The TAZ-CAMTA1 Fusion Protein Promotes Tumorigenesis via Connective Tissue Growth Factor and Ras-MAPK Signaling in Epithelioid Hemangioendothelioma

Shuang Ma, Ryan Kanai, Ajaybabu V Pobbati, Shuo Li, Kepeng Che, Caleb N Seavey, Andrea Hallett, Ashley Burtscher, John M Lamar, Brian P Rubin, Shuang Ma, Ryan Kanai, Ajaybabu V Pobbati, Shuo Li, Kepeng Che, Caleb N Seavey, Andrea Hallett, Ashley Burtscher, John M Lamar, Brian P Rubin

Abstract

Purpose: A consistent genetic alteration in vascular cancer epithelioid hemangioendothelioma (EHE) is the t(1;3)(p36;q25) chromosomal translocation, which generates a WWTR1(TAZ)-CAMTA1 (TC) fusion gene. TC is a transcriptional coactivator that drives EHE. Here, we aimed to identify the TC transcriptional targets and signaling mechanisms that underlie EHE tumorigenesis.

Experimental design: We used NIH3T3 cells transformed with TC (NIH3T3/TC) as a model system to uncover TC-dependent oncogenic signaling. These cells proliferated in an anchorage-independent manner in suspension and soft agar. The findings of the cell-based studies were validated in a xenograft model.

Results: We identified connective tissue growth factor (CTGF) as a tumorigenic transcriptional target of TC. We show that CTGF binds to integrin αIIbβ3, which is essential for sustaining the anchorage-independent proliferation of transformed NIH3T3/TC cells. NIH3T3/TC cells also have enhanced Ras and MAPK signaling, and the activity of these pathways is reduced upon CTGF knockdown, suggesting that CTGF signaling occurs via the Ras-MAPK cascade. Further, pharmacologic inhibition of MAPK signaling through PD 0325901 and trametinib abrogated TC-driven anchorage-independent growth. Likewise, for tumor growth in vivo, NIH3T3/TC cells require CTGF and MAPK signaling. NIH3T3/TC xenograft growth was profoundly reduced upon CTGF knockdown and after trametinib treatment.

