Deregulation of C-Myc in Primary Effusion Lymphoma by Kaposi's

Total Page:16

File Type:pdf, Size:1020Kb

Deregulation of C-Myc in Primary Effusion Lymphoma by Kaposi's Oncogene (2007) 26, 4979–4986 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc ORIGINAL ARTICLE Deregulation of c-Myc in primary effusion lymphoma by Kaposi’s sarcoma herpesvirus latency-associated nuclear antigen D Bubman1, I Guasparri2 and E Cesarman3 1Department of Pathology and Laboratory Medicine, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA; 2Department of Pathology and Laboratory Medicine, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA and 3Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, USA Primary effusion lymphoma (PEL) is a rare subtype of Introduction non-Hodgkin’s lymphoma, which is associated with infec- tion by Kaposi’s sarcoma herpesvirus (KSHV)/human Kaposi’s sarcoma herpesvirus (KSHV), or human herpesvirus-8. The c-Myc transcription factor plays an herpesvirus-8, is a lymphotrophic g2-herpesvirus that is important role in cellular proliferation, differentiation the etiologic agent of Kaposi’s sarcoma (Moore and and apoptosis. Lymphomas frequently have deregulated Chang, 1995), a subset of multicentric Castleman’s c-Myc expression owing to chromosomal translocations, disease (Soulier et al., 1995) and primary effusion amplifications or abnormal stabilization. However, no lymphoma (PEL) (Cesarman et al., 1995a). PEL is a structural abnormalities were found in the c-myc oncogene rare subtype of non-Hodgkin’s lymphoma that is most in PEL. Given that c-Myc is often involved in lympho- common in human immunodeficiency virus-infected magenesis, we hypothesized that it is deregulated in PEL. individuals, usually presenting as lymphomatous effu- We report that PEL cells have abnormally stable c-Myc sions in serous cavities. The expression of KSHV genes protein. The turnover of c-Myc protein is stringently is highly restricted in latently infected PEL cells. The few regulated by post-transcriptional modifications, including latent KSHV proteins expressed subvert various cellular phosphorylation of c-Myc threonine 58 (T58) by glycogen pathways in order to increase the proliferation and synthase kinase-3b (GSK-3b). Our data show that the survival of PEL cells. Most PEL cases are co-infected impaired c-Myc degradation in PEL cells is associated with Epstein–Barr virus (EBV), albeit with very with a significant underphosphorylation of c-Myc T58. restricted EBV latent gene expression (Horenstein The KSHV latency-associated nuclear antigen (LANA) is et al., 1997). Although KSHV is thought to be essential responsible for this deregulation. Overexpression of for PEL development, it is not sufficient, and EBV and LANA in human embryonic kidney 293 or peripheral other cellular factors are likely to play an important role blood B cells leads to post-transcriptional deregulation of (Fan et al., 2005). c-Myc protein. Conversely, when LANA is eliminated c-Myc is a major controller of cellular proliferation, from PEL cells using RNA interference, GSK-3b- and its deregulation is a common cause of malignancies mediated c-Myc T58 phosphorylation is restored. The (Pelengaris et al., 2002a). It is a basic helix–loop–helix- presence of c-Myc and LANA in GSK-3b-containing leucine-zipper transcription factor that regulates the complexes in PEL cells further confirms the significance transcription of numerous specific E-box-containing of these interactions in naturally KSHV-infected cells. genes as a heteromeric complex with a partner protein Oncogene (2007) 26, 4979–4986; doi:10.1038/sj.onc.1210299; Max (Henriksson and Luscher, 1996; Grandori and published online 19 February 2007 Eisenman, 1997). Constitutive expression of c-Myc suppresses exit from the cell cycle, interferes with cell Keywords: KSHV; HHV-8; PEL; c-Myc; LANA differentiation and can cause apoptotic cell death (Pelengaris et al., 2002b). Additionally, elevated expres- sion of c-Myc contributes to tumor formation