Download as pdf or txt
Download as pdf or txt
You are on page 1of 3

previews.

qxd 1/9/03 9:28 AM Page 1

P R E V I E W S

Initiation of angiogenic Kaposi’s sarcoma lesions


A new mouse model reported in this issue of Cancer Cell implies that VEGF/KDR-mediated paracrine effects induced by the
lytic cycle vGPCR signaling protein encoded by human Kaposi’s sarcoma-associated herpesvirus (KSHV or HHV8) may be
involved in promoting the proliferation of the KSHV latently infected spindle endothelial cells of Kaposi’s sarcoma.

One of the first hallmarks noted by the tics. In late stage nodular or disseminat- 1999). Most studies of clonality in KS
CDC at the beginning of what became ed KS, essentially all spindle cells are lesions have concluded that they are
the AIDS epidemic in the United States latently infected by KSHV and express either mixtures of oligoclonal cells or at
was the sudden unusual occurrence of just the viral LANA-1 protein and proba- best only partially monoclonal, suggest-
cancerous red skin lesions in a cohort of bly also the vCyc-D and vFLIP proteins ing that whereas some early develop-
young homosexual men in New York and (Dupin et al., 1999; Jenner and Boshoff, ment may involve spread of infection to
San Francisco. These angiogenic lesions 2002). LANA-1 is a chromatin-interacting new cells, the later nodular and invasive
known as Kaposi’s sarcoma (KS) resem- repressor that binds to the multicopy epi- stages probably involve cladal expansion
bled those in rare cases (defined origi- somal viral genomes giving a distinctive of proliferating populations that are now
nally in 1870) in elderly Mediterranean punctate nuclear pattern. It probably virtually totally infected and lack an apo-
Jewish men and also those in a later, plays a role in both segregation of ptotic subset.
more aggressive version in Central progeny genomes and cell growth con- The two great puzzles of KSHV
Africa (starting in the 1950s). We now trol. Both LANA-1 and vCyc-D have pro- oncogenesis are, firstly, considering that
appreciate that all forms of KS (epidem- mitotic properties, and vFLIP has KSHV clearly has the potential for caus-
ic, classic, introgenic, and endemic) are anti-apoptotic activity as well as upregu- ing neoplastic lesions and all KS lesions
associated with infections by a novel lating NFKB, a key feature of several rare have KSHV in them, why is KS so rare in
herpesvirus known as KSHV or HHV8 KSHV-associated lymphomas such as the absence of HIV? And secondly, how
discovered in KS lesion tissue in 1994 primary effusion lymphoma (PEL) and can one rationalize why virtually all of the
(Chang et al., 1994). This is normally an multicentric Castleman’s disease (MCD) unexpectedly large number of KSHV
extremely rare disease correlating with that also occur at increased frequency in genes that have been shown to have
low seropositivity rates in most parts of AIDS patients. Infectious KSHV virions some form of in vitro transforming or
the world, except sub-Saharan Africa derived from TPA-treated PEL cell lines potentially oncogenic properties fall into
(60%) and the Mediterranean (5% to can be used to infect cultured normal pri- the lytic cycle class rather than being
20%). However, the rates of KS disease mary human dermal microvascular latent state genes?
