Nucleocytoplasmic Shuttling of Bovine Herpesvirus 1 UL47 Protein in Infected Cells

Similar documents
Nuclear export of VP19C is not essential for replication of herpes simplex virus type 1

Live-Cell Analysis of a Green Fluorescent Protein-Tagged Herpes Simplex Virus Infection

Activation of Gene Expression by Human Herpes Virus 6

Herpesviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Materials and Methods , The two-hybrid principle.

The Herpes Simplex Virus Immediate-Early Protein ICP27 Shuttles between Nucleus and Cytoplasm

The First Identified Nucleocytoplasmic Shuttling Herpesviral Capsid Protein: Herpes Simplex Virus Type 1 VP19C

Conditional and reversible disruption of essential herpesvirus protein functions

Herpes Simplex Virus 1 Protein Kinase Us3 and Major Tegument Protein UL47 Reciprocally Regulate Their Subcellular Localization in Infected Cells

Supplemental information contains 7 movies and 4 supplemental Figures

Structural vs. nonstructural proteins

Supplementary Figure 1. SC35M polymerase activity in the presence of Bat or SC35M NP encoded from the phw2000 rescue plasmid.

Supplementary information. MARCH8 inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins

SUPPLEMENTARY INFORMATION

Under the Radar Screen: How Bugs Trick Our Immune Defenses

The Attenuated Pseudorabies Virus Strain Bartha Fails To Package the Tegument Proteins Us3 and VP22

NOTES. Matthew S. Walters, Christos A. Kyratsous, and Saul J. Silverstein*

Sarah A. Kopecky and Douglas S. Lyles*

Herpesvirus mrnas Are Sorted for Export via Crm1- Dependent and -Independent Pathways

Intracellular Trafficking and Localization of the Pseudorabies Virus Us9 Type II Envelope Protein to Host and Viral Membranes

Received 12 June 2002/Accepted 13 September 2002

Inner tegument protein pul37 of herpes simplex virus type 1 is involved in directing capsids to the trans-golgi network for envelopment

Polyomaviridae. Spring

in Cell Culture and Lack of Virulence in Cattle

Herpes Simplex Virus ICP27 Induces Cytoplasmic Accumulation of Unspliced Polyadenylated -Globin Pre-mRNA in Infected HeLa Cells

viruses ISSN

T H E J O U R N A L O F C E L L B I O L O G Y

HSV-1 gm and the gk/pul20 Complex Are Important for the Localization of gd and gh/l to Viral Assembly Sites

Certificate of Analysis

MOLECULAR CELL BIOLOGY

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

T H E J O U R N A L O F C E L L B I O L O G Y

Essential Function of the Pseudorabies Virus UL36 Gene Product Is Independent of Its Interaction with the UL37 Protein

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

Supplementary Fig. S1. Schematic diagram of minigenome segments.

Pseudorabies Virus UL37 Gene Product Is Involved in Secondary Envelopment

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR

Problem Set 5 KEY

Determination of the temporal pattern and importance of BALF1 expression in Epstein-Barr viral infection

Rabbit Polyclonal antibody to NFkB p65 (v-rel reticuloendotheliosis viral oncogene homolog A (avian))

Directional Transneuronal Infection by Pseudorabies Virus Is Dependent on an Acidic Internalization Motif in the Us9 Cytoplasmic Tail

Shin-Hee Kim, Yongqi Yan, and Siba K. Samal*

ICP27 Selectively Regulates the Cytoplasmic Localization of a Subset of Viral Transcripts in Herpes Simplex Virus Type 1-Infected Cells

Overview of virus life cycle

Regulation of a nuclear export signal by an adjacent inhibitory sequence: The effector domain of the influenza virus NS1 protein

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells

T H E J O U R N A L O F C E L L B I O L O G Y

Plasmid-Driven Formation of Influenza Virus-Like Particles

Muscular Dystrophy. Biol 405 Molecular Medicine

Cyclin-Dependent Kinase-Like Function Is Shared by the Beta- and Gamma- Subset of the Conserved Herpesvirus Protein Kinases

Role of the VP16-Binding Domain of vhs in Viral Growth, Host Shutoff Activity, and Pathogenesis

Reviewers' comments: Reviewer #1 (Remarks to the Author):

The functional investigation of the interaction between TATA-associated factor 3 (TAF3) and p53 protein

Genomic Alterations Associated with Persistent Infections by Equine Infectious Anaemia Virus, a Retrovirus

Zool 3200: Cell Biology Exam 4 Part II 2/3/15

Certificate of Analysis

Section 6. Junaid Malek, M.D.

