SPECIFIC AIMS

advertisement
Maciej Tarnowski
“A role of RasGRF1 in Rhabdomyosarcoma expansion and metastasis”
Bone marrow provides an environment for the survival and expansion of several
tumor types. It is a place where several pediatric sarcomas, including rhabdomyosarcoma
(RMS), metastasize from the primary tissue location to such a degree that, in rare
situations it is difficult to distinguish between them and acute lymphoid leukemia and
lymphoma. A bone marrow (BM) biopsy shows on such occasions so-called “small round
blue cells,” which are sarcoma cells infiltrating the BM (1, 2, 3)
Accumulating evidence indicates that some chemokines and growth factors play
important roles in the metastasis of cancer cells to bone marrow. Among potential
chemokine candidates, a crucial role in directing metastasis has been documented for the
SDF-1-CXCR4 receptor axis (4, 5). CXCR4 is a Gαi-protein-coupled, seven
transmembrane-spanning receptor that has important roles in the homing of
hematopoietic stem cells to the BM and their retention in the BM microenvironment. The
homing and engraftment of hematopoietic stem cells to BM is mediated by the interaction
between CXCR4 expressed on these cells and SDF-1 secreted by BM stroma (4). It was
demonstrated that CXCR4+ tumor cells (e.g., breast and prostate cancers, pediatric
sarcomas) exploit this mechanism to metastasize to SDF-1-expressing tissues, such as
BM, lungs, liver, or lymph nodes (5-8). A new receptor for SDF-1 named CXCR7 was
recently described, and the role of SDF-1 in metastasis of CXCR4+ and CXCR7+ tumors
become more complex (9-13). Furthermore, the presence of two SDF-1 binding receptors
on cancer cells complicates the rationale to employ anti-CXCR4 blocking agents only in
anti-metastatic therapy.
Rhabdomyosarcoma (RMS) is the most common sarcoma in children and there
are two major histologic subtypes: ARMS and ERMS. Clinical evidence indicates that
ARMS is more aggressive and associated with a significantly worse outcome than ERMS
(14-22) and is characterized by the t(2;13)(q35;q14) translocation in 55% of cases or the
variant t(1;13)(p36;q14) translocation in 22% of cases. However, 23% of ARMS cases
are fusion gene-negative (20). These translocations generate PAX3-FKHR or PAX71
FKHR fusion genes (17-22) that encode the fusion proteins PAX3-FKHR and PAX7FKHR that demonstrate enhanced transcriptional activity as compared to wild-type PAX3
and PAX7 and play an important role in survival and proliferation of ARMS cells (1422). In addition to PAX/FKHR fusion genes, the family of insulin factors, including
insulin (Ins), insulin-like growth factor-1 (Igf-1), and insulin-like growth factor-2 (Igf-2),
plays an important role both in proliferation and migration of RMS cells (23-26).
Aberrant expression of several other genes such as p53, p16 INK4A/p14ARF, and nonmutation-related activation of the H-Ras pathway have been postulated to function in
RMS pathogenesis (27, 28). The potential role of Ras pathway activation in RMS was
demonstrated in a zebrafish model where overexpression of H-RAS induced externally
visible RMS tumors by 10 day of life (29). Furthermore, activation of H-Ras in MSCs
that expressed dominant-negative p53 or SV40 early region and were transduced with
PAX3-FKHR lead to formation of ARMS-like cells (30). However, because Ras family
members are mutationally activated infrequently in pediatric RMS, it was postulated that
H-Ras pathway activation in the majority of RMS results from “as-yet-unidentified”
molecular mechanisms (29) and may involve other proteins like, for example, GTP
exchange factors (GEFs) .
The Ras superfamily of GTPases are regulated switches that control many
intracellular pathways. The Ras family, which includes H-, K-, and N-Ras and other
closely related isoforms, has been particularly associated with the control of cell
proliferation and migration (31-33). Ras superfamily small GTPases function through
their cycling between GTP-bound states that can couple to downstream events and
guanosine diphosphate (GDP)-bound states that do not activate those pathways (31). The
conversion between these states is governed by several groups of enzymes, including
GEFs, which catalyze the release of GDP and subsequent binding of GTP activate these
proteins, and GTPase-activating proteins (GAPs), which greatly stimulate the
endogenous GTPase activity of Ras proteins and so stimulate their inactivation.