increase up to 500-fold in solid organ endothelial cells (DMVEC) to produce In this issue of Cancer Cell,
transplant patients and up to 20,000-fold LANA-1-positive latently infected cells Montaner and colleagues (2003) make
in male homosexual AIDS patients. The with a proliferating spindle cell pheno- the best case yet that, despite being a
huge increase in KS in Southern Africa, type that closely resembles KS (Ciufo et classic lytic cycle gene product, the viral-
where it is now the most frequently al., 2001). ly encoded G protein-coupled receptor
encountered cancer, represents a com- Unlike HIV, KSHV is clearly an (vGPCR or ORF74) gene is indeed the
bination of the introduction of novel HIV ancient human virus that has coevolved predominant oncogene of KSHV. Most
infection-related immunosuppression with its migrating human host since at investigators have agreed that in isola-
with a population that was already least the origin of modern humans in tion, the vGPCR protein, as a “captured”
almost ubiquitously but asymptomatical- Africa, and almost certainly throughout relative of the cellular IL8 (α-chemokine)
ly infected by KSHV. Nevertheless, it is mammalian evolution, judged by the receptor, has all of the right characteris-
very evident that HIV alone does not presence of closely related viruses in tics, including constitutive (or ligand-
cause KS, whereas KSHV is essential chimpanzees and in a number of other independent) MAPK signaling and
for KS lesions to form (Jenner and old world primate species. However, pro-angiogenic properties to explain the
Boshoff, 2002). unlike with other more typical and ubiqui- vascular appearance of KS lesions. As
There is no clear explanation of why tous human herpesviruses, KSHV infec- the laboratories of Mesri and Cesarman
HIV infection specifically depresses nor- tion (presumed to be through saliva) is have elegantly demonstrated, stable
mal cell-mediated immunological control apparently relatively inefficient and may ectopically expressed vGPCR induces
of KSHV, which is presumed to be dor- not have had time to catch up with the focus formation in NIH-3T3 cells, and
mant in latently infected B cells and per- relatively recent rapid expansion of these cells in turn produce angiogenic
haps other myeloid or endothelial cells human populations (outside of Africa) lesions resembling KS in nude mice.
(EC); however, dysregulation of immune after migration to new continents or from Serine kinase-mediated intracellular sig-
cytokine and chemokines and perhaps the Ice Age refuges in Europe and Asia naling by the vGPCR produces upregu-
the Tat protein is presumed to lead to (Zong et al., 2002). lated PI3K/AKT and MAPK/ERK
reactivation and spread of KSHV to der- Early stage KS lesions, particularly pathways leading to increased AP1
mal vascular or lymphatic endothelial those that fit into the original description activity and secreted VEGF production
cells, which then convert into the charac- of patch and plaque types, rather than and, in EC, to upregulation of the VEGF
teristic spindle cell phenotype and nodular or invasive, tend to have many receptor KDR (FLT-2) (Bais et al., 1998;
proliferate as an unusual viral-driven fewer spindle and LANA-1-positive cells Cannon et al., 2003).
neoplasia with angiogenic characteris- and include apoptotic cells (Sturzl et al., In the other well-studied γ her-