in transgenic mice (Pelengaris and Khan, 2003). Frequent c-Myc deregulation occurs in lymphomas owing to chromosomal translocations and gene ampli- fication. The proto-oncogene is translocated to the immunoglobulin loci in nearly all Burkitt’s lymphomas (BL) (Dalla-Favera et al., 1982; Taub et al., 1982) and Correspondence: Associate Professor E Cesarman, Department of amplified in 16% of diffuse large B-cell lymphomas Pathology and Laboratory Medicine, Weill Medical College of Cornell (DLBCLs) (Rao et al., 1998). Mutations can result from University, 1300 York Ave, Room C410, New York, NY, USA. E-mail: [email protected] the germinal center somatic hypermutation machinery Received 17 June 2006; revised 12 August 2006; accepted 12 August in B cells (Gaidano et al., 2003) and can be found in the 2006; published online 19 February 2007 translocated alleles of primary tumors and cell lines c-Myc deregulation in KSHV-associated lymphomas D Bubman et al 4980 derived from BL and DLBCL. These mutations can be Results in regulatory elements and affect transcription (Cesar- man et al., 1987) or occur in coding regions (e.g., Myc c-Myc is stabilized in PEL cell lines Box 1 and threonine 58 (T58)), leading to impaired Previous studies evaluating c-myc in PEL did not find proteasome-mediated degradation of c-Myc protein translocations or mutations in the regulatory region of (Rabbitts et al., 1983; Bhatia et al., 1993; Yano et al., the proto-oncogene (Arvanitakis et al., 1996; Gaidano 1993). et al., 1996; Nador et al., 1996). However, analyses of Because of its transforming potential, c-Myc is a mutations in the coding region of c-Myc have not been short-lived protein with a strictly regulated half-life of reported. Therefore, we sequenced exon 2 of c-Myc about 30 min (Hann and Eisenman, 1984; King et al., DNA from a PEL cell line, BC-3 and found no 1986). Fine tuning of protein stability is achieved mutations in the Myc Box 1. As abnormal stabilization through phosphorylation of two specific residues in is a mechanism for c-Myc deregulation, we sought to Myc box 1 located in the N-terminal transactivation examine c-Myc protein turnover in PEL cell lines. BC-1, domain, T58 and serine 62 (S62) and subsequent BC-3, BC-5 and BCBL-1 cells were treated with a proteasomal degradation (Dominguez-Sola and protein synthesis inhibitor, cycloheximide and c-Myc Dalla-Favera, 2004). These phosphorylation events degradation was monitored using immunoblot analysis. take place in an orderly manner during the G1 stage BL cell lines Daudi and JD38 were previously reported of the cell cycle through the stimulation of Ras by to have a normal and a prolonged half-life of c-Myc, growth factors and its activation of the Raf-MAPK/ respectively (Gregory and Hann, 2000), and were used ERK kinase-extracellular signal-regulated kinase and as a negative and a positive control. Figure 1 shows that phosphatidylinositol 30-kinase/Akt pathways. Phos- the steady-state level of c-Myc declined rapidly at 30 min phorylation of c-Myc on S62 is necessary for its in Daudi as reported, but decayed at a much slower pace phosphorylation by glycogen synthase kinase-3b in the PEL cell lines. All PEL cells exhibited a similar or (GSK-3b) on T58, which is crucial for c-Myc degrada- more stable turnover profile to that of JD38 (positive tion in late G1 (Gregory and Hann, 2000; Sears et al., control), which has a deregulation in c-Myc stability. 