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors

Identification of Mutation(s) in. Associated with Neutralization Resistance. Miah Blomquist

Innate Immunity & Inflammation

Recombinant Protein Expression Retroviral system

7.012 Quiz 3 Answers

October 26, Lecture Readings. Vesicular Trafficking, Secretory Pathway, HIV Assembly and Exit from Cell

Mike Reichelt,* Jennifer Brady, and Ann M. Arvin

Auxin-Inducible Degron (AID) System Total Set

Advances in gene encoding proteins of human herpesvirus 6

33VASTVNGATSANNHGEPPS51PADARPR58

Viral Genetics. BIT 220 Chapter 16

Nature Medicine: doi: /nm.4322

Herpes Simplex Virus Type 1 and Bovine Herpesvirus 1 Latency

Mechanisms of alternative splicing regulation

Nature Structural & Molecular Biology: doi: /nsmb Supplementary Figure 1. Generation and validation of mtef4-knockout mice.

MCB130 Midterm. GSI s Name:

Supplementary Figure 1. Prevalence of U539C and G540A nucleotide and E172K amino acid substitutions among H9N2 viruses. Full-length H9N2 NS

Fig. S1. Subcellular localization of overexpressed LPP3wt-GFP in COS-7 and HeLa cells. Cos7 (top) and HeLa (bottom) cells expressing for 24 h human

Size nm m m

The Amino Terminus of the Herpes Simplex Virus 1 Protein Vhs Mediates Membrane Association and Tegument Incorporation

Stability determinants of Murine Cytomegalovirus long non-coding RNA7.2

GFP-LC3 +/+ CLU -/- kda CLU GFP. Actin. GFP-LC3 +/+ CLU -/- kda CLU GFP. Actin

~Lentivirus production~

04_polarity. The formation of synaptic vesicles

Week 5 Section. Junaid Malek, M.D.

SUPPLEMENTARY FIGURES

Identification of Functional Domains in Herpes Simplex Virus 2 Glycoprotein B

Interaction of the Influenza Virus Nucleoprotein with the Cellular CRM1-Mediated Nuclear Export Pathway

Animal hosts Natural host Laboratory animals Rabbits Mice Rats Hamsters Newborn or suckling rodents Animal models for viral pathogenesis 4 Growth of v

Role of ESCRT-I in Retroviral Budding

A glycoprotein M-deleted equid herpesvirus 4 is severely impaired in virus egress and cell-to-cell spread

VP22 of Herpes Simplex Virus 1 Promotes. and Accumulation of a Subset of Viral mrnas at Early Times in Infection

Variation in the HindlII Restriction Fragments of DNA from the Chinese Tian Tan Strain of Vaccinia Virus

Influenza virus exploits tunneling nanotubes for cell-to-cell spread

Structure of viruses

Supplementary Figure 1. AdipoR1 silencing and overexpression controls. (a) Representative blots (upper and lower panels) showing the AdipoR1 protein

Human Cytomegalovirus (HCMV) Immediate early proteins, gene expression and signaling

UL20 Protein Functions Precede and Are Required for the UL11 Functions of Herpes Simplex Virus Type 1 Cytoplasmic Virion Envelopment

Life Sciences 1A Midterm Exam 2. November 13, 2006

Tunneling nanotubes (TNT) as a novel route of cell-to-cell spread of herpesviruses

Received 22 November 1996/Accepted 3 April 1997

Supplementary Figure S1 Supplementary Figure S2

VIRUSES. 1. Describe the structure of a virus by completing the following chart.

