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
Gem

In Vivo Models of Oncoproteins Encoded by Kaposi’s Sarcoma-Associated Herpesvirus

Ariana G. Bravo Cruz, Blossom Damania
Michaela Ulrike Gack, Editor
Ariana G. Bravo Cruz
aDepartment of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Blossom Damania
aDepartment of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michaela Ulrike Gack
University of Chicago
Roles: Editor
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/JVI.01053-18
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Kaposi’s sarcoma-associated herpesvirus (KSHV) is a human oncogenic virus. KSHV utilizes its proteins to modify the cellular environment to promote viral replication and persistence. Some of these proteins are oncogenic, modulating cell proliferation, apoptosis, angiogenesis, genome stability, and immune responses, among other cancer hallmarks. These changes can lead to the development of KSHV-associated malignancies. In this Gem, we focus on animal models of oncogenic KSHV proteins that were developed to enable better understanding of KSHV tumorigenesis.

INTRODUCTION

Viruses are linked to approximately 12% of all human cancers (1). Kaposi’s sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is a member of the Gammaherpesvirinae subfamily and is linked to three human cancers: the endothelial cancer Kaposi’s sarcoma (KS) and two B-cell lymphoproliferative diseases, primary effusion lymphoma (PEL), a type of non-Hodgkin lymphoma (NHL), and multicentric Castleman’s disease (MCD) (2–4). These malignancies develop primarily in immunocompromised individuals, although they can occur in HIV-naive individuals as well (5). KSHV is classified as a category 1 carcinogen by IARC/WHO (6).

Like other members of the Herpesviridae family, KSHV establishes a lifelong infection in the host, alternating between two distinct life cycles: latent and lytic. During latency, a limited number of genes are expressed, and the viral genome is maintained as an episome and is passed to daughter cells during cell division. During lytic infection, most of the viral genes are expressed, the viral genome is replicated, and new virions are produced.

The mechanisms by which KSHV infection leads to the development of cancer are not completely understood. However, it is believed that KSHV proteins play a critical role in tumorigenesis. KSHV expresses a myriad of lytic and latent proteins that alter the cellular environment to promote the survival of the infected cell and the establishment of latency; such alterations, including the induction of cell proliferation, evasion of apoptosis, and immune evasion, coincide with hallmarks of cancer (1).

Several transgenic mouse models have been developed to study the contributions of KSHV proteins to oncogenesis in vivo. These genetically modified mice express one or a few KSHV proteins shown to have oncogenic properties in cultured cells. The transgenic KSHV mouse models developed to date are discussed in this Gem (Table 1). Expression of KSHV proteins in mice often results in the development of a variety of diseases similar to those linked to KSHV, confirming the contributions of these proteins to KSHV-induced tumorigenesis. These models represent a suitable alternative for the study of KSHV pathogenesis, not only to provide insights into the mechanisms of KSHV-associated tumorigenesis but also to evaluate drugs to treat KSHV-associated infections by targeting these viral proteins.

View this table:
  • View inline
  • View popup
TABLE 1

Transgenic mouse models of KSHV oncoproteins

MOUSE MODELS OF KSHV-ENCODED LATENT ONCOPROTEINS

The default phase of the KSHV life cycle is latency. Most KSHV-infected cells remain dormant throughout the lifetime of the host. During latent infection, few viral proteins are expressed; this ensures viral persistence by deregulating host oncogenic pathways, promoting cell survival, proliferation, and immune evasion. Such prolonged modulation of cellular homeostasis can lead to cell transformation and immortalization and to the subsequent development of KSHV-associated malignancies. In this section, we discuss the KSHV latent proteins that have been shown to have oncogenic properties in mouse models and the cellular pathways they modulate that potentially explain their roles in tumorigenesis (Fig. 1).

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

KSHV-encoded oncoproteins and their modulation of cancer-inducing pathways leading to tumorigenesis (see reference 1) in transgenic mouse models. Created with BioRender.

LANA.The latency-associated nuclear antigen (LANA) is known for its critical roles in latency maintenance, genomic viral DNA replication, and latent gene transcription (7–9). However, in this section, we focus on the oncogenic properties of LANA.

In cell culture models, LANA is implicated in KSHV-induced tumorigenesis by altering cellular pathways involved in cell cycle progression, proliferation, and apoptosis. LANA modulates retinoblastoma protein (Rb), p53 and its homolog p73, NOTCH, WNT, and mitogen-activated protein kinase (MAPK) signaling (10–14). In addition, LANA alters cell death, growth, and proliferation by modulating survivin and β-catenin as well as c-Myc and c-Jun signaling (15–19). Moreover, LANA modulates angiogenesis and induces chromosomal instability (20, 21). Given these oncogenic properties, it is not surprising that LANA transforms primary rat embryo fibroblasts (10), prolongs the life of primary human endothelial vein cells (22), and, as discussed below, induces lymphomagenesis in mice.

The LANA transgenic mouse model expresses LANA from its natural promoter (23), which has been shown to be B-cell specific (24). Eleven percent of LANA transgenic mice develop monoclonal tumors, which are of B-cell lineage and express LANA (23). In addition, mature B cells are activated and hyperproliferative. This expansion resembles KSHV-associated lymphomas, specifically MCD and plasmablastic lymphomas.

A follow-up study further elucidated that LANA lowers the activation threshold of B cells, leading to their continual activation without exhaustion (25). LANA transgenic mice have a significant increase in mature CD19+ IgM+ IgD+ B cells. In addition, early germinal center (GC) and activated follicular cells are increased. Crossing LANA mice with CD19−/− mice shows that while LANA is not sufficient to compensate for the lack of CD19 (which is needed for GC development), it can partially restore marginal-zone B-cell development (25). Taken together, these data suggest that LANA lowers the B-cell receptor (BCR) signaling threshold in KSHV-infected cells.

vCyclin.The KSHV gene ORF72 (open reading frame 72) encodes a homolog of cellular cyclin D known as viral cyclin (vCyclin) (26, 27), which is expressed during both the latent and lytic replication stages (28). vCyclin interacts with several cyclin-dependent kinases (CDKs), though preferentially with CDK6. Such vCyclin-CDK complexes are resistant to known CDK inhibitors and have an increased number of substrates (29–32), making vCyclin a strong deregulator of cell cycle progression. In addition, vCyclin has been shown to modulate p53 and Bcl-2 (33, 34) and to induce the DNA damage response (DDR) (35).

To evaluate the oncogenic properties of vCyclin in vivo, a transgenic mouse model was generated in which vCyclin is expressed under the transcriptional control of the Eμ promoter, which allows for the expression of vCyclin in B and T cells (36). Approximately 17% of 6- to 9-month-old vCyclin transgenic mice develop B- or T-cell lymphomas (37).

Several lines of evidence suggest that p53 contributes to the suppression of vCyclin-induced lymphomagenesis: (i) expression of vCyclin in cell culture leads to p53-dependent cell growth arrest (36); (ii) vCyclin tumors exhibit elevated levels of p53 and p19ARF, an inhibitor of p53 degradation (37); and (iii) crossing vCyclin transgenic mice with p53−/− mice results in much more rapid and frequent development of T- and B-cell lymphomas (36, 37). The B-cell lymphomas are IgM–, and the T-cell lymphomas are P/TCRlow, suggesting that vCyclin induces precursor B- and/or T-cell lymphoma in p53−/− mice. vCyclin/p53−/− mice show genome instability, demonstrated by a high frequency of aneuploidy.

To better study the contributions of vCyclin to KS development, a mouse model has been established which expresses vCyclin under the control of the vascular endothelial growth factor receptor 3 (VEGFR-3) promoter (38), resulting in vCyclin expression in lymphatic endothelial cells. In this model, 80% of vCyclin mice die prematurely by the age of 6 months and show pleural fluid accumulation, lymphatic dysfunction, and erythematous skin.

vFLIP.KSHV expresses the latent viral FADD-like interleukin-1-converting enzyme (FLICE) inhibitory protein (vFLIP), also known as K13, in both KS and PEL (39, 40). Like other cellular and viral FLIPs, vFLIP inhibits Fas-induced apoptosis by preventing the recruitment and processing of caspase 8 (41–44). In addition, vFLIP regulates cell death by inhibiting autophagy (45). Uniquely among FLIPs, vFLIP activates NF-κB (46, 47). Given vFLIP’s inhibition of apoptosis, activation of NF-κB, and subsequent transformation of Rat-1 and BALB/c 3T3 cells (48), a role for vFLIP in KSHV-induced lymphomagenesis has been proposed. Below, several in vivo experiments supporting this hypothesis are discussed.

A transgenic mouse model was generated in which the vFLIP transgene is expressed under the control of the H-2Kb promoter and the Ig heavy chain enhancer (49). Surprisingly, vFLIP does not inhibit intrinsic or extrinsic apoptosis in transgenic mice. However, it induces constitutive activation of NF-κB. vFLIP transgene expression leads to an increase in cell proliferation and a higher incidence of lymphoma (11.8%) than that in age-matched wild-type mice (1.8%). vFLIP-induced lymphomas are B220+, express vFLIP, and show constitutive NF-κB activation. Given the low lymphoma incidence and the inability to recapitulate PEL in these mice, two transgenic mouse lines expressing vFLIP at different stages of B-cell development were generated (50). One transgenic mouse line expresses vFLIP under the control of the CD19 promoter, resulting in vFLIP expression in all B cells. A second line expresses vFLIP under the control of the Cγ1 promoter, restricting expression to IgG1 GC B cells. While neither line recapitulates PEL, vFLIP expression results in B-cell abnormalities similar to those observed in MCD, including lack of GC formation, increased frequency of marginal-zone and FAS+ B cells, and failure to produce class-switched IgG.