2000). Subsequent dephosphorylation of S62 by protein We also looked at the levels of LANA protein in PEL phosphatase 2A (PP2A), is required for a proper cells, which went down slightly at the 2-h time point. recognition of c-Myc by the proteasomal machinery Immunoblot for actin confirmed equal protein loading. (Yeh et al., 2004). Toextend the cycloheximide experiments pulse chase Although aberrant c-Myc appears to be important for studies were performed, which confirmed that PEL cells lymphomagenesis, previous studies failed to detect have abnormally stable c-Myc (data not shown). From structural abnormalities in the c-myc gene in PELs these experiments we concluded that KSHV-infected (Arvanitakis et al., 1996; Gaidano et al., 1996; Nador PEL cell lines have an abnormally stable c-Myc protein. et al., 1996). Therefore, we wanted to determine whether Although c-Myc appears to have a shorter half-life it is deregulated in this malignancy, and if so, identify in the KSHV þ /EBV þ cell lines BC-1 and BC-5 than in the KSHV gene responsible for the deregulation. A the KSHV þ /EBVÀ cell lines BC-3 and BCBL-1, possible candidate is the latency-associated nuclear further studies are needed toascertain the effect of antigen (LANA). LANA is one of the few latent genes EBV on c-Myc stability in PEL cells. expressed in KSHV-infected PEL cell lines. It tethers viral DNA to host chromosomes to ensure efficient viral LANA enhances c-Myc stability in HEK 293 cells replication (Ballestas et al., 1999). In addition, LANA To test the hypothesis that LANA is responsible for associates with various proteins including p53 and pRb c-Myc deregulation, we transfected human embryonic and through these interactions stimulates pathways kidney (HEK) 293 cells with a FLAG-tagged c-Myc and involved in survival and proliferation (Friborg et al., empty vector or LANA-expressing vectors and per- 1999; Radkov et al., 2000). Recently, it was found that formed protein stability studies. Figure 2a and b shows LANA promotes stabilization of b-catenin in PEL cells that transfection of LANA significantly increased the by inhibiting its negative regulator, GSK-3b (Fujimuro stability of transfected c-Myc-FLAG protein. We et al., 2003). GSK-3b phosphorylates numerous sub- observed the same effect when looking at endogenous strates, including more than a dozen transcription c-Myc stability in LANA-transfected cells (data not factors, such as activator protein-1 (AP-1), cAMP shown).
Recommended publications
  • ORIGINAL ARTICLE Short Telomeres and High Telomerase Activity in T
    Leukemia (2007) 21, 2456–2462 & 2007 Nature Publishing Group All rights reserved 0887-6924/07 $30.00 www.nature.com/leu ORIGINAL ARTICLE Short telomeres and high telomerase activity in T-cell prolymphocytic leukemia ARo¨th1,JDu¨rig1, H Himmelreich2,3, S Bug4, R Siebert4,UDu¨hrsen1, PM Lansdorp5,6 and GM Baerlocher2,3 1Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; 2Department of Hematology, University Hospital, Bern, Switzerland; 3Department of Clinical Research, University Hospital, Bern, Switzerland; 4Institute of Human Genetics, Christian-Albrechts-University Hospital Schleswig-Holstein, Kiel, Germany; 5Terry Fox Laboratory, BC Cancer Research Centre, Vancouver, Canada and 6Department of Medicine, University of British Columbia, Vancouver, Canada 4 5 To test the role of telomere biology in T-cell prolymphocytic repeats of the sequence T2AG3 and associated proteins folded leukemia (T-PLL), a rare aggressive disease characterized by into a telomere loop structure.6 Telomeres are required to the expansion of a T-cell clone derived from immuno-compe- tent post-thymic T-lymphocytes, we analyzed telomere length maintain chromosomal integrity and prevent end-to-end fusions and telomerase activity in subsets of peripheral blood leuko- of chromosomes. When telomeric ends become too short, DNA cytes from 11 newly diagnosed or relapsed patients with damage signals from telomeres can induce apoptosis or a state sporadic T-PLL. Telomere length values of the leukemic T cells of replicative senescence.7,8 Telomeres shorten with each round (mean7s.d.: 1.5370.65 kb) were all below the 1st percentile of of cell division as a result of failure to completely replicate the 30 telomere length values observed in T cells from healthy age- end of chromosomes9,10 as well as other causes.11 The average matched controls whereas telomere length of normal T- and telomere length in cells from most human tissues decreases with B cells fell between the 1st and 99th percentile of the normal 12,13 distribution.