Transcription:

JOURNAL OF VIROLOGY, Jan. 2006, p. 1059 1063 Vol. 80, No. 2 0022-538X/06/$08.00 0 doi:10.1128/jvi.80.2.1059 1063.2006 Copyright 2006, American Society for Microbiology. All Rights Reserved. Nucleocytoplasmic Shuttling of Bovine Herpesvirus 1 UL47 Protein in Infected Cells Janneke Verhagen, Ian Hutchinson, and Gillian Elliott* Virus Assembly Group, Marie Curie Research Institute, Oxted, Surrey, United Kingdom Received 9 September 2005/Accepted 27 October 2005 Previous studies with transfected cells have shown that the herpes simplex virus type 1 (HSV-1) and bovine herpesvirus 1 (BHV-1) UL47 proteins shuttle between the nucleus and the cytoplasm. HSV-1 UL47 has also been shown to bind RNA. Here we examine the BHV-1 UL47 protein in infected cells using a green fluorescent protein-ul47-expressing virus. We show that UL47 is detected in the nucleus early in infection. We use fluorescence loss in photobleaching to show that nuclear UL47 undergoes rapid nucleocytoplasmic shuttling. Furthermore, we demonstrate that actinomycin D inhibits the reaccumulation of UL47 in the nuclei of infected cells. These results suggest that UL47 exhibits behavior similar to that of previously characterized RNAtransporting proteins. The alphaherpesvirus UL47 gene encodes a protein that is assembled into the tegument of the virion (1, 7, 10, 23). While the role of UL47 has not yet been defined, its deletion from herpes simplex virus type 1 (HSV-1), pseudorabies virus, or Marek s disease virus results in viruses that produce small plaques and replicate more slowly than wild-type (Wt) viruses (4, 7, 24, 25). We have previously studied the HSV-1 UL47- encoded protein VP13/14 expressed in infected cells and by transient transfection. In both cases, VP13/14 was targeted to the nucleus, and we have identified an arginine-rich nuclear localization signal in its N terminus (2, 3). Furthermore, we have shown that VP13/14 has the ability to shuttle between the nucleus and the cytoplasm when expressed alone (3). The bovine herpesvirus 1 (BHV-1) homologue of VP13/14, the UL47-encoded protein VP8, is by far the major structural protein of the BHV-1 virus particle (1), and as yet there has been no report of a UL47 knockout virus for BHV-1. Immunofluorescence studies of infected cells have shown VP8 in the nucleus (22), and a recent study demonstrated that as for VP13/14, VP8 appears to shuttle between the nucleus and the cytoplasm when expressed in isolation (26). Here we have examined the trafficking of the BHV-1 UL47 protein in infected cells by constructing a recombinant virus expressing green fluorescent protein (GFP)-tagged UL47. Infectious BHV-1 genomic DNA (strain P8-2) was cotransfected into COS-1 cells with plasmid pjv46, which consists of the GFP open reading frame flanked by 1 kb of the UL47 upstream sequence and the entire UL47 open reading frame. Green fluorescent plaques were purified three times on MDBK cells, resulting in the recombinant virus jv46v (Fig. 1A). The growth characteristics of jv46v were established by conducting a time course of infection with MDBK cells. Total cell lysates were analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and Western blotting with a polyclonal anti-bhv-1 UL47 antibody, indicating that the UL47- * Corresponding author. Mailing address: Virus Assembly Group, Marie Curie Research Institute, Oxted, Surrey RH8 OTL, United Kingdom. Phone: 44 1883 722306. Fax: 44 1883 714375. E-mail: g.elliott@mcri.ac.uk. reactive species in jv46v-infected cells was around 30 kda larger than Wt UL47 (Fig. 1B). This species also reacted with a monoclonal GFP antibody (Clontech), confirming that it represented a novel GFP-UL47 fusion protein (Fig. 1B). Importantly, the rates of expression of UL47 in the two viruses were similar, as were the expression kinetics of the btif protein encoded by UL48 (Fig. 1B). One-step growth curves for intracellular and extracellular virus also indicated that the introduction of GFP at the N terminus of UL47 had no effect on virus replication (Fig. 1C). Importantly, the recombinant virus also exhibited the same plaque size and the same replication rate under low-multiplicity conditions as the Wt virus (data not shown). We next addressed the assembly of GFP-UL47 into extracellular virions that had been purified from 5 10 8 infected MDBK cells. Wt and jv46v virions were isolated by centrifugation through a 5 to 15%Ficoll gradient and were analyzed by SDS-PAGE followed by Coomassie blue staining or Western blotting. The high level of UL47 is obvious from the protein profile of Wt BHV-1 virions (Fig. 2A). Furthermore, the profile of jv46v BHV-1 virions shows that the 105-kDa VP8 protein has been replaced by a larger species of around 135 kda (Fig. 2A). Western blotting with anti-ul47 and anti-gfp antibodies revealed that this novel component was GFP-UL47 and that it was packaged into the virus particle in amounts equivalent to those of native UL47 (Fig. 2B). To determine if we could establish the fate of virion UL47 at virus entry, MDBK cells were infected with jv46v in the presence of cycloheximide (CHX) (100 g/ml) and examined live 1 h later. GFP-UL47 was clearly localized in the nucleus of these cells, demonstrating that incoming UL47 was imported into the infected cell nucleus (Fig. 2C). Newly synthesized GFP-UL47 was also detected in the nucleus of infected cells as early as 4 h after infection of MDBK cells in the absence of CHX and increased in intensity until 6 h, when cytoplasmic GFP-UL47 also began to accumulate (Fig. 2D). Over the next 8 h, GFP- UL47 became increasingly cytoplasmic with little detectable nuclear fluorescence (Fig. 2D). The localization of UL47 as determined by the GFP-UL47-expressing virus was similar to that described previously in immunofluorescence studies (22). Downloaded from http://jvi.asm.org/ 1059