Latency locus.The KSHV latency locus (LL) includes LANA, vCyclin, vFLIP, K12, and all viral microRNAs (miRNAs). The genes in the LL are constitutively expressed from a single B-cell-specific promoter in all KSHV-infected cells. B cells from LL mice, which express the LL from its natural promoter, exhibit chronically activated mature B cells, leading to hyperglobulinemia due to increases in the frequencies of CD138+ plasma cells and marginal-zone B cells (51). In addition, the augmentation of marginal-zone and GC B-cell responses suggests that the LL drives both T-independent and T-dependent B-cell activation, respectively. The B-cell activation abilities of the LL were further demonstrated by showing that the LL can drive B-cell development even in the absence of miRNA-155, which is normally required for B- and T-cell function and is expressed in many cancers (52). In addition, the LL can compensate for the lack of interleukin 6 (IL-6) (53). Not only is IL-6 critical for B-cell function, but its signaling is deregulated in KSHV-associated malignancies. Collectively, KSHV latent genes might compensate for a lack of IL-6 in early B-cell development.

Given that LL mice do not display lymphomas until they age (51), it was suspected that additional cellular effects were needed to accelerate lymphomagenesis. One candidate was MYC deregulation, which occurs frequently in lymphomas. To test this hypothesis, LL transgenic mice were crossed to iMYCCα transgenic mice (54), in which the MYC coding region was inserted into the IgG Cα locus under the control of its natural promoter and the Eα enhancer. The iMYCCα/LL mice develop lymphomas that resemble PEL at a low rate and, like LL mice, after a long latency. This allowed study of the synergism between MYC and the KSHV LL. iMYCCα/LL mice display higher frequencies of plasmablasts and plasma cells, and more proliferation and GC formation, than iMYCCα mice or LL mice. Moreover, double transgenic mice develop lymphomas at a higher rate. Together, these findings suggest that MYC and the KSHV LL cooperate to promote KSHV-induced lymphomagenesis.

An additional LL transgenic mouse model is available in which the LL is expressed (i) in a BALB/c background and (ii) in the absence of the FCγRIIB inhibitory receptor (55). BALB/LL mice develop hyperglobulinemia, plasmacytosis, and B-cell hyperplasia. In addition, in accord with the lower B-cell activation threshold induced by the LL, these mice exhibit an augmented antibody response against a secondary viral infection.

MOUSE MODELS OF KSHV-ENCODED LYTIC ONCOPROTEINS

Under certain still-elusive physiological conditions, KSHV undergoes lytic reactivation. Initially, it was thought that only KSHV latent proteins contributed to tumorigenesis, while lytic proteins contributed to the production of infectious virions. However, it was found that latently KSHV infected cells sometimes express low levels of lytic genes in the absence of full-blown lytic replication. Additionally, abortive replication can lead to the expression of lytic genes. Furthermore, lytic genes may be expressed alone in the context of certain environmental stimuli, e.g., hypoxia. Interestingly, many KSHV lytic proteins display oncogenic properties, such as induction of cell proliferation, inhibition of apoptosis, and evasion of immune responses, suggesting a role in tumorigenesis. This gave rise to the concept that lytic proteins might contribute to tumor progression in a paracrine fashion. Several transgenic mouse models, each expressing one lytic gene, confirm the oncogenic potential of such proteins and are discussed below (Fig. 1).

vIL-6.KSHV encodes a structural homolog of mouse IL-6 (mIL-6) and human IL-6 (hIL-6). It is expressed from ORF K2 during lytic replication and at low levels during latency. Viral IL-6 (vIL-6) supports the proliferation of a mouse B-cell hybridoma and a human myeloma cell line, and it is expressed in all three KSHV-associated cancers (2, 56–58). vIL-6 activates the JAK/STAT signaling pathway similarly to hIL-6. However, it requires only one receptor subunit, gp130, rather than both the gp130 and IL-6Rα subunits (59). Stable expression of vIL-6 in NIH 3T3 cells induces hematopoiesis, plasmacytosis, and angiogenesis when the cells are injected into athymic mice (60).

vIL-6 transgenic mice were generated by expressing a codon-optimized vIL-6 gene under the control of the major histocompatibility complex (MHC) class I H-2 promoter in the B6D2 strain (61). vIL-6 is detected in mouse serum, with intolerably high concentrations (>130 ng/ml) resulting in death/euthanasia. B6D2/vIL-6 mice were crossed to C57BL/6 mice, giving rise to vIL-6 mice, which express vIL-6 in serum and in various tissues. At the age of 15 weeks, vIL-6 transgenic mice develop enlarged spleens and lymph nodes. While no malignant tumors are observed, vIL-6 mice show a massive accumulation of plasma cells, as well as enlarged B-cell follicles with hyperplastic GCs and vascular proliferation, which are hallmarks of plasma cell-type MCD. The spleens of vIL-6 mice show increased extramedullary hematopoiesis and plasma cell hyperplasia, which are also characteristic of MCD. GCs in lymph nodes show high expression of the proliferation marker Ki67, and phosphorylation of STAT3 is observed in the lymph nodes and spleens of vIL-6 mice. Taking these findings together, expression of vIL-6 in vivo leads to the induction of symptoms that resemble human MCD. Moreover, vIL-6 induces high expression levels of mIL-6, which has been shown to be essential for MCD-like symptom development in mice.

Since vIL-6 on the C57BL/6 genetic background acts as a weak oncogene, this transgenic model was refined by crossing vIL-6 mice with BALB/c mice (vIL-6/BALBc mice) (62), which are highly susceptible to neoplasms of plasmablasts and plasma cells (63). These mice were further intercrossed with C.iMYCΔEμ mice, which have deregulated expression of the oncoprotein MYC. vIL-6/BALBc mice show much faster proliferation of MCD-like plasma cells, with a higher tumor incidence (70%), than vIL-6 mice, while vIL-6/BALBc mice with deregulated MYC expression show much faster tumor development and a 100% tumor incidence. Tumors consist mostly of plasmablastic neoplasms similar to PEL, plasmablastic lymphoma (PBL), and immunoglobulin-producing, extramedullary plasmablastic plasma cell myeloma (PBM).

K1.The K1 protein is a viral immunoreceptor encoded by ORF-K1 (64, 65). It is expressed at high levels during the lytic stage of viral replication and at low levels during latency (66, 67).

The K1 protein has structural and functional similarity to the BCR and can activate B cells (64, 68). When K1 is stimulated, it recruits and activates signaling molecules such as the tyrosine kinase Lyn, the spleen tyrosine kinase (SYK), and the p85α subunit of phosphatidylinositol 3-kinase (PI3K) (69). K1 also induces the downregulation of BCR surface expression (70), a phenomenon observed in several cancers (71). Thus, K1, and not the BCR, controls the signaling environment of infected B cells, providing a survival advantage to virus-infected cells. K1 also promotes cell survival by activating the PI3K/Akt/mammalian target of rapamycin (mTOR) pathway (72, 73) and by protecting cells from Fas-mediated apoptosis (74, 75) in a heat shock protein 90 (HSP90)- and HSP40-dependent fashion (76). In addition, K1 induces cell migration and angiogenesis by inducing the upregulation of cytokines and angiogenic factors (68, 69, 77). Given the ability of K1 to induce multiple signaling pathways, it is not surprising that K1 plays a role in KSHV tumorigenesis. In cell culture, K1 immortalizes primary endothelial cells and transforms rodent fibroblasts (65, 66). In addition, marmosets develop lymphomas when inoculated with a recombinant herpesvirus saimiri (HVS) expressing the K1 protein instead of the saimiri transforming protein (STP) (65), which is essential for HVS oncogenicity (78). While K1 shares no amino acid sequence similarity with STP, the latter is located at an equivalent position in the HVS genome.

To further investigate the oncogenic properties of K1 in vivo, a transgenic mouse model was generated in which the K1 ORF is ubiquitously expressed under the transcriptional control of the simian virus 40 (SV40) promoter (79). B and T lymphocytes isolated from 6- to 8-month-old K1 transgenic mice show higher mRNA expression levels of NF-κB and basic fibroblast growth factor (bFGF) than wild-type mice. In contrast, K1 mice have decreased IL-12 transcripts in their B cells and lower levels of IL-12 in serum. In addition, K1 induces activation not only of NF-κB, but also of the B-cell transcription factor Oct-2. A majority of K1 mice develop hyperplasia and splenomegaly at the ages of 8 and 10 months, respectively (80). In addition, splenocytes from K1 mice display higher levels of IL-6 and CXCR5 mRNAs. At the age of 18 months, 87.5% of K1 mice show signs of lymphoid hyperplasia, with 25% having confirmed lymphoma and 50% showing abdominal and/or hepatic tumors. In accordance with the inhibition of Fas-mediated apoptosis by K1 in cell culture models, splenocytes from 6-month-old transgenic mice are significantly less responsive to Fas activation than those from wild-type mice. K1-induced tumor growth is also dependent on VEGF production and NF-κB activation, since both are necessary for the expansion of K1 lymphoma cells injected into nude mice (81).