RasGRF1 (or CDC25Mm) is a protein endowed with GEF activity that contains a
pleckstrin homology domain, which binds to phosphatidylinositol lipids within biological
membranes and assembles signaling proteins in membrane lipid rafts, coiled coil and
ilimaquinon regions, as well as catalytic Ras and Rac exchange factor domains. It has
2
been reported that RasGRF1 knockout mice are smaller than normal littermates and
display defects in memory consolidation associated with different areas of the brain, as
well as defects in  -cell development and glucose homeostasis (34). It has been
demonstrated that RasGRF1 is a c-Jun-regulated gene necessary for promoting nonadherent growth of c-Myc- or c-Jun-transduced fibroblasts. However, not much attention
has been paid to a potential role of RasGRF1 in tumorogenesis, despite this protein
having been observed to be expressed in several tumor types (35, 36).
In this proposal, we will test the hypothesis that a GEF protein, RasGRF1, plays a
crucial role in Ras hyperactivation in RMS. This hypothesis results from observation that
very small embryonic-like stem cells (VSELs) identified in adult tissues are kept
quiescent in adult murine tissues due to erasure of DNA methylation on differentially
methylated regions (DMRs) for two paternally imprinted genes, Igf2-H19 and RasGRF1,
which leads to downregulation of Igf2 and RasGRF1 expression respectively (37). In
contrast, the reverse situation is observed in human RMS (spontaneous or part of
Beckwith-Wiedemann syndrome), where imprinting of DMRs for Igf2 on chromosome
11p15.5 at the H19-Igf2 loci is re-established leading to overexpression of Igf2 (38-44).
Based on this and our preliminary data showing high expression of RasGRF1 in
RMS cells, we would like to ask if RasGRF1 could be overexpressed in RMS similarly
to the paternally imprinted Igf2 and if RasGRF1 is involved in SDF-1/I-TAC signaling
and metastasis.
To address these questions we would like to propose the following aims:
1.
By employing Real-Time RQ-PCR we will evaluate RasGRF1 expression on all
10 available to us RMS cell lines and compare its expression in RMS cells to
normal human skeletal muscles.
2.
We will downregulate RasGRF1 in selected ARMS and ERMS cells lines via a
shRNA strategy. Clones with more than 90% of RMS downregulation will be
eomployed for further studies.
3
3.
We will use in vitro assays (13, 45-48) to study the effect of RasGRF1
downregulation on RMS cell motility, directional chemotaxis, adhesion and
secretion of metalloproteinases.
4.
We will also assess whether downregulation of RasGRF1 affects serumdependent proliferation and whether addition of strong growth stimulators like
IGF2 or Insulin will provide rescue these cells from serum starvation and will
affect cell proliferation.
5.
In parallel, we will study SDF-1/I-TAC-mediated activation of MAPKp42/44,
AKT, and Janus kinase-signal transducers and activators of transcription (JAKSTAT) pathways in RMS cells with downregulated RasGRF1 as compared to
unmanipulated wild type cells. The rationale for selecting these pathways is their
involvement on cell migration/adhesion and our data showing that they are
activated in RMS cells after stimulation by SDF-1 or I-TAC (6, 13).
6.
We will also perform pull-down experiments using H-Ras-specific Abs to show
the effect of RasGRF1 on its phospohorylation/activation (Ras activation kit from
Upstate-Milipore). Based on data that RasGRF1 is also a GEF for Rac (49),
similarly as for H-Ras, we will employ a Rac1 activation assay kit (UpstateMilipore) to study effect of RasGRF1 on Rac1 activation.
7.
In parallel experiments, we will employ farnesyl transferase inhibitor (FTI) to
block H-Ras and NSC23766 inhibitor (Calbiochem) to inhibit Rac alone or in
combination. These investigations will allow us to dissect which SDF-1–
CXCR4/CXCR7-directed steps of metastasis involve RasGRF1.
8.