Downloaded from http://jvi.asm.org/ FIG. 2. Characterization of BHV-1 expressing GFP-UL47. (A) Equivalent amounts of extracellular virions purified from Wt- and jv46vinfected MDBK cells were analyzed by SDS-PAGE followed by Coomassie blue staining. (B) The same virions were analyzed by Western blotting with antibodies against UL47, GFP, and UL48. (C) MDBK cells grown in a coverslip chamber were infected with jv46v at a multiplicity of 10 in the presence of 100 g/ml cycloheximide. One hour later, the cells were imaged on a Zeiss LSM410 confocal microscope. (D) MDBK cells grown in a coverslip chamber were infected with jv46v at a multiplicity of 2. At various times after infection, representative images were collected using a Zeiss LSM410 confocal microscope. FIG. 1. Insertion of GFP at the N terminus of UL47 has no effect on BHV-1 replication. (A) To construct a plasmid expressing GFP- UL47, the UL47 open reading frame was first transferred as a BamHI/ XhoI fragment from plasmid pcvp8, in which the BamHI site is located seven bases upstream of the initiation codon (kindly provided by Vikram Misra, University of Saskatchewan) into pcdnai/amp. VP8 was then inserted into pegfpc1 (Clontech) as a BamHI/XbaI fragment to create pjv2. Finally, 1 kb upstream of the VP8 gene was amplified by PCR from the BHV-1 genome (strain P8-2) as an AseI/ AgeI fragment and inserted into AseI/AgeI-digested pjv2, resulting in plasmid pjv46 comprising GFP flanked by VP8 upstream sequences at the 5 end and the VP8 gene at the 3 end. Cotransfections were then carried out between pjv46 and infectious BHV-1 DNA, and resulting green fluorescent plaques were purified to generate recombinant virus jv46v. (B) MDBK cells infected with either wild-type BHV-1 (Wt) or the GFP-UL47-expressing virus (jv46v) at a multiplicity of 10 were harvested every 4 h up to 24 h after infection. Equal amounts of total cell extracts were analyzed by Western blotting with antibodies against UL47, GFP (Clontech), and UL48. (C) MDBK cells were infected with Wt or jv46v viruses at a multiplicity of 10 and were harvested every 4 h for both intracellular virus (IC) and extracellular virus (EC). Each sample was titrated on MDBK cells. 1060