    [Show full text]
  • S Sarcoma-Associated Herpesvirus Lana2 Protein Interacts with the Pocket Proteins and Inhibits Their Sumoylation
    Oncogene (2014) 33, 495–503 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc ORIGINAL ARTICLE Kaposi’s sarcoma-associated herpesvirus lana2 protein interacts with the pocket proteins and inhibits their sumoylation L Marcos-Villar1,5, P Gallego1,5, C Mun˜ oz-Fontela2, CF de la Cruz-Herrera1, M Campagna1, D Gonza´lez1, F Lopitz-Otsoa3, MS Rodrı´guez3,4 and C Rivas1 The pocket proteins retinoblastoma protein (pRb), p107 and p130 are the key targets of oncoproteins expressed by DNA tumor viruses. Some of these viral proteins contain an LXCXE motif that mediates the interaction with the three pocket proteins and the inhibition of the pRb SUMOylation. Kaposi’s sarcoma herpesvirus (KSHV) contains at least two proteins that can regulate pRb function but, so far, a KSHV-encoded protein targeting p107 and p130 has not been identified. Here, we show that the KSHV latent protein LANA2 binds to pRb, p107 and p130. LANA2 contains an LXCXE motif that is required for bypassing pRb-mediated cell-cycle arrest and for inhibiting pRb SUMOylation. Finally, we demonstrate that, in addition to pRb, both p107 and p130 can be SUMOylated, and this modification is also inhibited by LANA2 in an LXCXE-dependent manner. These results demonstrate, for the first time, the SUMOylation of p107 or p130 and, so far, they represent the first example of a KSHV protein able to interact with the three pocket proteins and to inhibit their conjugation to SUMO. Oncogene (2014) 33, 495–503; doi:10.1038/onc.2012.603; published online 14 January 2013 Keywords: pocket proteins; KSHV; LANA2; pocket proteins; sumoylation; LXCXE domain INTRODUCTION vIRF3, interacts with pRb, p130 and p107 in vitro and in vivo.
    [Show full text]
  • THE NANOARCHITECTURE of the KSHV LANA TETHER and APPROACHES to ITS DISRUPTION Margaret Josephine Grant Shelton, CT B.S., Brown U
    THE NANOARCHITECTURE OF THE KSHV LANA TETHER AND APPROACHES TO ITS DISRUPTION Margaret Josephine Grant Shelton, CT B.S., Brown University, 2007 A Dissertation presented to the Graduate Faculty of the University of Virginia in Candidacy for the degree of Doctor of Philosophy Department of Microbiology, Immunology, and Cancer Biology University of Virginia May 2018 _____________________________ _____________________________ _____________________________ _____________________________ _____________________________ _____________________________ ii ABSTRACT Kaposi’s sarcoma-associated herpesvirus (KSHV) is the causative agent of three human malignancies and presents a special concern to immunocompromised individuals. Its latency-associated nuclear antigen protein (LANA) tethers latent viral genomes to host chromatin, thereby maintaining viral infection, and as such, is an attractive target for therapeutic intervention. In an effort to better understand this protein and its tethering function, we have used super-resolution microscopy to examine LANA tethers, obtaining information that remained obscured in earlier studies using standard epifluorescence microscopy. We have determined several characteristics of these tethers, including the folding properties of the underlying viral DNA and occupancy of LANA on its available viral binding sites. Quantitative data support the prediction of a coiled-coil feature in LANA dimers, and computer modeling of a minimal LANA tether illustrates the importance of viral DNA bending and nucleosome positioning on tether structure. Preliminary data examining LANA tethers during cellular mitosis suggest a potential role for mitotic machinery in manipulating tether positioning and condensation. This work also begins to address the relative timing of host chromosome condensation and LANA tether formation. These promising early results compel further study of the mitotic LANA tether and its dynamics.