ORF36/vPK.Viral protein kinase (vPK) is a serine-threonine kinase encoded by KSHV ORF36. (82). The subcellular localization of vPK is predominantly nuclear, and it is expressed as an early-late gene during the lytic stage of KSHV infection. However, its mRNA has been found in 59% of KS biopsy specimens (83). vPK expression can be induced by hypoxia, since its promoter contains hypoxia-inducible elements (84).

vPK alters many cellular signaling pathways by phosphorylating a myriad of cellular substrates. Mitogen-activated kinases 4 and 7 (MKK4 and MKK7) are phosphorylated by vPK, resulting in the phosphorylation of c-Jun N-terminal kinase (JNK) and the subsequent phosphorylation of c-Jun transcription factor (85). One study suggests that vPK does not induce the DDR, given the absence of phosphorylation of the histone protein H2AX in vPK-expressing cells (86). However, vPK coimmunoprecipitates with and phosphorylates the histone acetyltransferase TIP60 (87), which is thought to be a tumor suppressor that participates in the oncogene-induced DDR (88).

vPK has been shown to have functional similarities to CDKs. For example, vPK phosphorylates the CDK substrates retinoblastoma and lamin A/C (89). In addition to functional similarity to CDKs, vPK mimics ribosomal protein S6 kinase (S6KB1) structurally and functionally, sharing several phosphorylation substrates, including ribosomal S6 protein (90). In the same study, vPK was shown to phosphorylate eukaryotic initiation factor 4E (eIF4E). vPK-induced S6 phosphorylation enhances several hallmarks of cancer, including cellular proliferation, protein synthesis, anchorage independence, and angiogenesis.

Two transgenic mouse lines were generated in which the vPK transgene is expressed under the control of a ubiquitin promoter (91). vPK expression alone increases the incidence of B-cell hyperproliferative disorder and/or a high-grade B-cell non-Hodgkin lymphoma (NHL) 8-fold over that for age-matched wild-type mice. Although vPK is expressed ubiquitously, the mice develop only B-cell tumors. vPK mice show increased B-cell activation in the absence of antigen stimulation and increased levels of IL-1β and IL-12 p40. Lymphomas from vPK mice consist predominantly of B220+ GL-7+ IgM– IgD– B cells, suggesting a GC origin. In addition, lymphomas express vPK, display monoclonal or polyclonal IgH rearrangements, and show robust expression of phosphorylated S6 and eIF4E, suggesting that vPK phosphorylation of these cellular substrates is important for tumorigenesis.

vGPCR.KSHV encodes a constitutively active G protein-coupled receptor (vGPCR) (26) functionally homologous to the human interleukin 8 receptor (IL-8R). It binds to the CXC chemokine IL-8 and signals through phosphoinositide phospholipase C (92). In contrast to IL-8R, vGPCR binds both CXC and CC chemokines (92). vGPCR can transform endothelial cells (93, 94) due to its deregulation of cellular signaling via a variety of ligand-independent paracrine and autocrine mechanisms (95). vGPCR directly modulates several intracellular signaling pathways, such as the JNK, extracellular signal-regulated kinase (ERK), p38, PI3K/AKT/mTOR, and WNT/β-catenin pathways (93, 94, 96–101). Subsequently, vGPCR activates key transcription factors, such as NF-κB, nuclear factor of activated T cells (NFAT), AP-1, and CREB (96, 102–106). Such modulation results in increased cell survival and proliferation and in the expression of angiogenic and inflammatory molecules such as CCL2 and VEGF. These cytokines and growth factors can activate receptors in neighboring infected cells, amplifying vGPCR signaling (92, 93, 107–111).

To date, four vGPCR transgenic mouse models have been developed. The first model expresses the vGPCR gene under the control of the T-cell-specific CD2 promoter, resulting in the expression of vGPCR predominantly in T and NK cells (112). By 30 to 90 days after birth, all vGPCR mice develop KS-like lesions with strong expression of the endothelial cell marker CD34, suggesting that vGPCR acts in a paracrine fashion. The second mouse model employs mice expressing the avian retroviral receptor under the control of the endothelial-cell-specific TIE2 promoter (113). This allows for the infection of mouse endothelial cells with avian leukosis virus (ALV) expressing a KSHV gene. vGPCR-expressing mice show a decreased median survival from that of uninfected mice. Mice infected with 107 copies of vGPCR-expressing ALV die at an incidence of 100% within 6 weeks after infection, and numerous microscopic tumors develop. Mice infected with 105 copies of vGPCR-expressing ALV develop angiogenic tumors by the age of 4 months. vGPCR-induced pathogenesis is abolished when an inactive vGPCR mutant is transduced. The third mouse model expresses vGPCR ubiquitously under the control of the SV40 promoter (114). These mice develop internal and/or external tumors at an incidence of 30% to 40%. The tumors are highly angiogenic, resembling KS given the presence of spindle cells, CD31+ cells, and expression of VEGF-C. In the fourth vGPCR model, vGPCR is conditionally expressed to bypass the potential adverse effects of vGPCR expression prior to birth (108, 115). Induction of vGPCR with doxycycline results in highly angiogenic lesions with spindle cells of endothelial origin, similar to KS. Angiogenic factors are significantly upregulated in vGPCR+ cells. In addition, proangiogenic chemokine receptors and chemokines are upregulated in doxycycline-induced mice only. The development of this angioproliferative disease is independent of T and B cells, and the continued expression of vGPCR is required.

CONCLUSION

KSHV encodes multiple proteins that potentially contribute to KSHV-related human cancers. The modulation of cellular signaling pathways by KSHV oncogenic proteins allows infected cells to bypass growth suppressors and apoptotic signals, often leading to cell proliferation and immortality. In addition, KSHV oncogenic proteins lead to other hallmarks of cancer, such as angiogenesis, immune evasion, and genome instability. One possible reason why KSHV encodes several oncoproteins is so that the virus can keep the infected cell alive and thereby itself survive and be transmitted to other hosts. However, the pathways that are required to keep the infected cell alive are the very same pathways that go awry in cancer. Hence, although the goal of the virus is to survive in its host cell, the unintended consequence is the development of cancer in the host.

While many of the oncogenic properties of these viral proteins have been discovered and further studied in a variety of cell culture models, the oncogenic potential of just a few have been confirmed in vivo by means of the transgenic mouse models discussed here. In such models, we can see how the expression of a multifunctional KSHV protein initiates the multistep process of cancer development, with mice showing symptoms that mimic KSHV-associated malignancies. Although these transgenic mouse models are an excellent tool for studying the mechanisms by which KSHV induces oncogenesis and the functional role of each protein, no model yet entirely recapitulates KSHV-associated cancers in humans. One reason is that each of these proteins is expressed individually or in a small group, as in the LL mice, and as outlined by this review, each protein contributes to KSHV tumorigenesis in a unique fashion. In addition, given that KSHV is a strictly human pathogen, some human factors present or absent in mice can affect disease development. Despite such caveats, these transgenic mouse models can be used not only to study how KSHV leads to tumorigenesis but also to evaluate drugs that target KSHV proteins with the aim of treating KSHV-related cancers.

ACKNOWLEDGMENTS

We thank members of the Damania lab for helpful edits. We apologize for not including the work of many colleagues due to the limited word count.

Our lab is supported by Public Health Service grants CA096500, CA019014, DE028211, and CA163217. B.D. is a Leukemia and Lymphoma Society Scholar and a Burroughs Welcome Fund Investigator in Infectious Disease. A.G.B.C. is supported by NIH grant T32CA009156.

FOOTNOTES

    • Received 23 February 2019.
    • Accepted 6 March 2019.
    • Accepted manuscript posted online 13 March 2019.
  • Copyright © 2019 American Society for Microbiology.

All Rights Reserved.