VOL. 80, 2006 NOTES 1061 Downloaded from http://jvi.asm.org/ FIG. 3. UL47 shuttles between the nucleus and the cytoplasm in infected cells. (A) MDBK cells grown in a coverslip chamber were infected with jv46v at a multiplicity of 10. At 4 h, the cells were examined using a Deltavision RT imaging system and individual cells chosen for FLIP analysis. The laser module was then used to carry out sequential photobleaching events of the area denoted by the white oval. This area was exposed to the laser for 1 s every 10 s over a period of 7 min. An image of the field was acquired after each bleaching event to determine the loss of fluorescence in the nucleus of the cell. Arrows denote the control cell. (B) The relative fluorescences in the nuclei of the bleached and the unbleached cells in A were quantitated using NIH-image software and plotted against time. (C) MDBK cells infected with HSV-1 expressing GFP-NLS-VP22 in place of Wt VP22 were subjected to FLIP analysis as described for A. Furthermore, our own comparative immunofluorescence studies between Wt- and jv46v-infected cells using the anti-ul47 antibody confirmed that the addition of GFP to the N terminus of UL47 had no effect on the localization of UL47 (data not shown). To address the possibility that UL47 may shuttle during virus infection, we employed fluorescence loss in photobleaching (FLIP) technology (6, 8, 17, 21). Four hours after infection, a small area of cytoplasm in jv46-infected MDBK cells was photobleached for1satintervals of 10 s using the laser module on a Deltavision imaging system. In the example shown, which is one of 10 analyses carried out, images of the cell were acquired after each bleach event, resulting in a time-lapse animation that shows a rapid loss of nuclear fluorescence in the bleached cell compared to the control cell in the same image (Fig. 3A and B). Furthermore, quantification of nuclear GFP-UL47 fluorescence showed that it was reduced to almost background in 7 min (Fig. 3B), suggesting that UL47 export from the nucleus was an extremely rapid event. To confirm that this photobleaching effect was truly indicative of UL47 shuttling, we used a control virus expressing a GFP-tagged protein that is also predominantly nuclear and subjected it to FLIP analysis as described above. For this purpose, we used recombinant HSV-1 that expresses GFP-UL49 (VP22), as described previously (5), but which also has the simian virus 40 nuclear localization signal (NLS) inserted at the N terminus of VP22 (GFP-NLS-VP22). As shown in Fig. 3C (0 s), this results in a GFP-VP22 fusion protein that is predominantly nuclear 4 h