Conclusions: Collectively, our results demonstrated that CTGF and the Ras-MAPK signaling cascade are essential for TC-mediated tumorigenesis. These studies provided the preclinical rationale for SARC033 (NCI 10015-NCT03148275), a nonrandomized, open-label, phase II study of trametinib in patients with unresectable or metastatic EHE.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
CTGF is a key downstream target of the TC fusion protein. A, Immunoblots to estimate the expression levels of CTGF and CYR61 in NIH3T3 cells transfected with either an EV or FLAG-TC fusion gene construct (TC). Cells were grown in suspension on HEMA-treated culture plates. Cell proliferation assay to measure the rate of anchorage-independent cell growth of NIH3T3/TC cells grown in suspension after the knockdown of CTGF (B) or CYR61 (C). D, Soft agar colonies of NIH3T3/TC cells expressing either control (SHC1/2), CTGF, or CYR61 shRNAs; quantification of the colony numbers is also shown. E, Soft agar assay to observe colony growth after CTGF overexpression. Data represent the mean ± SD of independent wells; experiments were performed at least twice.
Figure 2.
Figure 2.
Integrin αIIbβ3 is crucial for the TC signaling pathway. A, Lysates from NIH3T3 cells transfected with FLAG-CTGF were immunoprecipitated with an anti-FLAG antibody. Coimmunoprecipitated integrins were detected in an immunoblot using anti-integrin αIIb, anti-integrin β3, or anti-integrin αV antibodies. B, Immunoblots to detect the efficiency of shRNA-mediated knockdown of integrin β3, αV, or αIIb in NIH3T3/TC cells. SHC1 is an EV, SHC2 is a scrambled control shRNA. For integrin knockdowns, two independent shRNA were used. C, Quantification of the soft agar colony counts after knockdown of the indicated integrins in NIH3T3/TC cells. Silencing of integrin αIIb and integrin β3, but not integrin αV, inhibited the ability of NIH3T3/TC cells to form colonies. Data represent mean and SD of individual wells. IP, immunoprecipitated; IB, immunoblot.
Figure 3.
Figure 3.
Ras is an important member of the TC signaling pathway. A, Expression levels of active Ras (Ras-GTP) in NIH3T3/TC and NIH3T3/EV cells that were cultured in an anchorage-independent manner. Active Ras was precipitated using the RBD of Raf-1, and the precipitated Ras levels were determined by immunoblot using an anti-Ras antibody. B, Expression levels of active Ras in NIH3T3/TC cells upon CTGF knockdown using two independent shRNA; the cells were cultured in suspension. C, Immunoblots to estimate active Ras levels and D, the activity of MAPK signaling after sorafenib (Raf inhibitor) treatment. E, Soft agar assay in the presence of vehicle (DMSO) or sorafenib. Inhibiting RAS suppressed the ability of NIH3T3/TC cells to form colonies. Comparison is to DMSO vehicle. Error bars represent one SD, and the plot is representative of two independent experiments.
Figure 4.
Figure 4.
TC predominantly activates the MAPK signaling pathway. A, NIH3T3/EV and NIH3T3/TC cells were grown attached to dishes (day 0) or in suspension for 1, 3, 7, 9, 11, or 13 days before harvesting for lysate preparation. Levels of pERK, ERK, pAKT, AKT, pS6, and S6 in cell lysates were detected by immunoblots. B, MAPK activity was estimated using a pERK antibody in MS1 cells after exogenous TC expression. C, NIH3T3 cells expressing a double FLAG-CTGF were grown in suspension for 7 days before collection. Cell lysates were analyzed by immunoblot with antibodies to pERK and ERK. Densitometry data for pERK is also shown. Expression of pERK, ERK, pAKT, and AKT in NIH3T3/TC cells after (D) CTGF or (E) CYR61 knockdown. Actin is used as a loading control. F, Immunoblots to estimate the levels of pERK and ERK between normal and matched human EHE tumor tissue samples. N, normal tissue samples; T, tumor tissue samples.
Figure 5.
Figure 5.
Effects of pharmacologic inhibitors of MAPK or PI3K pathway on anchorage-independent growth of NIH3T3/TC cells. A, NIH3T3/TC cells were cultured in the presence of either PD 0325901 or GDC 0941 for 2 days after being grown in suspension for 7 days, lysed, and the cell lysates were analyzed by immunoblotting with the indicated antibodies. GDC 0941 is a PI3K inhibitor, and PD 0325901 is a MEK inhibitor. B, The change in cell number over time of NIH3T3/TC cells grown in suspension in the presence of GDC 0941 or PD 0325901. PD 0325901 effectively inhibited the anchorage-independent proliferation of NIH3T3/TC cells (P < 0.05). C, Estimating the cell number of NIH3T3/TC cells grown in suspension. Administration of trametinib significantly decreased cell proliferation in suspension in a dose-dependent manner compared withto DMSO only (P < 0.0001). D, Soft agar colony numbers of NIH3T3/TC cells in the presence of GDC 0941 or PD 0325901 as compared with vehicle DMSO. E, Quantification of soft agar colonies with NIH3T3/TC cells in the presence of vehicle (DMSO) or trametinib. Error bars represent one SD; experiments were performed at least twice.
Figure 6.
Figure 6.
CTGF and MAPK signaling sustain tumor growth in vivo. NIH3T3/TC cells were infected with a control GFP shRNA or a shRNA targeting CTGF and assayed for A,Ctgf mRNA expression by qPCR; data are shown as mean ± SD, samples were normalized to control cells lacking any shRNA (dotted line; n = 5 samples run in triplicate from five independent experiments, **P < 0.01). B, Control or CTGF knockdown cells were injected subcutaneously in NSG mice, and tumor volume was measured twice per week for 85 days. C, Tumors from (B) were isolated and weighed; data are shown as mean ± SD (n = 6 shGFP mice and n = 5 shCTGF-1 mice, *P < 0.05 by unpaired t test). D, NIH3T3 cells stably expressing TAZ-CAMTA1 were injected subcutaneously and allowed to form tumors. Once tumors reached a volume of between 150 to 250 mm3, mice received daily oral treatment with either vehicle or 1 mg/kg trametinib for 14 days. Plots show individual mouse tumor volume normalized to tumor volume at the start of treatment (solid Iines with dots). Solid blue lines represent linear regression with 95% CI (dashed blue lines) shown (n = 6 mice per group); the difference in the slopes of the linear regression was tested by ANOVA; **** P < 0.0001.

