Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Journal of Virology
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Genome Replication and Regulation of Viral Gene Expression | Spotlight

Involvement of SSRP1 in Latent Replication of Kaposi's Sarcoma-Associated Herpesvirus

Jianhong Hu, Eugene Liu, Rolf Renne
Jianhong Hu
Department of Molecular Genetics and Microbiology and UF Shands Cancer Center, University of Florida, Gainesville, Florida 32610
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Eugene Liu
Department of Molecular Genetics and Microbiology and UF Shands Cancer Center, University of Florida, Gainesville, Florida 32610
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rolf Renne
Department of Molecular Genetics and Microbiology and UF Shands Cancer Center, University of Florida, Gainesville, Florida 32610
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: rrenne@ufl.edu
DOI: 10.1128/JVI.00907-09
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Article Figures & Data

Figures

  • Tables
  • FIG. 1.
    • Open in new tab
    • Download powerpoint
    FIG. 1.

    The first 8 nt within the RE are pivotal for KSHV latent origin function. Replication assays were performed as previously described. Briefly, 8 μg of each origin-containing construct was cotransfected with 2 μg of pcDNA3/orf73 or carrier DNA into 293 cells. Episomal DNA was recovered by Hirt extraction 72 h posttransfection. Ten percent of the episomal DNA was digested with HindIII (input), while 90% was double digested with HindIII and DpnI for 16 h (DpnI digest). DNA were electrophoretically separated in an agarose gel, immobilized on nylon membranes, and hybridized with radiolabeled probes using pCRII-TR as the template. The replication efficiency of each plasmid was calculated by comparing the intensity of the replicated DNA band with that of its input from independent experiments. The replication activity of each mutant was compared with that of the wild-type MR, which was set at 100%. (A) The MR replicates with efficiency similar to that of the full-length TR. (B) Sequences of the wild-type RE and converted sequences within RE mutants, respectively. (C) The first 8 nt in the RE are critical for origin activity. The REm1-8-LBS1/2, REm9-16-LBS1/2, REm17-24-LBS1/2, and REm25-32-LBS1/2 mutants replicate with efficiencies of 4.8%, 9.6%, 27% and 60%, respectively, compared to that of the wild-type MR. (D) Fine mapping of the first 8 nt to determine the sequences important for LANA-dependent DNA replication. The REm1-4-LBS1/2, REm5-8-LBS1/2, REm9-12-LBS1/2, and REm13-16-LBS1/2 mutants replicate with efficiencies of 13.2%, 8.7%, 41%, and 100%, respectively, compared to that of the wild-type MR. The bracket and arrowheads indicate the positions of the origin-containing plasmids. LBSs, LBS1/2.

  • FIG. 2.
    • Open in new tab
    • Download powerpoint
    FIG. 2.

    The orientation and position of the RE element relative to LBS1/2 (LBSs) play an important role for origin function. The wild-type MR or mutants were constructed by inserting synthetic oligonucleotides into pCRII plasmids. Replication assays were performed as described in Materials and Methods. (A) Origin activity was diminished when the RE was flipped or moved downstream of LBS1/2. (B) Spacing changing between the RE and LBS1/2 abrogated the origin activity. The bracket and arrowhead indicate the positions of the origin-containing plasmids. The top panels show the representative structure of the wild-type MR and mutants; the bottom panels show the results of the replication assays with the wild-type MR and mutants.

  • FIG. 3.
    • Open in new tab
    • Download powerpoint
    FIG. 3.

    Proteomics approach to study RE binding proteins. (A) Flow chart for experimental design. Biotinylated PCR fragments containing the MR, REm1-8-LBS1/2, or Amp were immobilized on magnetic streptavidin beads. Nuclear extract and solubilized nuclear pellet extract mixture from BJAB Tet-on/ORF73 cells induced with doxycycline were subjected to two rounds of DNA affinity purification with the immobilized DNA. The second round of purified proteins was resolved on a 4 to 20% gradient SDS-PAGE gel. Gels in each lane were sliced and subjected for mass spectrometry analysis. (B) Colloidal blue staining of DNA affinity chromatography-purified proteins. Unique bands representing proteins specifically bound to REs are indicated with arrows. M, protein size marker. The molecular weight (in thousands) for each band of the marker is labeled as shown. LBSs, LBS1/2.

  • FIG. 4.
    • Open in new tab
    • Download powerpoint
    FIG. 4.