REFERENCES

  1. 1.↵
    1. Mesri EA,
    2. Feitelson MA,
    3. Munger K
    . 2014. Human viral oncogenesis: a cancer hallmarks analysis. Cell Host Microbe 15:266–282. doi:10.1016/j.chom.2014.02.011.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Chang Y,
    2. Cesarman E,
    3. Pessin MS,
    4. Lee F,
    5. Culpepper J,
    6. Knowles DM,
    7. Moore PS
    . 1994. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 266:1865–1869. doi:10.1126/science.7997879.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    1. Cesarman E,
    2. Chang Y,
    3. Moore PS,
    4. Said JW,
    5. Knowles DM
    . 1995. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity–based lymphomas. N Engl J Med 332:1186–1191. doi:10.1056/NEJM199505043321802.
    OpenUrlCrossRefPubMedWeb of Science
  4. 4.↵
    1. Soulier J,
    2. Grollet L,
    3. Oksenhendler E,
    4. Cacoub P,
    5. Cazals-Hatem D,
    6. Babinet P,
    7. D’Agay MF,
    8. Clauvel JP,
    9. Raphael M,
    10. Degos L
    . 1995. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman’s disease. Blood 86:1276–1280.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Yarchoan R,
    2. Uldrick TS
    . 2018. HIV-associated cancers and related diseases. N Engl J Med 378:1029–1041. doi:10.1056/NEJMra1615896.
    OpenUrlCrossRef
  6. 6.↵
    IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. 2012. A review of human carcinogens. Biological agents. IARC Monogr Eval Carcinog Risks Hum 100(Part B):1–441.
    OpenUrlPubMed
  7. 7.↵
    1. Ballestas ME,
    2. Chatis PA,
    3. Kaye KM
    . 1999. Efficient persistence of extrachromosomal KSHV DNA mediated by latency- associated nuclear antigen. Science 284:641–644. doi:10.1126/science.284.5414.641.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    1. Cotter MA,
    2. Robertson ES
    . 1999. The latency-associated nuclear antigen tethers the Kaposi’s sarcoma-associated herpesvirus genome to host chromosomes in body cavity-based lymphoma cells. Virology 264:254–264. doi:10.1006/viro.1999.9999.
    OpenUrlCrossRefPubMedWeb of Science
  9. 9.↵
    1. Garber AC,
    2. Shu MA,
    3. Hu J,
    4. Renne R
    . 2001. DNA binding and modulation of gene expression by the latency-associated nuclear antigen of Kaposi’s sarcoma-associated herpesvirus. J Virol 75:7882–7892. doi:10.1128/JVI.75.17.7882-7892.2001.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    1. Radkov SA,
    2. Kellam P,
    3. Boshoff C
    . 2000. The latent nuclear antigen of Kaposi sarcoma-associated herpesvirus targets the retinoblastoma–E2F pathway and with the oncogene Hras transforms primary rat cells. Nat Med 6:1121–1127. doi:10.1038/80459.
    OpenUrlCrossRefPubMedWeb of Science
  11. 11.↵
    1. Friborg J,
    2. Kong W,
    3. Hottiger MO,
    4. Nabel GJ
    . 1999. p53 inhibition by the LANA protein of KSHV protects against cell death. Nature 402:889–894. doi:10.1038/47266.
    OpenUrlCrossRefPubMedWeb of Science
  12. 12.↵
    1. Schulz TF,
    2. Santag S,
    3. Jäger W,
    4. Karsten CB,
    5. Kati S,
    6. Pietrek M,
    7. Steinemann D,
    8. Sarek G,
    9. Ojala PM
    . 2013. Recruitment of the tumour suppressor protein p73 by Kaposi’s sarcoma herpesvirus latent nuclear antigen contributes to the survival of primary effusion lymphoma cells. Oncogene 32:3676–3685. doi:10.1038/onc.2012.385.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Lan K,
    2. Verma SC,
    3. Murakami M,
    4. Bajaj B,
    5. Kaul R,
    6. Robertson ES
    . 2007. Kaposi’s sarcoma herpesvirus-encoded latency-associated nuclear antigen stabilizes intracellular activated Notch by targeting the Sel10 protein. Proc Natl Acad Sci U S A 104:16287–16292. doi:10.1073/pnas.0703508104.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    1. Roupelieva M,
    2. Griffiths SJ,
    3. Kremmer E,
    4. Meisterernst M,
    5. Viejo-Borbolla A,
    6. Schulz T,
    7. Haas J
    . 2010. Kaposi’s sarcoma-associated herpesvirus Lana-1 is a major activator of the serum response element and mitogen-activated protein kinase pathways via interactions with the Mediator complex. J Gen Virol 91:1138–1149. doi:10.1099/vir.0.017715-0.
    OpenUrlCrossRefPubMedWeb of Science
  15. 15.↵
    1. Lu J,
    2. Verma SC,
    3. Murakami M,
    4. Cai Q,
    5. Kumar P,
    6. Xiao B,
    7. Robertson ES
    . 2009. Latency-associated nuclear antigen of Kaposi’s sarcoma-associated herpesvirus (KSHV) upregulates survivin expression in KSHV-associated B-lymphoma cells and contributes to their proliferation. J Virol 83:7129–7141. doi:10.1128/JVI.00397-09.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Fujimuro M,
    2. Wu FY,
    3. ApRhys C,
    4. Kajumbula H,
    5. Young DB,
    6. Hayward GS,
    7. Hayward SD
    . 2003. A novel viral mechanism for dysregulation of β-catenin in Kaposi’s sarcoma-associated herpesvirus latency. Nat Med 9:300–306. doi:10.1038/nm829.
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.↵
    1. Bubman D,
    2. Guasparri I,
    3. Cesarman E
    . 2007. Deregulation of c-Myc in primary effusion lymphoma by Kaposi’s sarcoma herpesvirus latency-associated nuclear antigen. Oncogene 26:4979–4986. doi:10.1038/sj.onc.1210299.
    OpenUrlCrossRefPubMedWeb of Science
  18. 18.↵
    1. Liu J,
    2. Martin HJ,
    3. Liao G,
    4. Hayward SD
    . 2007. The Kaposi’s sarcoma-associated herpesvirus LANA protein stabilizes and activates c-Myc. J Virol 81:10451–10459. doi:10.1128/JVI.00804-07.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. An J,
    2. Sun Y,
    3. Rettig MB
    . 2004. Transcriptional coactivation of c-Jun by the KSHV-encoded LANA. Blood 103:222–228. doi:10.1182/blood-2003-05-1538.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Sun Z,
    2. Xiao B,
    3. Jha HC,
    4. Lu J,
    5. Banerjee S,
    6. Robertson ES
    . 2014. Kaposi’s sarcoma-associated herpesvirus-encoded LANA can induce chromosomal instability through targeted degradation of the mitotic checkpoint kinase Bub1. J Virol 88:7367–7378. doi:10.1128/JVI.00554-14.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    1. Thakker S,
    2. Strahan RC,
    3. Scurry AN,
    4. Uppal T,
    5. Verma SC
    . 2018. KSHV LANA upregulates the expression of epidermal growth factor like domain 7 to promote angiogenesis. Oncotarget 9:1210–1228. doi:10.18632/oncotarget.23456.
    OpenUrlCrossRef
  22. 22.↵
    1. Watanabe T,
    2. Sugaya M,
    3. Atkins AM,
    4. Aquilino EA,
    5. Yang A,
    6. Borris DL,
    7. Brady J,
    8. Blauvelt A
    . 2003. Kaposi’s sarcoma-associated herpesvirus latency-associated nuclear antigen prolongs the life span of primary human umbilical vein endothelial cells. J Virol 77:6188–6196. doi:10.1128/JVI.77.11.6188-6196.2003.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Fakhari FD,
    2. Jeong JH,
    3. Kanan Y,
    4. Dittmer DP
    . 2006. The latency-associated nuclear antigen of Kaposi sarcoma-associated herpesvirus induces B cell hyperplasia and lymphoma. J Clin Invest 116:735–742. doi:10.1172/JCI26190.
    OpenUrlCrossRefPubMedWeb of Science
  24. 24.↵
    1. Jeong JH,
    2. Hines-Boykin R,
    3. Ash JD,
    4. Dittmer DP
    . 2002. Tissue specificity of the Kaposi’s sarcoma-associated herpesvirus latent nuclear antigen (LANA/orf73) promoter in transgenic mice. J Virol 76:11024–11032. doi:10.1128/JVI.76.21.11024-11032.2002.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    1. Sin S-H,
    2. Fakhari FD,
    3. Dittmer DP
    . 2010. The viral latency-associated nuclear antigen augments the B-cell response to antigen in vivo. J Virol 84:10653–10660. doi:10.1128/JVI.00848-10.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. Cesarman E,
    2. Nador R,
    3. Bai F,
    4. Bohenzky R,
    5. Russo J,
    6. Moore P,