1062 NOTES J. VIROL. FIG. 4. Actinomycin D treatment inhibits the accumulation of UL47 in the nuclei of infected cells. (A and B) Hep2 cells grown on coverslips were exposed to either 20 g/ml CHX alone ( Act D) or 20 g/ml CHX and 5 g/ml Act D ( Act D) for 3 h prior to fixation with 100% methanol. Immunofluorescence was then carried out with antibodies against either hnrnpc1 (A) or hnrnpa1 (B). (C) MDBK cells grown in a coverslip chamber were infected with jv46v at a multiplicity of 2. Four hours later, the cells were exposed to either 20 g/ml CHX alone ( Act D) or 20 g/ml CHX and 5 g/ml Act D ( Act D). After 1 h, representative images of live cells were acquired using a Zeiss LSM 410 confocal microscope. after infection, rather than the cytoplasmic localization previously observed for Wt GFP-VP22 (5). Importantly, photobleaching of the cytoplasm of a cell containing nuclear GFP- NLS-VP22 conducted in the same way as for GFP-UL47 had no effect on the level of fluorescence present in the nucleus, thus confirming that the effect we observe with GFP-UL47 is specifically due to its shuttling between the nucleus and cytoplasm. (Fig. 3C). To further investigate UL47 shuttling, we used the drug actinomycin D (Act D), which affects the shuttling dynamics of many nucleocytoplasmic trafficking proteins and causes the accumulation of such proteins in the cytoplasm rather than the nucleus (12 15). We first tested the experimental conditions for this assay using two endogenous cellular proteins, namely, the known shuttling protein hnrnpa1 and hnrnpc1, a nuclear protein that does not shuttle (14 16). Hep2 cells grown on coverslips were treated with CHX alone or CHX and 5 g/ml Act D, fixed 3 h later in methanol, and processed for immunofluorescence with antibodies against hnrnpa1 and hnrnpc1. As expected, the addition of Act D had no effect on the nuclear localization of hnrnpc1 (Fig. 4A). By contrast, Act D altered the steady-state localization of hnrnpa1 from entirely nuclear to nuclear and cytoplasmic distributions (Fig. 4B), confirming the efficacy of Act D. MDBK cells infected 4 h previously with jv46v were treated in the same way and examined after 1 h. As for hnrnpa1, there was a clear shift of UL47 into the cytoplasm of cells treated with Act D, suggesting that UL47 shuttling was impaired in the presence of Act D (Fig. 4C). In this study we have provided the first evidence of shuttling by any alphaherpesvirus UL47 gene product during virus infection. This confirms the relevance of previous studies carried out on the nucleocytoplasmic shuttling of UL47-encoded proteins (3, 26). Although the exact role of UL47 shuttling has yet to be determined, it has been reported previously that HSV-1 UL47 binds RNA (19). Here we demonstrate that Act D, a drug that inhibits RNA transcription by RNA polymerase II and is known to inhibit the nuclear reaccumulation of exported RNA binding proteins (15, 16), also alters the localization of UL47. This suggests that UL47 shuttling may be linked in some way to RNA biogenesis. The essential HSV-1 immediate-early protein ICP27 is an RNA binding protein that has already been proposed, among other things, to be involved in the nuclear export of viral RNAs (11, 13, 18, 20). Nonetheless, viruses expressing ICP27 mutated in its RNA binding domain or its nuclear export signal can still replicate, suggesting that this function of ICP27 is not absolutely crucial to the virus (9). It is thus tempting to speculate that UL47 may also function in the export of at least some viral RNAs and that this function may in part compensate for the absence of ICP27-directed RNA export. Furthermore, because UL47 is delivered to the nucleus immediately upon infection, it would be available to function in the transport of the immediate-early viral transcripts. The shift of BHV-1 UL47 steady-state localization from the nucleus to the cytoplasm at later times in infection may simply reflect the high levels of viral RNAs that are being synthesized at this time. However, it may also be indicative of the active recruitment and retention of UL47 in cytoplasmic sites of virus assembly. We thank Vikram Misra, University of Saskatchewan, for BHV-1 strain P8-2, plasmid pcvp8, and polyclonal UL47 and UL48 antibodies. We also thank G. Dreyfuss for antibodies against hnrnpa1 and hnrnpc1 and Michelle Donnelly for useful discussion. This work was funded by Marie Curie Cancer Care. REFERENCES 1. Carpenter, D. E., and V. Misra. 1991. The most abundant protein in bovine herpes 1 virions is a homologue of herpes simplex virus type 1 UL47. J Gen. Virol. 72:3077 3084. 2. Donnelly, M., and G. Elliott. 2001. Fluorescent tagging of herpes simplex virus tegument protein VP13/14 in virus infection. J. Virol. 75:2575 2583. 3. Donnelly, M., and G. Elliott. 2001. Nuclear localization and shuttling of herpes simplex virus tegument protein VP13/14. J. Virol. 75:2566 2574. 4. Dorange, F., B. K. Tischer, J. F. Vautherot, and N. Osterrieder. 2002. Characterization of Marek s disease virus serotype 1 (MDV-1) deletion mutants that lack UL46 to UL49 genes: MDV-1 UL49, encoding VP22, is indispensable for virus growth. J. Virol. 76:1959 1970. Downloaded from http://jvi.asm.org/