    [Show full text]
  • Transcription Profile of Kaposi's Sarcoma-Associated Herpesvirus in Primary Kaposi's Sarcoma Lesions As Determined by Real-Time PCR Arrays
    [CANCER RESEARCH 63, 2010–2015, May 1, 2003] Advances in Brief Transcription Profile of Kaposi’s Sarcoma-associated Herpesvirus in Primary Kaposi’s Sarcoma Lesions as Determined by Real-Time PCR Arrays1 Dirk P. Dittmer2 Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 Abstract tumor cell expresses at least one viral protein: the LANA/orf73 (2). Antibodies to LANA exist in virtually all HIV-infected as well as Kaposi’s sarcoma (KS) is the signature pathology of AIDS. KS-associ- non-HIV-infected KS patients, and other viral antigens have also been ated herpesvirus (KSHV/HHV-8) is causally linked to KS. Here, we report identified as the targets of this response. Prospective, longitudinal the first complete profile of KSHV transcription in primary KS lesions studies found that increases in peripheral blood viral load as well as using a novel, real-time PCR array. The KSHV latency I mRNAs [latency- associated nuclear antigen (LANA)/orf73, v-cyclin/orf72, and v-FLIP/ KSHV-specific antibody titers precede the onset of disease and cor- orf71] were invariably present in all biopsies. Yet, viral lytic mRNAs were relate with increased risk for KS. In addition, two lymphoproliferative detectable in only a subset of tumors. Interestingly, there was a difference disorders, primary effusion lymphoma (PEL) and multicentric Castle- in the expression pattern of the viral IFN-regulatory factors (vIRFs) man’s disease (MCD), are associated with KSHV (3, 4). These ob- encoded by KSHV. The vIRF-1/K9 clustered with LANA in KS, in con- servations imply (a) that KSHV viral oncogenes are required for KS trast to its homologue, vIRF-3/LANA-2, which is transcribed only in development and (b) that the identification of these viral genes may KSHV-associated lymphomas.
    [Show full text]
  • Visualization of Molecular Biology: the LANA Tether Vaibhav Jaina and Rolf Rennea,B,C,1
    COMMENTARY COMMENTARY Visualization of molecular biology: The LANA tether Vaibhav Jaina and Rolf Rennea,b,c,1 The latency-associated nuclear antigen (LANA) plays we focus on the role of LANA with respect to ge- a central role in the biology and pathogenesis of nome persistence during latency. The first evidence Kaposi’s sarcoma (KS)-associated herpesvirus (KSHV). that KSHV LANA, like EBNA1 from the related hu- Both classical and endemic KS in HIV-infected individ- man tumor virus Epstein–Barr virus (EBV), is re- uals and two lymphoproliferative diseases are associ- sponsibleforgenomesegregationcamein1999, ated with KSHV. During the latent phase of the viral when it was demonstrated that plasmids containing TR life cycle in dividing tumor cells, the LANA protein en- sequences were stably segregated in cells expressing sures that viral genomes persist by supporting both LANA (9, 10). Multiple groups identified the TR se- the initiation of DNA replication and segregation of quences as cis-regulatory elements essential for both viral episomes (nonintegrated circular viral genomes) the initiation of DNA replication and the segregation into daughter cells. Arguably, interrupting these com- of TR-containing plasmids during mitosis. The LANA plex LANA-dependent processes could be one of the C-terminal domain was mapped and shown to bind most promising antiviral and antitumor therapeutic to two LANA binding sites (LBS1 and LBS2) in a co- strategies. Hence, studying the molecular and cell bio- operative manner (11). Next, elegant structural and ge- logical details of LANA’s host/viral protein and chro- netic approaches demonstrated that an 18-aa-long matin interactions has been a focus in a number of N-terminal peptide specifically interacts with the H2A/ laboratories since 1996.