    7. Chang Y,
    8. Knowles D
    . 1996. Kaposi’s sarcoma-associated herpesvirus contains G protein-coupled receptor and cyclin D homologs which are expressed in Kaposi’s sarcoma and malignant lymphoma. J Virol 70:8218–8223.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Yuan C,
    2. Moore PS,
    3. Talbot SJ,
    4. Boshoff CH,
    5. Zarkowska T,
    6. Godden- Kent D,
    7. Paterson H,
    8. Weiss RA,
    9. Mittnacht S
    . 1996. Cyclin encoded by KS herpesvirus. Nature 382:410.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Verschuren EW,
    2. Jones N,
    3. Evan GI
    . 2004. The cell cycle and how it is steered by Kaposi’s sarcoma-associated herpesvirus cyclin. J Gen Virol 85(Part 6):1347–1361. doi:10.1099/vir.0.79812-0.
    OpenUrlCrossRefPubMedWeb of Science
  29. 29.↵
    1. Li M,
    2. Lee H,
    3. Yoon D,
    4. Albrecht J,
    5. Fleckenstein B,
    6. Neipel F,
    7. Jung J
    . 1997. Kaposi’s sarcoma-associated herpesvirus encodes a functional cyclin. J Virol 71:1984–1991.
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    1. Godden-Kent D,
    2. Talbot SJ,
    3. Boshoff C,
    4. Chang Y,
    5. Moore P,
    6. Weiss RA,
    7. Mittnacht S
    . 1997. The cyclin encoded by Kaposi’s sarcoma-associated herpesvirus stimulates cdk6 to phosphorylate the retinoblastoma protein and histone H1. J Virol 71:4193–4198.
    OpenUrlAbstract/FREE Full Text
  31. 31.↵
    1. Swanton C,
    2. Mann DJ,
    3. Fleckenstein B,
    4. Neipel F,
    5. Peters G,
    6. Jones N
    . 1997. Herpes viral cyclin/Cdk6 complexes evade inhibition by CDK inhibitor proteins. Nature 390:184–187. doi:10.1038/36606.
    OpenUrlCrossRefPubMedWeb of Science
  32. 32.↵
    1. Ellis M,
    2. Chew YP,
    3. Fallis L,
    4. Freddersdorf S,
    5. Boshoff C,
    6. Weiss RA,
    7. Lu X,
    8. Mittnacht S
    . 1999. Degradation of p27Kip cdk inhibitor triggered by Kaposi’s sarcoma virus cyclin-cdk6 complex. EMBO J 18:644–653. doi:10.1093/emboj/18.3.644.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    1. Ojala PM,
    2. Tiainen M,
    3. Salven P,
    4. Veikkola T,
    5. Castaños-Vélez E,
    6. Sarid R,
    7. Biberfeld P,
    8. Mäkelä TP
    . 1999. Kaposi’s sarcoma-associated herpesvirus-encoded v-cyclin triggers apoptosis in cells with high levels of cyclin-dependent kinase 6. Cancer Res 59:4984–4989.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    1. Ojala PM,
    2. Yamamoto K,
    3. Castaños-Vélez E,
    4. Biberfeld P,
    5. Korsmeyer SJ,
    6. Mäkelä TP
    . 2000. The apoptotic v-cyclin-CDK6 complex phosphorylates and inactivates Bcl-2. Nat Cell Biol 2:819–825. doi:10.1038/35041064.
    OpenUrlCrossRefPubMedWeb of Science
  35. 35.↵
    1. Koopal S,
    2. Furuhjelm JH,
    3. Järviluoma A,
    4. Jäämaa S,
    5. Pyakurel P,
    6. Pussinen C,
    7. Wirzenius M,
    8. Biberfeld P,
    9. Alitalo K,
    10. Laiho M,
    11. Ojala PM
    . 2007. Viral oncogene-induced DNA damage response is activated in Kaposi sarcoma tumorigenesis. PLoS Pathog 3:1348–1360. doi:10.1371/journal.ppat.0030140.
    OpenUrlCrossRefPubMed
  36. 36.↵
    1. Verschuren EW,
    2. Klefstrom J,
    3. Evan GI,
    4. Jones N
    . 2002. The oncogenic potential of Kaposi’s sarcoma-associated herpesvirus cyclin is exposed by p53 loss in vitro and in vivo. Cancer Cell 2:229–241. doi:10.1016/S1535-6108(02)00123-X.
    OpenUrlCrossRefPubMedWeb of Science
  37. 37.↵
    1. Verschuren EW,
    2. Hodgson JG,
    3. Gray JW,
    4. Kogan S,
    5. Jones N,
    6. Evan GI
    . 2004. The role of p53 in suppression of KSHV cyclin-induced lymphomagenesis. Cancer Res 64:581–589. doi:10.1158/0008-5472.CAN-03-1863.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Sugaya M,
    2. Watanabe T,
    3. Yang A,
    4. Starost MF,
    5. Kobayashi H,
    6. Atkins AM,
    7. Borris DL,
    8. Hanan EA,
    9. Schimel D,
    10. Bryant MA,
    11. Roberts N,
    12. Skobe M,
    13. Staskus KA,
    14. Kaldis P,
    15. Blauvelt A
    . 2005. Lymphatic dysfunction in transgenic mice expressing KSHV k-cyclin under the control of the VEGFR-3 promoter. Blood 105:2356–2363. doi:10.1182/blood-2004-08-3364.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Talbot SJ,
    2. Weiss RA,
    3. Kellam P,
    4. Boshoff C
    . 1999. Transcriptional analysis of human herpesvirus-8 open reading frames 71, 72, 73, K14, and 74 in a primary effusion lymphoma cell line. Virology 257:84–94. doi:10.1006/viro.1999.9672.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Stürzl M,
    2. Hohenadl C,
    3. Zietz C,
    4. Castanos-Velez E,
    5. Wunderlich A,
    6. Ascherl G,
    7. Biberfeld P,
    8. Monini P,
    9. Browning PJ,
    10. Ensoli B
    . 1999. Expression of K13/v-FLIP gene of human herpesvirus 8 and apoptosis in Kaposi’s sarcoma spindle cells. J Natl Cancer Inst 91:1725–1733. doi:10.1093/jnci/91.20.1725.
    OpenUrlCrossRefPubMedWeb of Science
  41. 41.↵
    1. Shisler JL
    . 2014. Viral and cellular FLICE-inhibitory proteins: a comparison of their roles in regulating intrinsic immune responses. J Virol 88:6539–6541. doi:10.1128/JVI.00276-14.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Irmler M,
    2. Thome M,
    3. Hahne M,
    4. Schneider P,
    5. Hofmann K,
    6. Steiner V,
    7. Bodmer JL,
    8. Schröter M,
    9. Burns K,
    10. Mattmann C,
    11. Rimoldi D,
    12. French LE,
    13. Tschopp J
    . 1997. Inhibition of death receptor signals by cellular FLIP. Nature 388:190–195. doi:10.1038/40657.
    OpenUrlCrossRefPubMedWeb of Science
  43. 43.↵
    1. Thome M,
    2. Schneider P,
    3. Hofmann K,
    4. Fickenscher H,
    5. Meinl E,
    6. Neipel F,
    7. Mattmann C,
    8. Burns K,
    9. Bodmer JL,
    10. Schröter M,
    11. Scaffidi C,
    12. Krammer PH,
    13. Peter ME,
    14. Tschopp J
    . 1997. Viral FLICE-inhibitory proteins (FLIPs) prevent apoptosis induced by death receptors. Nature 386:517–521. doi:10.1038/386517a0.
    OpenUrlCrossRefPubMedWeb of Science
  44. 44.↵
    1. Bertin J,
    2. Armstrong RC,
    3. Ottilie S,
    4. Martin DA,
    5. Wang Y,
    6. Banks S,
    7. Wang GH,
    8. Senkevich TG,
    9. Alnemri ES,
    10. Moss B,
    11. Lenardo MJ,
    12. Tomaselli KJ,
    13. Cohen JI
    . 1997. Death effector domain-containing herpesvirus and poxvirus proteins inhibit both Fas- and TNFR1-induced apoptosis. Proc Natl Acad Sci U S A 94:1172–1176. doi:10.1073/pnas.94.4.1172.
    OpenUrlAbstract/FREE Full Text
  45. 45.↵
    1. Lee JS,
    2. Li Q,
    3. Lee JY,
    4. Lee SH,
    5. Jeong JH,
    6. Lee HR,
    7. Chang H,
    8. Zhou FC,
    9. Gao SJ,
    10. Liang C,
    11. Jung JU
    . 2009. FLIP-mediated autophagy regulation in cell death control. Nat Cell Biol 11:1355–1362. doi:10.1038/ncb1980.
    OpenUrlCrossRefPubMedWeb of Science
  46. 46.↵
    1. Chaudhary PM,
    2. Jasmin A,
    3. Eby MT,
    4. Hood L
    . 1999. Modulation of the NF-κB pathway by virally encoded death effector domains-containing proteins. Oncogene 18:5738–5746. doi:10.1038/sj.onc.1202976.
    OpenUrlCrossRefPubMedWeb of Science
  47. 47.↵
    1. Field N,
    2. Low W,
    3. Daniels M,
    4. Howell S,
    5. Daviet L,
    6. Boshoff C,
    7. Collins M
    . 2003. KSHV vFLIP binds to IKK-γ to activate IKK. J Cell Sci 116:3721–3728. doi:10.1242/jcs.00691.
    OpenUrlAbstract/FREE Full Text
  48. 48.↵
    1. Sun Q,
    2. Zachariah S,
    3. Chaudhary PM
    . 2003. The human herpes virus 8-encoded viral FLICE-inhibitory protein induces cellular transformation via NF-κB activation. J Biol Chem 278:52437–52445. doi:10.1074/jbc.M304199200.
    OpenUrlAbstract/FREE Full Text
  49. 49.↵
    1. Chugh P,
    2. Matta H,
    3. Schamus S,
    4. Zachariah S,
    5. Kumar A,
    6. Richardson JA,
    7. Smith AL,