VOL. 80, 2006 NOTES 1063 5. Elliott, G., and P. O Hare. 1999. Live-cell analysis of a green fluorescent protein-tagged herpes simplex virus infection. J. Virol. 73:4110 4119. 6. Griffis, E. R., N. Altan, J. Lippincott-Schwartz, and M. A. Powers. 2002. Nup98 is a mobile nucleoporin with transcription-dependent dynamics. Mol. Biol. Cell. 13:1282 1297. 7. Kopp, M., B. G. Klupp, H. Granzow, W. Fuchs, and T. C. Mettenleiter. 2002. Identification and characterization of the pseudorabies virus tegument proteins UL46 and UL47: role for UL47 in virion morphogenesis in the cytoplasm. J. Virol. 76:8820 8833. 8. Koster, M., T. Frahm, and H. Hauser. 2005. Nucleocytoplasmic shuttling revealed by FRAP and FLIP technologies. Curr. Opin. Biotechnol. 16:28 34. 9. Lengyel, J., C. Guy, V. Leong, S. Borge, and S. A. Rice. 2002. Mapping of functional regions in the amino-terminal portion of the herpes simplex virus ICP27 regulatory protein: importance of the leucine-rich nuclear export signal and RGG Box RNA-binding domain. J. Virol. 76:11866 11879. 10. McLean, G., F. Rixon, N. Langeland, L. Haarr, and H. Marsden. 1990. Identification and characterization of the virion protein products of herpes simplex virus type 1 gene UL47. J. Gen. Virol. 71:2953 2960. 11. Mears, W. E., and S. A. Rice. 1998. The herpes simplex virus immediate-early protein ICP27 shuttles between nucleus and cytoplasm. Virology 242:128 137. 12. Meyer, B. E., and M. H. Malim. 1994. The HIV-1 Rev trans-activator shuttles between the nucleus and the cytoplasm. Genes Dev. 8:1538 1547. 13. Phelan, A., and J. B. Clements. 1997. Herpes simplex virus type 1 immediate early protein IE63 shuttles between nuclear compartments and the cytoplasm. J Gen. Virol. 78:3327 3331. 14. Pinol-Roma, S., and G. Dreyfuss. 1993. hnrnp proteins: localization and transport between the nucleus and the cytoplasm. Trends Cell Biol. 3:151 155. 15. Pinol-Roma, S., and G. Dreyfuss. 1992. Shuttling of pre-mrna binding proteins between nucleus and cytoplasm. Nature 355:730 732. 16. Pinol-Roma, S., and G. Dreyfuss. 1991. Transcription-dependent and transcription-independent nuclear transport of hnrnp proteins. Science 253: 312 314. 17. Rosin-Arbesfeld, R., A. Cliffe, T. Brabletz, and M. Bienz. 2003. Nuclear export of the APC tumour suppressor controls beta-catenin function in transcription. EMBO J. 22:1101 1113. 18. Sandri-Goldin, R. M. 1998. ICP27 mediates HSV RNA export by shuttling through a leucine-rich nuclear export signal and binding viral intronless RNAs through an RGG motif. Genes Dev. 12:868 879. 19. Sciortino, M. T., B. Taddeo, A. P. Poon, A. Mastino, and B. Roizman. 2002. Of the three tegument proteins that package mrna in herpes simplex virions, one (VP22) transports the mrna to uninfected cells for expression prior to viral infection. Proc. Natl. Acad. Sci. USA 99:8318 8323. 20. Soliman, T. M., R. M. Sandri-Goldin, and S. J. Silverstein. 1997. Shuttling of the herpes simplex virus type 1 regulatory protein ICP27 between the nucleus and cytoplasm mediates the expression of late proteins. J. Virol. 71:9188 9197. 21. van Drogen, F., and M. Peter. 2001. MAP kinase dynamics in yeast. Biol. Cell. 93:63 70. 22. van Drunen Littel-van den Hurk, S., S. Garzon, J. V. van den Hurk, L. A. Babiuk, and P. Tijssen. 1995. The role of the major tegument protein VP8 of bovine herpesvirus-1 in infection and immunity. Virology 206:413 425. 23. Whittaker, G. R., M. P. Riggio, I. W. Halliburton, R. A. Killington, G. P. Allen, and D. M. Meredith. 1991. Antigenic and protein sequence homology between VP13/14, a herpes simplex virus type 1 tegument protein, and gp10, a glycoprotein of equine herpesvirus 1 and 4. J. Virol. 65:2320 2326. 24. Zhang, Y., and J. L. McKnight. 1993. Herpes simplex virus type 1 UL46 and UL47 deletion mutants lack VP11 and VP12 or VP13 and VP14, respectively, and exhibit altered viral thymidine kinase expression. J. Virol. 67: 1482 1492. 25. Zhang, Y., D. A. Sirko, and J. L. McKnight. 1991. Role of herpes simplex virus type 1 UL46 and UL47 in alpha TIF-mediated transcriptional induction: characterization of three viral deletion mutants. J. Virol. 65:829 841. 26. Zheng, C., R. Brownlie, L. A. Babiuk, and S. van Drunen Littel-van den Hurk. 2004. Characterization of nuclear localization and export signals of the major tegument protein VP8 of bovine herpesvirus-1. Virology 324:327 339. Downloaded from http://jvi.asm.org/