    [Show full text]
  • Herpesviral Latency—Common Themes
    pathogens Review Herpesviral Latency—Common Themes Magdalena Weidner-Glunde * , Ewa Kruminis-Kaszkiel and Mamata Savanagouder Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Str. 10, 10-748 Olsztyn, Poland; [email protected] (E.K.-K.); [email protected] (M.S.) * Correspondence: [email protected] Received: 22 January 2020; Accepted: 14 February 2020; Published: 15 February 2020 Abstract: Latency establishment is the hallmark feature of herpesviruses, a group of viruses, of which nine are known to infect humans. They have co-evolved alongside their hosts, and mastered manipulation of cellular pathways and tweaking various processes to their advantage. As a result, they are very well adapted to persistence. The members of the three subfamilies belonging to the family Herpesviridae differ with regard to cell tropism, target cells for the latent reservoir, and characteristics of the infection. The mechanisms governing the latent state also seem quite different. Our knowledge about latency is most complete for the gammaherpesviruses due to previously missing adequate latency models for the alpha and beta-herpesviruses. Nevertheless, with advances in cell biology and the availability of appropriate cell-culture and animal models, the common features of the latency in the different subfamilies began to emerge. Three criteria have been set forth to define latency and differentiate it from persistent or abortive infection: 1) persistence of the viral genome, 2) limited viral gene expression with no viral particle production, and 3) the ability to reactivate to a lytic cycle. This review discusses these criteria for each of the subfamilies and highlights the common strategies adopted by herpesviruses to establish latency.
    [Show full text]
  • Latency-Associated Nuclear Antigen of Kaposi's Sarcoma-Associated
    JOURNAL OF VIROLOGY, Oct. 2004, p. 10348–10359 Vol. 78, No. 19 0022-538X/04/$08.00ϩ0 DOI: 10.1128/JVI.78.19.10348–10359.2004 Copyright © 2004, American Society for Microbiology. All Rights Reserved. Latency-Associated Nuclear Antigen of Kaposi’s Sarcoma-Associated Herpesvirus Up-Regulates Transcription of Human Telomerase Reverse Transcriptase Promoter through Interaction with Transcription Factor Sp1 Subhash C. Verma, Sumit Borah, and Erle S. Robertson* Department of Microbiology and Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania Received 20 March 2004/Accepted 11 May 2004 Telomerase is required for the maintenance of telomere length and is an important determinant for cell immortalization. In human cells, telomerase activity is due to the expression of its enzymatic subunit, human telomerase reverse transcriptase (hTERT). The expression of hTERT is not typically detectable in healthy somatic human cells but is present in cancerous tissues and immortalized cells. We have previously shown that hTERT promoter activity is up-regulated by the Kaposi’s sarcoma-associated herpesvirus (KSHV)-encoded latency-associated nuclear antigen (LANA). LANA is expressed in all forms of human malignancies associated with KSHV. The hTERT promoter sequence located at positions ؊130 to ؉5 contains several Sp1 binding motifs and was shown be important for up-regulation by LANA. In this report, we demonstrate that hTERT promoter activity is due to the direct interaction of LANA with Sp1. The interaction of LANA with Sp1 was demonstrated through in vitro binding experiments and coimmunoprecipitation and is supported by the colocalization of these two molecules in the nuclei of KSHV-infected cells.
