence that viruses could usurp the Ub system came from the DNA tumor viruses and Adenoviruses, each of which use Ub to dysregulate the host cell cycle (Scheffner et al., 1990; Querido et al., 2001). Today, the list of viruses that utilize Ub includes members from almost every viral class, encompassing both RNA and DNA viruses. Among these, there are examples of Ub usage at every stage of the viral life cycle, involving both ubiquitination and de-ubiquitination. In addition to viruses that merely modify the host Ub system, many of the large DNA viruses encode their own Ub modifying machinery. In this review, we highlight the latest discoveries regarding the myriad ways that viruses utilize Ub to their advantage.
Collapse
Affiliation(s)
- Jean K Gustin
- Vaccine and Gene Therapy Institute, Oregon Health & Science University Beaverton, OR, USA
| | | | | | | |
Collapse
|
249
|
Cuchet-Lourenço D, Boutell C, Lukashchuk V, Grant K, Sykes A, Murray J, Orr A, Everett RD. SUMO pathway dependent recruitment of cellular repressors to herpes simplex virus type 1 genomes. PLoS Pathog 2011; 7:e1002123. [PMID: 21779164 PMCID: PMC3136452 DOI: 10.1371/journal.ppat.1002123] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 05/03/2011] [Indexed: 12/12/2022] Open
Abstract
Components of promyelocytic leukaemia (PML) nuclear bodies (ND10) are recruited to sites associated with herpes simplex virus type 1 (HSV-1) genomes soon after they enter the nucleus. This cellular response is linked to intrinsic antiviral resistance and is counteracted by viral regulatory protein ICP0. We report that the SUMO interaction motifs of PML, Sp100 and hDaxx are required for recruitment of these repressive proteins to HSV-1 induced foci, which also contain SUMO conjugates and PIAS2β, a SUMO E3 ligase. SUMO modification of PML and elements of its tripartite motif (TRIM) are also required for recruitment in cells lacking endogenous PML. Mutants of PML isoform I and hDaxx that are not recruited to virus induced foci are unable to reproduce the repression of ICP0 null mutant HSV-1 infection mediated by their wild type counterparts. We conclude that recruitment of ND10 components to sites associated with HSV-1 genomes reflects a cellular defence against invading pathogen DNA that is regulated through the SUMO modification pathway.
Collapse
Affiliation(s)
| | - Chris Boutell
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Vera Lukashchuk
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Kyle Grant
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Amanda Sykes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Jill Murray
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Anne Orr
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Roger D. Everett
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| |
Collapse
|
250
|
Kobiler O, Lipman Y, Therkelsen K, Daubechies I, Enquist LW. Herpesviruses carrying a Brainbow cassette reveal replication and expression of limited numbers of incoming genomes. Nat Commun 2011; 1:146. [PMID: 21266996 PMCID: PMC3079281 DOI: 10.1038/ncomms1145] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 11/24/2010] [Indexed: 01/12/2023] Open
Abstract
Whether all the infectious herpesvirus particles entering a cell are able to replicate and/or express their genomes is not known. Here, we developed a general method to determine the number of viral genomes expressed in an infected cell. We constructed and analysed fluorophore expression from a recombinant pseudorabies virus (PRV263) carrying a Brainbow cassette (Cre-conditional expression of different fluorophores). Using three isogenic strains derived from PRV263, each expressing a single fluorophore, we analysed the colour composition of cells infected with these three viruses at different multiplicities. We estimate that fewer than seven incoming genomes are expressed per cell. In addition, those templates that are expressed are the genomes selected for replication and packaging into virions. This finite limit on the number of viral genomes that can be expressed is an intrinsic property of the infected cell and may be influenced by viral and cellular factors. The replication of viral genomes in infected cells is required for successful infection. In this study, using Cre-conditional expression of multiple coloured fluorophores, the authors demonstrate that the number of viral genomes expressed and replicated in a cell is surprisingly limited.
Collapse
Affiliation(s)
- Oren Kobiler
- Department of Molecular Biology and the Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544, USA
| | | | | | | | | |
Collapse
|