    8. Chaudhary PM
    . 2005. Constitutive NF-κB activation, normal Fas-induced apoptosis, and increased incidence of lymphoma in human herpes virus 8 K13 transgenic mice. Proc Natl Acad Sci U S A 102:12885–12890. doi:10.1073/pnas.0408577102.
    OpenUrlAbstract/FREE Full Text
  50. 50.↵
    1. Ballon G,
    2. Chen K,
    3. Perez R,
    4. Tam W,
    5. Cesarman E
    . 2011. Kaposi sarcoma herpesvirus (KSHV) vFLIP oncoprotein induces B cell transdifferentiation and tumorigenesis in mice. J Clin Invest 121:1141–1153. doi:10.1172/JCI44417.
    OpenUrlCrossRefPubMedWeb of Science
  51. 51.↵
    1. Sin SH,
    2. Dittmer DP
    . 2013. Viral latency locus augments B-cell response in vivo to induce chronic marginal zone enlargement, plasma cell hyperplasia, and lymphoma. Blood 121:2952–2963. doi:10.1182/blood-2012-03-415620.
    OpenUrlAbstract/FREE Full Text
  52. 52.↵
    1. Sin S-H,
    2. Kim YB,
    3. Dittmer DP
    . 2013. Latency locus complements microRNA 155 deficiency in vivo. J Virol 87:11908–11911. doi:10.1128/JVI.01620-13.
    OpenUrlAbstract/FREE Full Text
  53. 53.↵
    1. Sin S-H,
    2. Kang SA,
    3. Kim Y,
    4. Eason A,
    5. Tan K,
    6. An H,
    7. Dittmer DP
    . 2016. Kaposi’s sarcoma-associated herpesvirus latency locus compensates for interleukin-6 in initial B cell activation. J Virol 90:2150–2154. doi:10.1128/JVI.02456-15.
    OpenUrlAbstract/FREE Full Text
  54. 54.↵
    1. Sin SH,
    2. Kim Y,
    3. Eason A,
    4. Dittmer DP
    . 2015. KSHV latency locus cooperates with Myc to drive lymphoma in mice. PLoS Pathog 11:e1005135. doi:10.1371/journal.ppat.1005135.
    OpenUrlCrossRef
  55. 55.↵
    1. Sin S-H,
    2. Eason AB,
    3. Bigi R,
    4. Kim Y,
    5. Kang S,
    6. Tan K,
    7. Seltzer TA,
    8. Venkataramanan R,
    9. An H,
    10. Dittmer DP
    . 2018. Kaposi’s sarcoma-associated herpesvirus latency locus renders B cells hyperresponsive to secondary infections. J Virol 92:e01138-18. doi:10.1128/JVI.01138-18.
    OpenUrlAbstract/FREE Full Text
  56. 56.↵
    1. Moore PS,
    2. Boshoff C,
    3. Weiss RA,
    4. Chang Y
    . 1996. Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV. Science 274:1739–1744. doi:10.1126/science.274.5293.1739.
    OpenUrlAbstract/FREE Full Text
  57. 57.↵
    1. Neipel F,
    2. Albrecht J-C,
    3. Ensser A,
    4. Huang Y-Q,
    5. Li JJ,
    6. Friedman-Kien AE,
    7. Fleckenstein B
    . 1997. Human herpesvirus 8 encodes a homolog of interleukin-6. J Virol 71:839–842.
    OpenUrlAbstract/FREE Full Text
  58. 58.↵
    1. Molden J,
    2. Chang Y,
    3. You Y,
    4. Moore PS,
    5. Goldsmith MA
    . 1997. A Kaposi’s sarcoma-associated herpesvirus-encoded cytokine homolog (vIL-6) activates signaling through the shared gp130 receptor subunit. J Biol Chem 272:19625–19631. doi:10.1074/jbc.272.31.19625.
    OpenUrlAbstract/FREE Full Text
  59. 59.↵
    1. Nicholas J,
    2. Ruvolo VR,
    3. Burns WH,
    4. Sandford G,
    5. Wan X,
    6. Ciufo D,
    7. Hendrickson SB,
    8. Guo H-G,
    9. Hayward GS,
    10. Reitz MS
    . 1997. Kaposi’s sarcoma-associated human herpesvirus-8 encodes homologues of macrophage inflammatory protein-1 and interleukin-6. Nat Med 3:287–292. doi:10.1038/nm0397-287.
    OpenUrlCrossRefPubMedWeb of Science
  60. 60.↵
    1. Aoki Y,
    2. Jaffe ES,
    3. Chang Y,
    4. Jones K,
    5. Teruya-Feldstein J,
    6. Moore PS,
    7. Tosato G
    . 1999. Angiogenesis and hematopoiesis induced by Kaposi’s sarcoma-associated herpesvirus-encoded interleukin-6. Blood 93:4034–4043.
    OpenUrlAbstract/FREE Full Text
  61. 61.↵
    1. Suthaus J,
    2. Stuhlmann-Laeisz C,
    3. Van Tompkins S,
    4. Rosean TR,
    5. Klapper W,
    6. Tosato G,
    7. Janz S,
    8. Scheller J,
    9. Rose-John S
    . 2012. HHV-8-encoded viral IL-6 collaborates with mouse IL-6 in the development of multicentric Castleman disease in mice. Blood 119:5173–5181. doi:10.1182/blood-2011-09-377705.
    OpenUrlAbstract/FREE Full Text
  62. 62.↵
    1. Rosean TR,
    2. Holman CJ,
    3. Tompkins VS,
    4. Jing X,
    5. Krasowski MD,
    6. Rose-John S,
    7. Janz S
    . 2016. KSHV-encoded vIL-6 collaborates with deregulated c-Myc to drive plasmablastic neoplasms in mice. Blood Cancer J 6:e398. doi:10.1038/bcj.2016.6.
    OpenUrlCrossRef
  63. 63.↵
    1. Zhang K,
    2. Kagan D,
    3. DuBois W,
    4. Robinson R,
    5. Bliskovsky V,
    6. Vass WC,
    7. Zhang S,
    8. Mock BA
    . 2009. Mndal, a new interferon-inducible family member, is highly polymorphic, suppresses cell growth, and may modify plasmacytoma susceptibility. Blood 114:2952–2960. doi:10.1182/blood-2009-01-198812.
    OpenUrlAbstract/FREE Full Text
  64. 64.↵
    1. Lee H,
    2. Guo J,
    3. Li M,
    4. Choi J-K,
    5. DeMaria M,
    6. Rosenzweig M,
    7. Jung JU
    . 1998. Identification of an immunoreceptor tyrosine-based activation motif of K1 transforming protein of Kaposi’s sarcoma-associated herpesvirus. Mol Cell Biol 18:5219–5228. doi:10.1128/MCB.18.9.5219.
    OpenUrlAbstract/FREE Full Text
  65. 65.↵
    1. Lee H,
    2. Veazey R,
    3. Williams K,
    4. Li M,
    5. Guo J,
    6. Neipel F,
    7. Fleckenstein B,
    8. Lackner A,
    9. Desrosiers RC,
    10. Jung JU
    . 1998. Deregulation of cell growth by the K1 gene of Kaposi’s sarcoma-associated herpesvirus. Nat Med 4:435–440. doi:10.1038/nm0498-435.
    OpenUrlCrossRefPubMedWeb of Science
  66. 66.↵
    1. Wang L,
    2. Dittmer DP,
    3. Tomlinson CC,
    4. Fakhari FD,
    5. Damania B
    . 2006. Immortalization of primary endothelial cells by the K1 protein of Kaposi’s sarcoma-associated herpesvirus. Cancer Res 66:3658–3666. doi:10.1158/0008-5472.CAN-05-3680.
    OpenUrlAbstract/FREE Full Text
  67. 67.↵
    1. Chandriani S,
    2. Ganem D
    . 2010. Array-based transcript profiling and limiting-dilution reverse transcription-PCR analysis identify additional latent genes in Kaposi’s sarcoma-associated herpesvirus. J Virol 84:5565–5573. doi:10.1128/JVI.02723-09.
    OpenUrlAbstract/FREE Full Text
  68. 68.↵
    1. Lagunoff M,
    2. Majeti R,
    3. Weiss A,
    4. Ganem D
    . 1999. Deregulated signal transduction by the K1 gene product of Kaposi’s sarcoma-associated herpesvirus. Proc Natl Acad Sci U S A 96:5704–5709. doi:10.1073/pnas.96.10.5704.
    OpenUrlAbstract/FREE Full Text
  69. 69.↵
    1. Lee BS,
    2. Lee SH,
    3. Feng P,
    4. Chang H,
    5. Cho NH,
    6. Jung JU
    . 2005. Characterization of the Kaposi’s sarcoma-associatEd herpesvirus K1 signalosome. J Virol 79:12173–12184. doi:10.1128/JVI.79.19.12173-12184.2005.
    OpenUrlAbstract/FREE Full Text
  70. 70.↵
    1. Lee BS,
    2. Alvarez X,
    3. Ishido S,
    4. Lackner AA,
    5. Jung JU
    . 2000. Inhibition of intracellular transport of B cell antigen receptor complexes by Kaposi’s sarcoma-associated herpesvirus K1. J Exp Med 192:11–21. doi:10.1084/jem.192.1.11.
    OpenUrlAbstract/FREE Full Text
  71. 71.↵
    1. Chapman J,
    2. Gentles AJ,
    3. Sujoy V,
    4. Vega F,
    5. Dumur CI,
    6. Blevins TL,
    7. Bernal-Mizrachi L,
    8. Mosunjac M,
    9. Pimentel A,
    10. Zhu D,
    11. Lossos IS
    . 2015. Gene expression analysis of plasmablastic lymphoma identifies downregulation of B-cell receptor signaling and additional unique transcriptional programs. Leukemia 29:2270–2273. doi:10.1038/leu.2015.109.
    OpenUrlCrossRef
  72. 72.↵
    1. Tomlinson CC,
    2. Damania B
    . 2004. The K1 protein of Kaposi’s sarcoma-associated herpesvirus activates the Akt signaling pathway. J Virol 78:1918–1927. doi:10.1128/JVI.78.4.1918-1927.2004.
    OpenUrlAbstract/FREE Full Text
  73. 73.↵
    1. Anders PM,
    2. Zhang Z,
    3. Bhende PM,
    4. Giffin L,
    5. Damania B