    Analysis of the TRF2 interaction with LANA and role in KSHV DNA replication. (A) LANA interacts with TRF2 in an vitro pulldown assay. BJAB LANA Tet-on nuclear extracts and solubilized nuclear pellets were subject to affinity purification with either the wild-type MR or the REm1-8-LBS1/2 mutant. The isolated proteins were analyzed by Western blotting with antibody specifically against human TRF2. A total of 0.5 ml nuclear protein mixture was loaded as the input (lane 4). NS, nonspecific signal. (B) LANA interacts with TRF2 in vivo in cotransfected cells. 293 cells were cotransfected with LANA and Myc-TRF2 expression vectors in conjunction with REm1-8-LBS1/2 (lane 1) or MR (lanes 2 and 3) plasmid. The nuclear extracts were immunoprecipitated with either monoclonal antibody against Myc (lanes 1 and 3) or control mouse IgG (lane 2). The immunoprecipitated proteins were separated on an 8% SDS-PAGE gel and immunoblotted with rabbit polyclonal antibody against LANA. (C) Expression of DN-TRF2. 293 cells were mock transfected or transfected with FLAG-DN-TRF2. Cell lysate was hybridized with antibody against FLAG. (D) DN-TRF2 did not interfere with LANA-dependent DNA replication. 293 cells were transfected with 8 mg TR-containing plasmid, 2 mg LANA expression vector in the absence of DN-TRF2 (lanes 2 and 6), and 2 mg (lanes 3 and 7) or 5 mg (lanes 4 and 8) DN-TRF2 expression vector. The short-term DNA replication assay was performed as described. The position of the TR plasmid is shown with an arrowhead LBSs, LBS1/2.

  • FIG. 5.
    • Open in new tab
    • Download powerpoint
    FIG. 5.

    Complex formation of SSRP1 with LANA/origin in vitro and in vivo. (A) SSRP1 was pulled down by the wild-type and mutant MR but not the amp control fragment when LANA was present. Affinity chromatography was performed as described in Materials and Methods. A small amount of pulldown lysates was separated on an 8% SDS-PAGE gel and transferred to a polyvinylidene difluoride membrane. The blot was immunoblotted with antibody specifically against SSRP1. Lanes 1 to 3 showed the lysates affinity purified with the amp fragment, mutant REm1-8-LBS1/2, or the wild-type MR fragment. Lane 4 showed 5% of the extract as input. (B) SSRP1 forms a complex with LANA/origin through protein-protein interaction. 293 cells were transfected with LANA expression vector and mutant REm1-8-LBS1/2 (lane 1), MR (lanes 2 to 4), or no origin (lane 5) in the presence (lanes 1, 3, 4, and 5) or absence (lane 2) of FLAG-tagged SSRP1. Cells were harvested 48 h posttransfection. Cells were lysed and immunoprecipitated with FLAG antibody (lanes 1, 2, 4, and 5) or mock antibody (lane 3) as a negative control. Immune complexes were separated on an 8% SDS-PAGE gel and immunoblotted with LANA antibody or FLAG antibody (bottom panels). The expression of LANA and FLAG was tested with 3% total cell lysate with antibodies specifically against LANA or FLAG (top two panels, shown as input). A faint background band was observed with the FLAG antibody (lane 2) compared to that with the control with no antibody (lane 3). LBSs, LBS1/2.

  • FIG. 6.
    • Open in new tab
    • Download powerpoint
    FIG. 6.

    SSRP1 forms a complex with KSHV latent origin in a cell cycle-dependent manner. (A) SSRP1 binds to the wild-type MR and mutant REm1-8-LBS1/2 when LANA is expressed. A total of 3 × 106 293 cells were cotransfected with vector pcDNA3.1-LANA, pcDNA-2xFLAG-SSRP1 and pCRII-REm1-8-LBS1/2, or pCRII-MR. Cells were harvested 48 h posttransfection for the ChIP assay. SSRP1 IP significantly enriches for MR and REm1-8-LBS1/2 (lanes 1 and 3) but not for a genomic control (lane 4); IgG IP demonstrates the specificity of the SSRP1 antibody (lane 2). (B) SSRP1 binds to the MR in a cell cycle-dependent manner. 293 cells were cotransfected with pcDNA3.1-LANA, pcDNA-2xFLAG-SSRP1 and pCRII-REm1-8-LBS1/2, or pCRII-MR. At 48 h after transfection, cells were asynchronized (bar 2) or synchronized to G1/S phase (bars 1, 3, and 4), S phase (bar 5), or M phase (bar 6) with drug treatments. Cells were harvested for the ChIP assay to detect the binding of SSRP1 on the MR or REm1-8-LBS1/2. Relative amounts of immunoprecipitated DNA were normalized to input for each sample. LBSs, LBS1/2.

  • FIG. 7.
    • Open in new tab
    • Download powerpoint
    FIG. 7.

    SSRP1 is involved in KSHV latent origin replication. A total of 1.5 × 106 293 cells were transfected with 100 nM scrambled or SSRP1 siRNA with Lipofectamine 2000. At 24 h posttransfection, cells were transfected again with 8 μg TR-containing plasmid pCRII-TR and 2 μg empty pcDNA3.1 (panels A and B, lanes 1) or pcDNA3.1/LANA (panels A and B, lanes 2, 3, and 4). At 72 h after the second transfection, cells were harvested. One of 20 of the total cells was collected for Western blotting to detect the expression of SSRP1. The remaining cells were assayed for the replication assay as described in Materials and Methods. (A) Western blot analysis indicated efficient SSRP1 knockdown by siRNA at the time when replicating DNA was extracted. Lanes 1 and 2, no siRNA transfection; lane 3, scrambled siRNA-transfected samples; lane 4, SSRP1 siRNA-transfected cells. (B) The knockdown of SSRP1 inhibits the replication efficiency of the TR plasmid. The top panel shows a representative image of the results from three independent replication assays. Lanes 1 to 4 (same samples as for panel A) contain 10% of the Hirt-extracted DNA digested with HindIII; lanes 5 to 8 have 90% of the Hirt-extracted DNA double digested with HindIII and DpnI, corresponding to lanes 1 to 4, respectively. Arrowheads indicate the positions of linearized pcDNA3.1/LANA and pCRII-TR. The bottom panel shows the quantification and statistical analysis of the results from three independent assays. (C) Cell profiling with mock-, scrambled siRNA-, or SSRP1 siRNA-transfected cells did not show any differences. DNA content was analyzed by flow cytometry after cells were fixed and stained with propidium iodide.