    [Show full text]
  • Ribosomal Protein S6 Interacts with the Latency-Associated Nuclear Antigen of Kaposi’S Sarcoma-Associated Herpesvirusᰔ Wuguo Chen and Dirk P
    JOURNAL OF VIROLOGY, Sept. 2011, p. 9495–9505 Vol. 85, No. 18 0022-538X/11/$12.00 doi:10.1128/JVI.02620-10 Copyright © 2011, American Society for Microbiology. All Rights Reserved. Ribosomal Protein S6 Interacts with the Latency-Associated Nuclear Antigen of Kaposi’s Sarcoma-Associated Herpesvirusᰔ Wuguo Chen and Dirk P. Dittmer* Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, Center for AIDS Research (CfAR), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290 Received 16 December 2010/Accepted 28 June 2011 The latency-associated nuclear antigen (LANA) is central to the maintenance of Kaposi’s sarcoma-associ- ated herpesvirus (KSHV) and to the survival of KSHV-carrying tumor cells. In an effort to identify interaction partners of LANA, we purified authentic high-molecular-weight complexes of LANA by conventional chroma- tography followed by immunoprecipitation from the BC-3 cell line. This is the first analysis of LANA- interacting partners that is not based on forced ectopic expression of LANA. Subsequent tandem mass spectrometry (MS/MS) analysis identified many of the known LANA-interacting proteins. We confirmed LANA’s interactions with histones. Three classes of proteins survived our stringent four-step purification procedure (size, heparin, anion, and immunoaffinity chromatography): two heat shock proteins (Hsp70 and Hsp96 precursor), signal recognition particle 72 (SRP72), and 10 different ribosomal proteins. These proteins are likely involved in structural interactions within LANA high-molecular-weight complexes. Here, we show that ribosomal protein S6 (RPS6) interacts with LANA. This interaction is mediated by the N-terminal domain of LANA and does not require DNA or RNA.
    [Show full text]
  • CRISPR Interference Efficiently Silences Latent and Lytic Viral Genes in Kaposi’S Sarcoma-Associated Herpesvirus-Infected Cells
    viruses Article CRISPR Interference Efficiently Silences Latent and Lytic Viral Genes in Kaposi’s Sarcoma-Associated Herpesvirus-Infected Cells Kevin Brackett 1, Ameera Mungale 1, Mary Lopez-Isidro 1, Duncan A. Proctor 1, Guillermo Najarro 1 and Carolina Arias 1,2,3,* 1 Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, USA; [email protected] (K.B.); [email protected] (A.M.); [email protected] (M.L.-I.); [email protected] (D.A.P.); [email protected] (G.N.) 2 Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA 3 Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, CA 93106, USA * Correspondence: [email protected] Abstract: Uncovering viral gene functions requires the modulation of gene expression through overexpression or loss-of-function. CRISPR interference (CRISPRi), a modification of the CRISPR- Cas9 gene editing technology, allows specific and efficient transcriptional silencing without genetic ablation. CRISPRi has been used to silence eukaryotic and prokaryotic genes at the single-gene and genome-wide levels. Here, we report the use of CRISPRi to silence latent and lytic viral genes, with an efficiency of ~80–90%, in epithelial and B-cells carrying multiple copies of the Kaposi’s sarcoma-associated herpesvirus (KSHV) genome. Our results validate CRISPRi for the analysis of KSHV viral elements, providing a functional genomics tool for studying virus–host interactions. Citation: Brackett, K.; Mungale, A.; Keywords: KSHV; CRISPR-interference; dCas9-KRAB; Kaposi’s sarcoma-associated herpesvirus; Lopez-Isidro, M.; Proctor, D.A.; gene expression; gene silencing Najarro, G.; Arias, C.