Finally, we will employ RMS cells with downregulated RasGRF1 and evaluate
their tumorogenic potential by employing a long-term metastasis/tumor formation
assay, xenotranplants in SCID-Beige mice. At set time intervals, we will measure
tumor size and, at time of sacrifice, we will isolate BM, liver, lungs, peripheral
blood to evaluate dissemination of RMS cells. Human DNA in these tissues will
be detected and quantified by RQ-PCR of human α-satellite DNA sequences as
described by us (46-48).
4
Based on our data, we expect to better explain a role of Ras pathway activation in
pathogenesis of RMS and to confirm our hypothesis that downregulation of RasGRF1
will impair in vitro and in vivo metastatic properties of RMS cells. The successful
perturbation of RasGRF1/Ras/Rac signaling in RMS could find immediate translational
application. We anticipate that high expression of RasGRF1 in patient samples will
correlate with a more malignant and metastatic phenotype of RMS, which may have
some prognostic value.
The impact of the proposed studies is identification of another potentially paternally
imprinted gene, RasGRF1, in RMS pathogenesis, the molecular explanation of H-Ras
activation pathway observed in RMS, and evaluation of RasGRF1 as a new potential
RMS therapeutic target and prognostic factor.
Literature Cited:
1. Sandberg AA, Stone JF, Czarnecki L, et al. Hematologic masquerade of rhabdomyosarcoma. Am J
Hematol. 2001;68:51-57.
2. Shinkoda Y, Nagatoshi Y, Fukano R, et al. Rhabdomyosarcoma masquerading as acute leukemia. Pediatr
Blood Cancer 2009;52:286-7.
3. Lisboa S, Cerveira N, Vieira J, et al. Genetic diagnosis of alveolar rhabdomyosarcoma in the bone
marrow of a patient without evidence of primary tumor. Pediatr Blood Cancer 2008;51:554-7.
4. Kucia M, Reca R, Miekus K, et al. Trafficking of Normal Stem Cells and Metastasis of Cancer Stem
Cells Involve Similar Mechanisms: Pivotal Role of the SDF-1–CXCR4 Axis. Stem Cells 2005;23:879-894.
5. Mueller A, Homey B, Soto H, et al. A., Involvement of chemokine receptors in breast cancer metastasis,
Nature. 2000;410:50–56.
6. Libura J, Drukala J, Majka M, et al. CXCR4-SDF-1 signaling is active in rhabdomyosarcoma cells and
regulates locomotion, chemotaxis, and adhesion. Blood 2002;100:2597-2606.
7. Taichman RS, Cooper C, Keller ET, et al. Use of the stromal cell-derived factor-1/CXCR4 pathway in
prostate cancer metastasis to bone. Cancer Res 2002;62:1832-7.
8. Geminder H, Sagi-Assif O, Goldberg L, et al. A possible role for CXCR4 and its ligand, the CXC
Chemokine Stromal Cell-Derived Factor-1, in the development of bone marrow metastases in
neuroblastoma. J Immunol 2001;167:4747-4757.
9. Maksym RB, Tarnowski M, Grymula K, et al. The role of stromal-derived factor-1-CXCR7 axis in
development and cancer. Eur J Pharmacol 2009;625:31-40.
10. Wang J, Shiozawa Y, Wang Y, et al. The role of CXCR7/RDC1 as a chemokine receptor for
CXCL12/SDF-1 in prostate cancer. J Biol Chem 2008;283:4283-94.
11. Burns JM, Summers BC, Wang Y, et al. A novel chemokine receptor for SDF-1 and I-TAC involved in
cell survival, cell adhesion, and tumor development. J Exp Med 2006;203:2201-13.
12. Miao Z, Luker KE, Summers BC, et al. CXCR7 (RDC1) promotes breast and lung tumor growth in
vivo and is expressed on tumor-associated vasculature. Proc Natl Acad Sci U S A 2007;104:15735-40.
13. Grymula K, Tarnowski M, Wysoczynski M, et al. Overlapping and distinct role of CXCR7-SDF1/ITAC and CXCR4-SDF-1 axes in regulating metastatic behavior of human rhabdomyosarcomas. Int J
Cancer. 2010 DOI. 10.1002/ijc.25245
14. Ruymann FB, Newton WA, Ragab AH, et al. Bone marrow metastases at diagnosis in children and
adolescents with rhabdomyosarcoma, Cancer. 1984;53:368-373.