References

    1. Tanas MR, Sboner A, Oliveira AM, Erickson-Johnson MR, Hespelt J, Hanwright PJ, et al. . Identification of a disease-defining gene fusion in epithelioid hemangioendothelioma. Sci Transl Med 2011;3:98ra82.
    1. Errani C, Zhang L, Sung YS, Hajdu M, Singer S, Maki RG, et al. . A novel WWTR1-CAMTA1 gene fusion is a consistent abnormality in epithelioid hemangioendothelioma of different anatomic sites. Genes Chromosomes Cancer 2011;50:644–53.
    1. Seavey CN, Pobbati AV, Hallett A, Ma S, Reynolds JP, Kanai R, et al. . WWTR1(TAZ)-CAMTA1 gene fusion is sufficient to dysregulate YAP/TAZ signaling and drive epithelioid hemangioendothelioma tumorigenesis. Genes Dev 2021;35:512–27.
    1. Antonescu CR, Le Loarer F, Mosquera JM, Sboner A, Zhang L, Chen CL, et al. . Novel YAP1-TFE3 fusion defines a distinct subset of epithelioid hemangioendothelioma. Genes Chromosomes Cancer 2013;52:775–84.
    1. Dermawan JK, Azzato EM, Billings SD, Fritchie KJ, Aubert S, Bahrami A, et al. . YAP1-TFE3-fused hemangioendothelioma: a multi-institutional clinicopathologic study of 24 genetically-confirmed cases. Mod Pathol 2021;34:2211–21.
    1. Zanconato F, Cordenonsi M, Piccolo S. YAP/TAZ at the roots of cancer. Cancer Cell 2016;29:783–803.
    1. Okawa ER, Gotoh T, Manne J, Igarashi J, Fujita T, Silverman KA, et al. . Expression and sequence analysis of candidates for the 1p36.31 tumor suppressor gene deleted in neuroblastomas. Oncogene 2008;27:803–10.
    1. Henrich KO, Bauer T, Schulte J, Ehemann V, Deubzer H, Gogolin S, et al. . CAMTA1, a 1p36 tumor suppressor candidate, inhibits growth and activates differentiation programs in neuroblastoma cells. Cancer Res 2011;71:3142–51.
    1. Tanas MR, Ma S, Jadaan FO, Ng CK, Weigelt B, Reis-Filho JS, et al. . Mechanism of action of a WWTR1(TAZ)-CAMTA1 fusion oncoprotein. Oncogene 2016;35:929–38.
    1. Liu CY, Zha ZY, Zhou X, Zhang H, Huang W, Zhao D, et al. . The hippo tumor pathway promotes TAZ degradation by phosphorylating a phosphodegron and recruiting the SCF{beta}-TrCP E3 ligase. J Biol Chem 2010;285:37159–69.
    1. Merritt N, Garcia K, Rajendran D, Lin ZY, Zhang X, Mitchell KA, et al. . TAZ-CAMTA1 and YAP-TFE3 alter the TAZ/YAP transcriptome by recruiting the ATAC histone acetyltransferase complex. Elife 2021;10.
    1. Lai D, Ho KC, Hao Y, Yang X. Taxol resistance in breast cancer cells is mediated by the hippo pathway component TAZ and its downstream transcriptional targets Cyr61 and CTGF. Cancer Res 2011;71:2728–38.
    1. Chu CY, Chang CC, Prakash E, Kuo ML. Connective tissue growth factor (CTGF) and cancer progression. J Biomed Sci 2008;15:675–85.
    1. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell 2002;110:673–87.
    1. Chen PS, Wang MY, Wu SN, Su JL, Hong CC, Chuang SE, et al. . CTGF enhances the motility of breast cancer cells via an integrin-alphavbeta3-ERK1/2-dependent S100A4-upregulated pathway. J Cell Sci 2007;120(Pt 12):2053–65.
    1. Tan TW, Lai CH, Huang CY, Yang WH, Chen HT, Hsu HC, et al. . CTGF enhances migration and MMP-13 up-regulation via alphavbeta3 integrin, FAK, ERK, and NF-kappaB-dependent pathway in human chondrosarcoma cells. J Cell Biochem 2009;107:345–56.
    1. Jedsadayanmata A, Chen CC, Kireeva ML, Lau LF, Lam SC. Activation-dependent adhesion of human platelets to Cyr61 and Fisp12/mouse connective tissue growth factor is mediated through integrin alpha(IIb)beta (3). J Biol Chem 1999;274:24321–7.
    1. Kinbara K, Goldfinger LE, Hansen M, Chou FL, Ginsberg MH. Ras GTPases: integrins' friends or foes? Nat Rev Mol Cell Biol 2003;4:767–76.
    1. Clark EA, Hynes RO. Ras activation is necessary for integrin-mediated activation of extracellular signal-regulated kinase 2 and cytosolic phospholipase A2 but not for cytoskeletal organization. J Biol Chem 1996;271:14814–8.