    [Show full text]
  • Superresolution Microscopy Reveals Structural Mechanisms Driving the Nanoarchitecture of a Viral Chromatin Tether Margaret J
    Superresolution microscopy reveals structural mechanisms driving the nanoarchitecture of a viral chromatin tether Margaret J. Granta, Matthew S. Loftusa, Aiola P. Stojaa, Dean H. Kedesa,b,1, and M. Mitchell Smitha,1 aDepartment of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908; and bDepartment of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908 Edited by Donald E. Ganem, Novartis Institutes for Biomedical Research, Inc., Emeryville, CA, and approved March 2, 2018 (received for review December 13, 2017) By tethering their circular genomes (episomes) to host chromatin, qPCR to demonstrate a direct proportionality between the num- DNA tumor viruses ensure retention and segregation of their ber of LANA punctae and the amount of viral DNA, leading to genetic material during cell divisions. Despite functional genetic the conclusion that each nuclear dot represented a single viral and crystallographic studies, there is little information address- episome. Previous studies examining these punctae showed asso- ing the 3D structure of these tethers in cells, issues critical for ciation of LANA with mitotic chromosomes at the resolution understanding persistent infection by these viruses. Here, we of epifluorescence microscopy (12, 27). Kelley-Clarke et al. (28) have applied direct stochastic optical reconstruction microscopy later suggested preferential localization of these LANA punc- (dSTORM) to establish the nanoarchitecture of tethers within cells tae to centromeric and telomeric regions on metaphase chromo- latently infected with the oncogenic human pathogen, Kaposi’s some. Such studies have provided a solid foundation for further sarcoma-associated herpesvirus (KSHV). Each KSHV tether com- inquiry into the nature of this tethering mechanism.
    [Show full text]
  • Cell Cycle Regulatory Functions of the KSHV Oncoprotein LANA
    fmicb-07-00334 March 25, 2016 Time: 17:58 # 1 REVIEW published: 30 March 2016 doi: 10.3389/fmicb.2016.00334 Cell Cycle Regulatory Functions of the KSHV Oncoprotein LANA Fang Wei1, Jin Gan2, Chong Wang2, Caixia Zhu2 and Qiliang Cai2* 1 ShengYushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China, 2 MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China Manipulation of cell cycle is a commonly employed strategy of viruses for achieving a favorable cellular environment during infection. Kaposi’s sarcoma-associated herpesvirus (KSHV), the primary etiological agent of several human malignancies including Kaposi’s sarcoma, and primary effusion lymphoma, encodes several oncoproteins that deregulate normal physiology of cell cycle machinery to persist with endothelial cells and B cells and subsequently establish a latent infection. During latency, only a small subset of viral proteins is expressed. Latency-associated nuclear antigen (LANA) is one of the latent antigens shown to be essential for transformation of endothelial cells in vitro. It has been well demonstrated that LANA is critical for the maintenance of latency, episome DNA replication, segregation and gene transcription. In this review, we summarize recent studies and address how LANA Edited by: functions as an oncoprotein to steer host cell cycle-related events including proliferation Erle S. Robertson, and apoptosis by interacting with various cellular and viral factors, and highlight the University of Pennsylvania, USA potential therapeutic strategy of disrupting LANA-dependent signaling as targets in Reviewed by: Masahiro Fujimuro, KSHV-associated cancers.
    [Show full text]
  • Virus-Driven Carcinogenesis
    cancers Review Virus-Driven Carcinogenesis Yuichiro Hatano 1,* , Takayasu Ideta 2,3, Akihiro Hirata 4, Kayoko Hatano 5, Hiroyuki Tomita 1 , Hideshi Okada 6 , Masahito Shimizu 2 , Takuji Tanaka 7 and Akira Hara 1 1 Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; [email protected] (H.T.); [email protected] (A.H.) 2 Department of Gastroenterology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; [email protected] (T.I.); [email protected] (M.S.) 3 Department of Laboratory Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan 4 Laboratory of Veterinary Pathology, Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1194, Japan; [email protected] 5 Department of Obstetrics and Gynecology, Gifu University Hospital, Gifu 501-1194, Japan; [email protected] 6 Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; [email protected] 7 Department of Diagnostic Pathology (DDP) and Research Center of Diagnostic Pathology (RC-DiP), Gifu Municipal Hospital, Gifu 500-8513, Japan; [email protected] * Correspondence: [email protected]; Tel.: +81-58-230-6225 Simple Summary: Carcinogens, causes of cancer, are usually invisible and therefore in vivo carcino- genesis is difficult to detect. Tumor viruses, definitive carcinogens, are also usually unremarkable, particularly due to latent infection. However, recent developments in tumor virology are unraveling how a single infected cell becomes a life-threatening cell population from a molecular perspective.
    [Show full text]