5
15. Dickman PS, Tsokos M, Triche TJ. Biology of rhabdomyosarcoma: cell culture, xenografts and animal
models. In: Maurer HM, Ruymann FB, Pochedly C, eds. Rhabdomyosarcoma and related tumours in
children and adolescents. Florida: CRC Press 1991:49-88.
16. Barr FG, Galili N, Holick J, et al. Rearrangement of the PAX3 paired box gene in the paediatric solid
tumour alveolar rhabdomyosarcoma. Nat Genet. 1993;3:113-117.
17. Anderson MJ, Shelton GD, Cavenee KW, et al. Embryonic expression of the tumour-associated PAX3FKHR fusion protein interferes with the developmental functions of PAX3. Proc Natl Acad Sci USA
2001;98:1589-1594.
18. Davis RJ, Barr FG. Fusion genes resulting from alternative chromosomal translocations are
overexpressed by gene-specific mechanisms in alveolar rhabdomyosarcoma. Proc Natl Acad Sci USA
1997;94:8047-8051.
19. Davis RJ, D’Cruz CM, Lovell MA, et al. Fusion of PAX7 to FKHR by the variant t(1;13)(p36;q14)
translocation in alveolar rhabdomyosarcoma. Cancer Res. 1994;54:2869-2872.
20. Collins MH, Zhao H, Womer RB, et al. Proliferative and apoptotic differences between alveolar
rhabdomyosarcoma subtypes: a comparative study of tumors containing PAX3-FKHR gene fusions. Med
Pediatr Oncol. 2000;37:83-89.
21. Bennicelli JL, Advani S, Schafer BW, et al. PAX3 and PAX7 exhibit conserved cis-acting transcription
repression domains and utilize a common gain of function mechanism in alveolar rhabdomyosarcoma.
Oncogene. 1999;18:4348-4356.
22. Kelly KM, Womer RB, Barr F. PAX3-FKHR and PAX7-FKHR fusions in rhabdomyosarcoma. J
Pediatr Hematol Oncol. 1998;20:517-518.
23. Rikhof B, de Jong S, Suurmeijer AJ, Meijer C, van der Graaf WT. The insulin-like growth factor
system and sarcomas. J Pathol. 2009 Mar;217(4):469-82.
24. Makawita S, Ho M, Durbin AD, Thorner PS, Malkin D, Somers GR.
Expression of insulin-like growth factor pathway proteins in rhabdomyosarcoma: IGF-2 expression is
associated with translocation-negative tumors. Pediatr Dev Pathol. 2009 Mar-Apr;12(2):127-35.
25. Hahn H, Wojnowski L, Specht K, Kappler R, Calzada-Wack J, Potter D, Zimmer A, Müller U, Samson
E, Quintanilla-Martinez L, Zimmer A. Patched target Igf2 is indispensable for the formation of
medulloblastoma and rhabdomyosarcoma.
J Biol Chem. 2000;275(37):28341-4
26. Wang W, Kumar P, Wang W, Epstein J, Helman L, Moore JV, Kumar S.
Insulin-like growth factor II and PAX3-FKHR cooperate in the oncogenesis of rhabdomyosarcoma. Cancer
Res. 1998 Oct 1;58(19):4426-33.
27. Naini S, Etheridge KT, Adam SJ, et al. Defining the cooperative genetic changes that temporally drive
alveolar rhabdomyosarcoma. Cancer Res 2008;68:9583-8.
28. Linardic CM, Downie DL, Qualman S, Bentley RC, Counter CM.Genetic modeling of human
rhabdomyosarcoma.Cancer Res. 2005 Jun 1;65(11):4490-5.
29. Langenau DM, Keefe MD, Storer NY, et al. Effects of RAS on the genesis of embryonal
rhabdomyosarcoma. Genes Dev 2007;21:1382-95.
30. Ren YX, Finckenstein FG, Abdueva DA, et al. Mouse mesenchymal stem cells expressing PAX-FKHR
form alveolar rhabdomyosarcomas by cooperating with secondary mutations. Cancer Res 2008;68:6587-97.
31. Rossman KL, Der CJ, Sondek J. GEF means go: turning on RHO GTPases with guanine nucleotideexchange factors. Nat Rev Mol Cell Biol 2005;6:167-80.