    . 2016. The KSHV K1 protein modulates AMPK function to enhance cell survival. PLoS Pathog 12:e1005985. doi:10.1371/journal.ppat.1005985.
    OpenUrlCrossRef
  74. 74.↵
    1. Wang S,
    2. Wang S,
    3. Maeng H,
    4. Young DP,
    5. Prakash O,
    6. Fayad LE,
    7. Younes A,
    8. Samaniego F
    . 2007. K1 protein of human herpesvirus 8 suppresses lymphoma cell Fas-mediated apoptosis. Blood 109:2174–2182. doi:10.1182/blood-2006-02-003178.
    OpenUrlAbstract/FREE Full Text
  75. 75.↵
    1. Berkova Z,
    2. Wang S,
    3. Wise JF,
    4. Maeng H,
    5. Ji Y,
    6. Samaniego F
    . 2009. Mechanism of Fas signaling regulation by human herpesvirus 8 K1 oncoprotein. J Natl Cancer Inst 101:399–411. doi:10.1093/jnci/djn516.
    OpenUrlCrossRefPubMedWeb of Science
  76. 76.↵
    1. Wen KW,
    2. Damania B
    . 2010. Hsp90 and Hsp40/Erdj3 are required for the expression and anti-apoptotic function of KSHV K1. Oncogene 29:3532–3544. doi:10.1038/onc.2010.124.
    OpenUrlCrossRefPubMed
  77. 77.↵
    1. Wang L,
    2. Wakisaka N,
    3. Tomlinson CC,
    4. DeWire SM,
    5. Krall S,
    6. Pagano JS,
    7. Damania B
    . 2004. The Kaposi’s sarcoma-associated herpesvirus (KSHV/HHV-8) K1 protein induces expression of angiogenic and invasion factors. Cancer Res 64:2774–2781. doi:10.1158/0008-5472.CAN-03-3653.
    OpenUrlAbstract/FREE Full Text
  78. 78.↵
    1. Duboise SM,
    2. Guo J,
    3. Czajak S,
    4. Desrosiers RC,
    5. Jung JU
    . 1998. STP and Tip are essential for herpesvirus saimiri oncogenicity. J Virol 72:1308–1313.
    OpenUrlAbstract/FREE Full Text
  79. 79.↵
    1. Prakash O,
    2. Tang ZY,
    3. Peng X,
    4. Coleman R,
    5. Gill J,
    6. Farr G,
    7. Samaniego F
    . 2002. Tumorigenesis and aberrant signaling in transgenic mice expressing the human herpesvirus-8 K1 gene. J Natl Cancer Inst 94:926–935. doi:10.1093/jnci/94.12.926.
    OpenUrlCrossRefPubMedWeb of Science
  80. 80.↵
    1. Berkova Z,
    2. Wang S,
    3. Sehgal L,
    4. Patel KP,
    5. Prakash O,
    6. Samaniego F
    . 2015. Lymphoid hyperplasia and lymphoma in KSHV K1 transgenic mice. Histol Histopathol 30:559–568. doi:10.14670/HH-30.559.
    OpenUrlCrossRef
  81. 81.↵
    1. Prakash O,
    2. Swamy OR,
    3. Peng X,
    4. Tang ZY,
    5. Li L,
    6. Larson JE,
    7. Cohen JC,
    8. Gill J,
    9. Farr G,
    10. Wang S,
    11. Samaniego F
    . 2005. Activation of Src kinase Lyn by the Kaposi sarcoma-associated herpesvirus K1 protein: implications for lymphomagenesis. Blood 105:3987–3994. doi:10.1182/blood-2004-07-2781.
    OpenUrlAbstract/FREE Full Text
  82. 82.↵
    1. Park J,
    2. Lee D,
    3. Seo T,
    4. Chung J,
    5. Choe J
    . 2000. Kaposi’s sarcoma-associated herpesvirus (human herpesvirus-8) open reading frame 36 protein is a serine protein kinase. J Gen Virol 81:1067–1071. doi:10.1099/0022-1317-81-4-1067.
    OpenUrlCrossRefPubMedWeb of Science
  83. 83.↵
    1. Hosseinipour MC,
    2. Sweet KM,
    3. Xiong J,
    4. Namarika D,
    5. Mwafongo A,
    6. Nyirenda M,
    7. Chiwoko L,
    8. Kamwendo D,
    9. Hoffman I,
    10. Lee J,
    11. Phiri S,
    12. Vahrson W,
    13. Damania B,
    14. Dittmer DP
    . 2014. Viral profiling identifies multiple subtypes of Kaposi’s sarcoma. mBio 5:e01633-14. doi:10.1128/mBio.01633-14.
    OpenUrlAbstract/FREE Full Text
  84. 84.↵
    1. Haque M,
    2. Wang V,
    3. Davis DA,
    4. Zheng Z-M,
    5. Yarchoan R
    . 2006. Genetic organization and hypoxic activation of the Kaposi’s sarcoma-associated herpesvirus ORF34-37 gene cluster. J Virol 80:7037–7051. doi:10.1128/JVI.00553-06.
    OpenUrlAbstract/FREE Full Text
  85. 85.↵
    1. Hamza MS,
    2. Reyes RA,
    3. Izumiya Y,
    4. Wisdom R,
    5. Kung HJ,
    6. Luciw PA
    . 2004. ORF36 protein kinase of Kaposi’s sarcoma herpesvirus activates the c-Jun N-terminal kinase signaling pathway. J Biol Chem 279:38325–38330. doi:10.1074/jbc.M400964200.
    OpenUrlAbstract/FREE Full Text
  86. 86.↵
    1. Tarakanova VL,
    2. Leung-Pineda V,
    3. Hwang S,
    4. Yang CW,
    5. Matatall K,
    6. Basson M,
    7. Sun R,
    8. Piwnica-Worms H,
    9. Sleckman BP,
    10. Virgin HW, IV.
    2007. γ-Herpesvirus kinase actively initiates a DNA damage response by inducing phosphorylation of H2AX to foster viral replication. Cell Host Microbe 1:275–286. doi:10.1016/j.chom.2007.05.008.
    OpenUrlCrossRefPubMed
  87. 87.↵
    1. Li R,
    2. Zhu J,
    3. Xie Z,
    4. Liao G,
    5. Liu J,
    6. Chen MR,
    7. Hu S,
    8. Woodard C,
    9. Lin J,
    10. Taverna SD,
    11. Desai P,
    12. Ambinder RF,
    13. Hayward GS,
    14. Qian J,
    15. Zhu H,
    16. Hayward SD
    . 2011. Conserved herpesvirus kinases target the DNA damage response pathway and TIP60 histone acetyltransferase to promote virus replication. Cell Host Microbe 10:390–400. doi:10.1016/j.chom.2011.08.013.
    OpenUrlCrossRefPubMed
  88. 88.↵
    1. Gorrini C,
    2. Squatrito M,
    3. Luise C,
    4. Syed N,
    5. Perna D,
    6. Wark L,
    7. Martinato F,
    8. Sardella D,
    9. Verrecchia A,
    10. Bennett S,
    11. Confalonieri S,
    12. Cesaroni M,
    13. Marchesi F,
    14. Gasco M,
    15. Scanziani E,
    16. Capra M,
    17. Mai S,
    18. Nuciforo P,
    19. Crook T,
    20. Lough J,
    21. Amati B
    . 2007. Tip60 is a haplo-insufficient tumour suppressor required for an oncogene-induced DNA damage response. Nature 448:1063–1067. doi:10.1038/nature06055.
    OpenUrlCrossRefPubMedWeb of Science
  89. 89.↵
    1. Kuny CV,
    2. Chinchilla K,
    3. Culbertson MR,
    4. Kalejta RF
    . 2010. Cyclin-dependent kinase-like function is shared by the beta- and gamma- subset of the conserved herpesvirus protein kinases. PLoS Pathog 6:e1001092. doi:10.1371/journal.ppat.1001092.
    OpenUrlCrossRef
  90. 90.↵
    1. Bhatt AP,
    2. Wong JP,
    3. Weinberg MS,
    4. Host KM,
    5. Giffin LC,
    6. Buijnink J,
    7. van Dijk E,
    8. Izumiya Y,
    9. Kung HJ,
    10. Temple BR,
    11. Damania B
    . 2016. A viral kinase mimics S6 kinase to enhance cell proliferation. Proc Natl Acad Sci U S A 113:7876–7881. doi:10.1073/pnas.1600587113.
    OpenUrlAbstract/FREE Full Text
  91. 91.↵
    1. Anders PM,
    2. Montgomery ND,
    3. Montgomery SA,
    4. Bhatt AP,
    5. Dittmer DP,
    6. Damania B
    . 2018. Human herpesvirus-encoded kinase induces B cell lymphomas in vivo. J Clin Invest 128:2519–2534. doi:10.1172/JCI97053.
    OpenUrlCrossRef
  92. 92.↵
    1. Arvanitakis L,
    2. Geras-Raaka E,
    3. Varma A,
    4. Gershengorn MC,
    5. Cesarman E
    . 1997. Human herpesvirus KSHV encodes a constitutively active G-protein-coupled receptor linked to cell proliferation. Nature 385:347–349. doi:10.1038/385347a0.
    OpenUrlCrossRefPubMedWeb of Science
  93. 93.↵
    1. Bais C,
    2. Santomasso B,
    3. Coso O,
    4. Arvanitakis L,
    5. Raaka EG,
    6. Gutkind JS,
    7. Asch AS,
    8. Cesarman E,
    9. Gershengorn MC,
    10. Mesri EA
    . 1998. G-protein-coupled receptor of Kaposi’s sarcoma-associated herpesvirus is a viral oncogene and angiogenesis activator. Nature 391:86–89. doi:10.1038/34193.
    OpenUrlCrossRefPubMedWeb of Science
  94. 94.↵
    1. Sodhi A,
    2. Chaisuparat R,
    3. Hu J,
    4. Ramsdell AK,
    5. Manning BD,
    6. Sausville EA,
    7. Sawai ET,
    8. Molinolo A,
    9. Gutkind JS,
    10. Montaner S
    . 2006. The TSC2/mTOR pathway drives endothelial cell transformation induced by the Kaposi’s sarcoma-associated herpesvirus G protein-coupled receptor. Cancer Cell 10:133–143. doi:10.1016/j.ccr.2006.05.026.
    OpenUrlCrossRefPubMedWeb of Science
  95. 95.↵
    1. Sodhi A,
    2. Montaner S,
    3. Gutkind JS
    . 2004. Viral hijacking of G-protein-coupled-receptor signalling networks. Nat Rev Mol Cell Biol 5:998–1012. doi:10.1038/nrm1529.
    OpenUrlCrossRefPubMedWeb of Science