Tables

  • Figures
  • TABLE 1.

    Proteins identified in DNA affinity chromatography which bind only to the MR or have higher binding affinity to the MR than the mutants

    NameaAccession no.Predicted molecular mass (kDa)No. of peptides identified in MR pulldownFunction(s)
    DNA topoisomerase II β IPI0021770918346Control and alter the topologic states of DNA during transcription and replication; DNA damage response
    DNA topoisomerase II α IPI0021875317937Control and alter the topologic states of DNA during transcription and replication
    Structural maintenance of chromosome 1IPI002919391435Mitotic spindle organization and biogenesis; DNA repair
    Structural maintenance of chromosome 3IPI002194201425Mitotic spindle organization and biogenesis
    AF5q31 proteinIPI000043441274Transcription regulation
    Scaffold attachment factor B IPI003006311035Nuclear matrix component; transcription regulation
    M-phase phosphoprotein 8IPI00030408973N/Ab
    Gamma interferon-inducible protein Ifi-16IPI00003443889Transcription regulation; DNA damage signaling; cell cycle checkpoint
    Structure-specific recognition protein 1IPI00005154815Transcription regulation; chromatin remodeling; DNA replication
    DEAD box polypeptide 17 isoform p82 IPI00023785808RNA metabolism
    Heterogeneous nuclear ribonucleoprotein M isoform aIPI00171903784RNA metabolism
    Splicing factor proline/glutamine-rich (SFPQ) IPI00010740763RNA metabolism; DNA repair
    Lamina-associated polypeptide 2αIPI002162307533Nuclear architecture
    Interleukin enhancer-binding factor 3IPI00219330756Transcription regulation
    ALB proteinIPI00022434723Carrier protein
    Probable RNA-dependent helicase p68IPI00017617699RNA processing
    Nucleolar protein Nop56IPI00411937663RNA processing
    Telomeric repeat binding factor 2IPI00024214566Telomere maintenance; DNA replication
    Serine/threonine-protein kinase VRK3IPI000092915311Protein binding
    DERPCIPI00171540515DNA replication
    Elongation factor 1-α 2IPI00014424502Protein synthesis
    Heterogeneous nuclear ribonucleoprotein H1 IPI00013881492RNA processing
    KH domain-containing, RNA binding, signal transduction-associated protein 1 (Sam68)IPI00008575488RNA processing
    Gamma interferon-inducible protein Ifi-16 isoform 4IPI003848364710Transcription regulation
    JKTBP2IPI00011274463RNA processing
    SRP55-1 of splicing factorIPI00012345402RNA processing
    Pre-mRNA cleavage factor I 25-kDa subunitIPI00012995262RNA processing
    FUS-interacting serine-arginine-rich protein 1IPI00009071222RNA processing
    CGG triplet repeat binding protein 1IPI002955851912Transcription regulation
    • ↵ a Proteins in bold are six proteins that have previously been described as either LANA-interacting proteins or proteins which bind to the full-length TRs (41).

    • ↵ b N/A, not applicable.

PreviousNext
Back to top
Download PDF
Citation Tools
Involvement of SSRP1 in Latent Replication of Kaposi's Sarcoma-Associated Herpesvirus
Jianhong Hu, Eugene Liu, Rolf Renne
Journal of Virology Oct 2009, 83 (21) 11051-11063; DOI: 10.1128/JVI.00907-09

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Journal of Virology article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Involvement of SSRP1 in Latent Replication of Kaposi's Sarcoma-Associated Herpesvirus
(Your Name) has forwarded a page to you from Journal of Virology
(Your Name) thought you would be interested in this article in Journal of Virology.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Involvement of SSRP1 in Latent Replication of Kaposi's Sarcoma-Associated Herpesvirus
Jianhong Hu, Eugene Liu, Rolf Renne
Journal of Virology Oct 2009, 83 (21) 11051-11063; DOI: 10.1128/JVI.00907-09
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

Antigens, Viral
DNA replication
DNA, Viral
DNA-binding proteins
Herpesvirus 8, Human
High Mobility Group Proteins
Nuclear Proteins
Transcriptional Elongation Factors
Virus Latency
virus replication

Related Articles

Cited By...

About

  • About JVI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #Jvirology

@ASMicrobiology

       

 

JVI in collaboration with

American Society for Virology

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0022-538X; Online ISSN: 1098-5514