32. Innocenti M, Zippel R, Brambilla R, et al. CDC25(Mm)/Ras-GRF1 regulates both Ras and Rac
signaling pathways. FEBS Lett 1999;460:357-62.
33. Lavagni P, Indrigo M, Colombo G, et al. Identification of novel RasGRF1 interacting partners by largescale proteomic analysis. J Mol Neurosci 2009;37:212-24.
34. Font de Mora J, Esteban LM, Burks DJ, Núñez A, Garcés C, García-Barrado MJ, Iglesias-Osma MC,
Moratinos J, Ward JM, Santos E. Ras-GRF1 signaling is required for normal beta-cell development and
glucose homeostasis. EMBO J. 2003;22(12):3039-49.
35. Abreu JR, de Launay D, Sanders ME, et al The Ras guanine nucleotide exchange factor RasGRF1
promotes matrix metalloproteinase-3 production in rheumatoid arthritis synovial tissue. Arthritis Res Ther.
2009;11(4):R121.
36. Guerrero C, Rojas JM, Chedid M, Esteban LM, Zimonjic DB, Popescu NC, Font de Mora J, Santos E.
Expression of alternative forms of Ras exchange factors GRF and SOS1 in different human tissues and cell
lines. Oncogene. 1996 Mar 7;12(5):1097-107
6
37. Shin DM, Zuba-Surma EK, Wu W, et al. Novel epigenetic mechanisms that control pluripotency and
quiescence of adult bone marrow-derived Oct-4+ very small embryonic-like stem cells. Leukemia 2009;23:
2042-2051.
38. Anderson J, Gordon A, McManus A, et al. Disruption of imprinted genes at chromosome region
11p15.5 in paediatric rhabdomyosarcoma. Neoplasia 1999;1:340-8.
39. Casola S, Pedone PV, Cavazzana AO, et al. Expression and parental imprinting of the H19 gene in
human rhabdomyosarcoma. Oncogene 1997;14:1503-10.
40. Feinberg AP. Genomic imprinting and gene activation in cancer. Nat Genet 1993;4:110-3.
41. Scrable H, Cavenee W, Ghavimi F, et al. A model for embryonal rhabdomyosarcoma tumorigenesis
that involves genome imprinting. Proc Natl Acad Sci U S A 1989;86:7480-4.
42. Zhan S, Shapiro DN, Helman LJ. Activation of an imprinted allele of the insulin-like growth factor II
gene implicated in rhabdomyosarcoma. J Clin Invest 1994;94:445-8.
43. Feinberg AP. Phenotypic plasticity and the epigenetics of human disease. Nature 2007;447:433-40.
44. Eggenschwiler J, Ludwig T, Fisher P, et al. Mouse mutant embryos overexpressing IGF-II exhibit
phenotypic features of the Beckwith-Wiedemann and Simpson-Golabi-Behmel syndromes. Genes Dev
1997;11:3128-42.
45. Tarnowski M, Grymula K, Reca R, et al. Regulation of expression of stromal-derived factor-1
receptors: CXCR4 and CXCR7 in human rhabdomyosarcomas. Mol Cancer Res 2010;8:1-14.
46. Jankowski K, Kucia M, Wysoczynski M, et al. Both hepatocyte growth factor (HGF) and stromalderived factor-1 regulate the metastatic behavior of human rhabdomyosarcoma cells, but only HGF
enhances their resistance to radiochemotherapy. Cancer Res. 2003;63:7926-35.
47. Wysoczynski M, Miekus K, Jankowski K, et al. Leukemia inhibitory factor: a newly identified
metastatic factor in rhabdomyosarcomas. Cancer Res 2007;67:2131-40.
48. Wysoczynski M, Shin DM, Kucia M, et al. Selective up-regulation of interleukin-8 by human
rhabdomyosarcomas in response to hypoxia: therapeutic implications. Int. J. Cancer 2010;126:371-381.
49. Yang H, Mattingly RR. The Ras-GRF1 exchange factor coordinates activation of H-Ras and Rac1 to
control neuronal morphology. Mol Biol Cell 2006;17:2177-2189.
7
Download