CANCER CELL : JANUARY 2003 · VOL. 3 · COPYRIGHT © 2003 CELL PRESS 1


previews.qxd 1/9/03 9:28 AM Page 2

P R E V I E W S

Figure 1. Contributions of both latent and lytic viral proteins to KS pathogenesis


A: Schematic diagram illustrating the current model supported by Montaner et al. (2003) for conversion of endothelial cells (yellow cytoplasm) to
elongated spindle cells (LANA-positive nuclei in red) by KSHV latent infection, and the subsequent proliferative neoplasia of Kaposi’s sarcoma dri-
ven by paracrine effects from sporadic lytic cycle induction of vGPCR protein expression (black cytoplasm).
B: Histological image of a human nodular KS biopsy tissue with large numbers of spindle cells (paraffin section, hematoxylin stain).
C: Double label immunohistochemistry of a human nodular KS biopsy specimen showing both latent state infected LANA-positive spindle cells
(brown nuclear spots) and an occasional cell (arrowed) expressing the lytic cycle vGPCR protein (red cytoplasm) (from Chiou et al., 2002).

pesvirus models for oncogenesis, only effects. Normally we think of the lytic gene, both the viral mRNA and its isolat-
latency genes are known to be essential cycle in herpesviruses as a process that ed upstream promotor display typical
as contributors to immortalization of B leads to rapid cell death. However, the PAA-insensitive early lytic cycle proper-
cells or T cells and for oncogenicity (e.g., pattern of lytic cycle expression in both ties in both PELs and DMVEC, and are
LMP1 and EBNA2 in Epstein-Barr virus PELs and DMVEC instead suggests a not expressed significantly in the vast
or STP in Herpesvirus saimiri). In con- hierarchy of abortive lytic progression in majority of otherwise latently infected
trast, KSHV encodes at least five lytic which some cells express only one lytic cells. This applies also to vGPCR mRNA
cycle proteins that either behave as gene (vIL6) or just the earliest set of lytic and protein expression detected by in
oncogenes in focus formation assays genes, including the immediate early situ hybridization or immunohistochem-
(K1, vIRF1, vGPCR) or at least have transcriptional regulators and one or istry in KS lesion samples. However, a
growth-promoting antiapoptotic or angio- more viral β-chemokines (vMIP). Even small fraction of cells in some KS sam-
genic properties (vIL6, vMIP). Because fewer cells also express the core DNA ples (particularly nodular cases) do
most PELs, MCD, and some KS sam- replication proteins and the late early reveal typical lytic cycle levels of vGPCR
ples, as well as KSHV-infected cultured vGPCR, and often there are very few or mRNA and protein expression (Chiou et
DMVEC spindle cells, display a low level no cells at all that express the true late al., 2002; Kirshner et al., 1999).
of spontaneous lytic cycle gene expres- gpK8.1 protein (Chiou et al., 2002; Ciufo Montaner et al. (2003) have asked
sion (usually in about 1% of the cells), et al., 2001). Therefore, some of these whether in vivo retrovirus-mediated
many authors have speculated that lytic abortive lytic stages may still be compat- infection leading to specific expression of
cycle gene expression may contribute to ible with cell survival or at least delayed vGPCR, vCYC-D, vFLIP, vIRF1, or
the KS phenotype possibly via paracrine cell death. With regard to the vGPCR vBCL2 genes can produce KS-like