    1. Wellbrock C, Karasarides M, Marais R. The RAF proteins take centre stage. Nat Rev Mol Cell Biol 2004;5:875–85.
    1. Adnane L, Trail PA, Taylor I, Wilhelm SM. Sorafenib (BAY 43–9006, Nexavar), a dual-action inhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature. Methods Enzymol 2006;407:597–612.
    1. Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, et al. . Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov 2006;5:835–44.
    1. Simpson CD, Anyiwe K, Schimmer AD. Anoikis resistance and tumor metastasis. Cancer Lett 2008;272:177–85.
    1. Lipson KE, Wong C, Teng Y, Spong S. CTGF is a central mediator of tissue remodeling and fibrosis and its inhibition can reverse the process of fibrosis. Fibrogenesis Tissue Repair 2012;5Suppl 1:S24.
    1. Zhao B, Ye X, Yu J, Li L, Li W, Li S, et al. . TEAD mediates YAP-dependent gene induction and growth control. Genes Dev 2008;22:1962–71.
    1. Dhar A, Ray A. The CCN family proteins in carcinogenesis. Exp Oncol 2010;32:2–9.
    1. Huang YT, Lan Q, Lorusso G, Duffey N, Ruegg C. The matricellular protein CYR61 promotes breast cancer lung metastasis by facilitating tumor cell extravasation and suppressing anoikis. Oncotarget 2017;8:9200–15.
    1. Kleer CG. Dual roles of CCN proteins in breast cancer progression. J Cell Commun Signal 2016;10:217–22.
    1. Ladwa R, Pringle H, Kumar R, West K. Expression of CTGF and Cyr61 in colorectal cancer. J Clin Pathol 2011;64:58–64.
    1. Zhou ZQ, Cao WH, Xie JJ, Lin J, Shen ZY, Zhang QY, et al. . Expression and prognostic significance of THBS1, Cyr61 and CTGF in esophageal squamous cell carcinoma. BMC Cancer 2009;9:291.
    1. Xie D, Yin D, Wang HJ, Liu GT, Elashoff R, Black K, et al. . Levels of expression of CYR61 and CTGF are prognostic for tumor progression and survival of individuals with gliomas. Clin Cancer Res 2004;10:2072–81.
    1. Driskill JH, Zheng Y, Wu BK, Wang L, Cai J, Rakheja D, et al. . WWTR1(TAZ)-CAMTA1 reprograms endothelial cells to drive epithelioid hemangioendothelioma. Genes Dev 2021;35:495–511.
    1. Bennett JS. Structure and function of the platelet integrin alphaIIbbeta3. J Clin Invest 2005;115:3363–9.
    1. Lavergne M, Janus-Bell E, Schaff M, Gachet C, Mangin PH. Platelet integrins in tumor metastasis: do they represent a therapeutic target? Cancers (Basel) 2017;9:133.
    1. Trikha M, Timar J, Lundy SK, Szekeres K, Cai Y, Porter AT, et al. . The high affinity alphaIIb beta3 integrin is involved in invasion of human melanoma cells. Cancer Res 1997;57:2522–8.
    1. Shattil SJ, Kim C, Ginsberg MH. The final steps of integrin activation: the end game. Nat Rev Mol Cell Biol 2010;11:288–300.
    1. Molina JR, Adjei AA. The Ras/Raf/MAPK pathway. J Thorac Oncol 2006;1:7–9.
    1. Castellano E, Downward J. Role of RAS in the regulation of PI 3-kinase. Curr Top Microbiol Immunol 2010;346:143–69.
    1. Stacchiotti S, Simeone N, Lo Vullo S, Baldi GG, Brunello A, Vincenzi B, et al. . Activity of sirolimus in patients with progressive epithelioid hemangioendothelioma: A case-series analysis within the Italian Rare Cancer Network. Cancer. 2021;127:569–76.
    1. Chevreau C, Le Cesne A, Ray-Coquard I, Italiano A, Cioffi A, Isambert N, et al. . Sorafenib in patients with progressive epithelioid hemangioendothelioma: a phase 2 study by the French Sarcoma Group (GSF/GETO). Cancer 2013;119:2639–44.
    1. Agulnik M, Yarber JL, Okuno SH, von Mehren M, Jovanovic BD, Brockstein BE, et al. . An open-label, multicenter, phase II study of bevacizumab for the treatment of angiosarcoma and epithelioid hemangioendotheliomas. Ann Oncol 2013;24:257–63.

Source: PubMed

3
Iratkozz fel