  96. 96.↵
    1. He M,
    2. Bakken T,
    3. Kassimova A,
    4. Boshoff C,
    5. Philpott N,
    6. Cannon ML
    . 2012. Focal adhesion kinase is required for KSHV vGPCR signaling. Mol Carcinog 51:339–351. doi:10.1002/mc.20790.
    OpenUrlCrossRef
  97. 97.↵
    1. Sodhi A,
    2. Montaner S,
    3. Patel V,
    4. Gomez-Roman JJ,
    5. Li Y,
    6. Sausville EA,
    7. Sawai ET,
    8. Gutkind JS
    . 2004. Akt plays a central role in sarcomagenesis induced by Kaposi’s sarcoma herpesvirus-encoded G protein-coupled receptor. Proc Natl Acad Sci U S A 101:4821–4826. doi:10.1073/pnas.0400835101.
    OpenUrlAbstract/FREE Full Text
  98. 98.↵
    1. Martin D,
    2. Galisteo R,
    3. Molinolo AA,
    4. Wetzker R,
    5. Hirsch E,
    6. Gutkind J
    . 2011. PI3Kγ mediates Kaposi’s sarcoma-associated herpesvirus vGPCR-induced sarcomagenesis. Cancer Cell 19:805–813. doi:10.1016/j.ccr.2011.05.005.
    OpenUrlCrossRefPubMedWeb of Science
  99. 99.↵
    1. Angelova M,
    2. Ferris MB,
    3. Swan KF,
    4. McFerrin HE,
    5. Pridjian G,
    6. Morris CA,
    7. Sullivan DE
    . 2014. Kaposi’s sarcoma-associated herpesvirus G-protein coupled receptor activates the canonical Wnt/β-catenin signaling pathway. Virol J 11:218. doi:10.1186/s12985-014-0218-8.
    OpenUrlCrossRefPubMed
  100. 100.↵
    1. Jham BC,
    2. Ma T,
    3. Hu J,
    4. Chaisuparat R,
    5. Friedman ER,
    6. Pandolfi PP,
    7. Schneider A,
    8. Sodhi A,
    9. Montaner S
    . 2011. Amplification of the angiogenic signal through the activation of the TSC/mTOR/HIF axis by the KSHV vGPCR in Kaposi’s sarcoma. PLoS One 6:e19103. doi:10.1371/journal.pone.0019103.
    OpenUrlCrossRef
  101. 101.↵
    1. Montaner S
    . 2007. Akt/TSC/mTOR activation by the KSHV G protein-coupled receptor: emerging insights into the molecular oncogenesis and treatment of Kaposi’s sarcoma. Cell Cycle 6:438–443. doi:10.4161/cc.6.4.3843.
    OpenUrlCrossRefPubMedWeb of Science
  102. 102.↵
    1. Pati S,
    2. Cavrois M,
    3. Guo HG,
    4. Foulke JS,
    5. Kim J,
    6. Feldman RA,
    7. Reitz M
    . 2001. Activation of NF-κB by the human herpesvirus 8 chemokine receptor ORF74: evidence for a paracrine model of Kaposi’s sarcoma pathogenesis. J Virol 75:8660–8673. doi:10.1128/JVI.75.18.8660-8673.2001.
    OpenUrlAbstract/FREE Full Text
  103. 103.↵
    1. Martin D,
    2. Galisteo R,
    3. Ji Y,
    4. Montaner S,
    5. Gutkind JS
    . 2008. An NF-κB gene expression signature contributes to Kaposi’s sarcoma virus vGPCR-induced direct and paracrine neoplasia. Oncogene 27:1844–1852. doi:10.1038/sj.onc.1210817.
    OpenUrlCrossRefPubMedWeb of Science
  104. 104.↵
    1. Pati S,
    2. Foulke JS,
    3. Barabitskaya O,
    4. Kim J,
    5. Nair BC,
    6. Hone D,
    7. Smart J,
    8. Feldman RA,
    9. Reitz M
    . 2003. Human herpesvirus 8-encoded vGPCR activates nuclear factor of activated T cells and collaborates with human immunodeficiency virus type 1 Tat. J Virol 77:5759–5773. doi:10.1128/JVI.77.10.5759-5773.2003.
    OpenUrlAbstract/FREE Full Text
  105. 105.↵
    1. Cannon ML,
    2. Cesarman E
    . 2004. The KSHV G protein-coupled receptor signals via multiple pathways to induce transcription factor activation in primary effusion lymphoma cells. Oncogene 23:514–523. doi:10.1038/sj.onc.1207021.
    OpenUrlCrossRefPubMedWeb of Science
  106. 106.↵
    1. Montaner S,
    2. Sodhi A,
    3. Servitja JM,
    4. Ramsdell AK,
    5. Barac A,
    6. Sawai ET,
    7. Gutkind JS
    . 2004. The small GTPase Rac1 links the Kaposi sarcoma-associated herpesvirus vGPCR to cytokine secretion and paracrine neoplasia. Blood 104:2903–2911. doi:10.1182/blood-2003-12-4436.
    OpenUrlAbstract/FREE Full Text
  107. 107.↵
    1. Montaner S,
    2. Sodhi A,
    3. Pece S,
    4. Mesri EA,
    5. Gutkind JS
    . 2001. The Kaposi’s sarcoma-associated herpesvirus G protein-coupled receptor promotes endothelial cell survival through the activation of Akt/protein kinase B. Cancer Res 61:2641–2648.
    OpenUrlAbstract/FREE Full Text
  108. 108.↵
    1. Grisotto MG,
    2. Garin A,
    3. Martin AP,
    4. Jensen KK,
    5. Chan P,
    6. Sealfon SC,
    7. Lira SA
    . 2006. The human herpesvirus 8 chemokine receptor vGPCR triggers autonomous proliferation of endothelial cells. J Clin Invest 116:1264–1273. doi:10.1172/JCI26666.
    OpenUrlCrossRefPubMedWeb of Science
  109. 109.↵
    1. Choi YB,
    2. Nicholas J
    . 2010. Induction of angiogenic chemokine CCL2 by human herpesvirus 8 chemokine receptor. Virology 397:369–378. doi:10.1016/j.virol.2009.11.024.
    OpenUrlCrossRefPubMed
  110. 110.↵
    1. Ma T,
    2. Patel H,
    3. Babapoor-Farrokhran S,
    4. Franklin R,
    5. Semenza GL,
    6. Sodhi A,
    7. Montaner S
    . 2015. KSHV induces aerobic glycolysis and angiogenesis through HIF-1-dependent upregulation of pyruvate kinase 2 in Kaposi’s sarcoma. Angiogenesis 18:477–488. doi:10.1007/s10456-015-9475-4.
    OpenUrlCrossRef
  111. 111.↵
    1. Jham BC,
    2. Montaner S
    . 2010. The Kaposi’s sarcoma-associated herpesvirus G protein-coupled receptor: lessons on dysregulated angiogenesis from a viral oncogene. J Cell Biochem 110:1–9. doi:10.1002/jcb.22524.
    OpenUrlCrossRefPubMedWeb of Science
  112. 112.↵
    1. Yang TY,
    2. Chen SC,
    3. Leach MW,
    4. Manfra D,
    5. Homey B,
    6. Wiekowski M,
    7. Sullivan L,
    8. Jenh CH,
    9. Narula SK,
    10. Chensue SW,
    11. Lira SA
    . 2000. Transgenic expression of the chemokine receptor encoded by human herpesvirus 8 induces an angioproliferative disease resembling Kaposi’s sarcoma. J Exp Med 191:445–454. doi:10.1084/jem.191.3.445.
    OpenUrlAbstract/FREE Full Text
  113. 113.↵
    1. Montaner S,
    2. Sodhi A,
    3. Molinolo A,
    4. Bugge TH,
    5. Sawai ET,
    6. He Y,
    7. Li Y,
    8. Ray PE,
    9. Gutkind JS
    . 2003. Endothelial infection with KSHV genes in vivo reveals that vGPCR initiates Kaposi’s sarcomagenesis and can promote the tumorigenic potential of viral latent genes. Cancer Cell 3:23–36. doi:10.1016/S1535-6108(02)00237-4.
    OpenUrlCrossRefPubMedWeb of Science
  114. 114.↵
    1. Guo H-G,
    2. Sadowska M,
    3. Reid W,
    4. Tschachler E,
    5. Hayward G,
    6. Reitz M
    . 2003. Kaposi’s sarcoma-like tumors in a human herpesvirus 8 ORF74 transgenic mouse. J Virol 77:2631–2639. doi:10.1128/JVI.77.4.2631-2639.2003.
    OpenUrlAbstract/FREE Full Text
  115. 115.↵
    1. Jensen KK,
    2. Manfra DJ,
    3. Grisotto MG,
    4. Martin AP,
    5. Vassileva G,
    6. Kelley K,
    7. Schwartz TW,
    8. Lira SA
    . 2005. The human herpes virus 8-encoded chemokine receptor is required for angioproliferation in a murine model of Kaposi’s sarcoma. J Immunol 174:3686–3694. doi:10.4049/jimmunol.174.6.3686.
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top
Download PDF
Citation Tools
In Vivo Models of Oncoproteins Encoded by Kaposi’s Sarcoma-Associated Herpesvirus
Ariana G. Bravo Cruz, Blossom Damania
Journal of Virology May 2019, 93 (11) e01053-18; DOI: 10.1128/JVI.01053-18

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.
In Vivo Models of Oncoproteins Encoded by 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
In Vivo Models of Oncoproteins Encoded by Kaposi’s Sarcoma-Associated Herpesvirus
Ariana G. Bravo Cruz, Blossom Damania
Journal of Virology May 2019, 93 (11) e01053-18; DOI: 10.1128/JVI.01053-18
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • INTRODUCTION
    • MOUSE MODELS OF KSHV-ENCODED LATENT ONCOPROTEINS
    • MOUSE MODELS OF KSHV-ENCODED LYTIC ONCOPROTEINS
    • CONCLUSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

KS
Kaposi’s sarcoma-associated herpesvirus
MCD
PEL
animal models
lymphoma

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