2 CANCER CELL : JANUARY 2003


previews.qxd 1/9/03 9:28 AM Page 3

P R E V I E W S

lesions in mice. They found that this was gene under the control of the SV40- Selected reading
indeed the case, but only for vGPCR and enhancer again produced KS-like skin
not for the other genes in isolation. The EC lesions with confirmed vGPCR Bais, C., Santomasso, B., Coso, O., Arvanitakis,
authors chose an apparently very suc- expression in just a small fraction of the L., Raaka, E.G., Gutkind, J.S., Asch, A.S.,
Cesarman, E., Gerhengorn, M.C., and Mesri,
cessful and elegant approach for target- tumor spindle cells (M. Reitz, personal E.A. (1998). Nature 391, 86–89.
ed expression in vascular EC using a communication). Whether or not the
transgenic mouse expressing an avian occasional cells that express vGPCR Cannon, M., Philpott, N.J., and Cesarman, E.
(2003). J. Virol. 77, 57–67.
retrovirus receptor under the control of survive long-term, these results create
an EC-specific enhancer domain. an attractive scenario in which KS devel- Chang, Y., Cesarman, E., Pessin, M.S., Lee, F.,
Control infections with ALV-GFP showed opment involves initial latent infection of Culpepper, J., Knowles, D.M., and Moore, P.S.
(1994). Science 266, 1865–1869.
expression only in CD31-positive vascu- normal vascular EC, which converts
lar EC, and the use of ALV expressing them into spindle cells, followed by a pro- Chiou, C.-J., Poole, L., Ciufo, D.M., Cannon, J.S.,
wild-type vGPCR but not mutant vGPCR liferative phase requiring expression of Alcendor, D.J., Zong, J.-C., Ambinder, R.F., and
Hayward, G.S. (2002). J. Virol. 76, 3421–3439.
yielded fast growing nodular KS-like vas- the early lytic cycle vGPCR protein in at
cular lesions at high efficiency. Note that, least a few spindle cells, with subse- Ciufo, D.M., Cannon, J.S., Poole, L.J., Wang, F.,
unlike human KS lesions, these mouse quent activation of a VEGF/KDR Murray, P., Ambinder, R.F., and Hayward, G.S.
(2001). J. Virol. 75, 5614–5626.
tumors do not contain or express any autocrine loop plus apparent paracrine
KSHV latency genes, but curiously just effects on adjacent spindle cells that are Dupin, N., Fisher, C., Kellam, P., Ariad, S., Tulliez,
like human KS, PEL, and infected only latently infected (Figure 1). M., Franck, N., van Marck, E., Salmon, D., Gorin,
DMVEC, vGPCR expression occurred in This experimental model offers the I., Escande, J.-P., et al. (1999). Proc. Natl. Acad.
Sci. USA 96, 4546–4551.
just a small fraction of the spindle-like possibility of additional exciting develop-
tumor cells. A similar fraction of tumor ments in the future such as asking Gershengorn, M.C., Geras-Raaka, E., Varma, A.,
cells (presumably the same cells) also whether including LANA-1 or vMIP or and Clark-Lewis, I. (1998). J. Clin. Invest. 102,
1469–1472.
expressed VEGF and KDR. Most inter- even HIV TAT in vGPCR-driven tumors
estingly, when infected EC expressing contributes to tumor progression. Jenner, R.G., and Boshoff, C. (2002). Biochim.
either GFP or vCyc-D/vFLIP were coin- Although, in contrast to its cellular Biophys. Acta 1602, 1–22.
jected together with vGPCR-expressing counterparts such as CCR1, vGPCR evi- Kirshner, J.R., Staskus, K., Haase, A., Lagunoff,
cells, the former also contributed cooper- dently has acquired the novel ability to M., and Ganem, D. (1999). J. Virol. 73,
atively to the mass of the tumor, although signal in a ligand-independent manner, 6006–6014.
they did not proliferate in the absence of this signaling can still be up or down- Montaner, S., Sodhi, A., Molinolo, A., Bugge, T.,
vGPCR. Furthermore, tumors that regulated by certain α-chemokine lig- Sawai, E., He, Y., Li, Y., Ray, P., and Gutkind, J.S.
included cells expressing the latency ands such as GROα and IP10, (2003). Cancer Cell 3, this issue, 23–36.
genes (but not those including cells respectively (Gershengorn et al., 1998). Sturzl, M., Hohenadl, C., Zietz, C., Castanos-
expressing GFP) grew faster than those If indeed the growth of the mouse KS-like Velez, E., Wunderlich, A., Ascherl, G., Biberfeld,
with vGPCR only. Evidently, the pres- tumors can be manipulated positively or P., Monini, P., Browning, P.J., and Ensoli, B.
(1999). J. Natl. Cancer Inst. 91, 1725–1733.
ence of vGPCR-positive cells produced negatively by GROα or IP10, this could
paracrine or bystander effects that pro- provide further strong support for a key Zong, J.C., Ciufo, D.M., Viscidi, R., Alagiozoglou,
moted the propagation and survival of role of vGPCR signaling in KS L., Tyring, S., Rady, P., Orenstein, J., Boto, W.,
the cells expressing latency genes, pathogenesis. Kajumbula, H., Romano, N., et al. (2002). J. Clin.
Virol. 23, 119–148.
which must already have exhibited some
extra level of growth dysregulation over
Gary S. Hayward
the GFP-only infected cells.
The strange pattern of vGPCR Viral Oncology Program
expression in just a small percentage of Sydney Kimmel Oncology Center
the mouse tumor cells is unexplained, Bunting-Blaustein Building
1650 Orleans Street, CRB 3M09
but is remarkably similar to the situation
Johns Hopkins University School of
in human KS, and even to that in another Medicine
about to be published study in which Baltimore, Maryland 21231
transgenic mice receiving a vGPCR E-mail: ghayward@jhmi.edu

CANCER CELL : JANUARY 2003 3

You might also like