Elsevier

Gene

Volume 363, 19 December 2005, Pages 1-14
Gene

Review
EWS–ETS oncoproteins: The linchpins of Ewing tumors

https://doi.org/10.1016/j.gene.2005.08.007Get rights and content

Abstract

Ewing tumors, which comprise Ewing's sarcoma and peripheral primitive neuroectodermal tumors, are highly aggressive and mostly affect children and adolescents. Their molecular signature is a chromosomal translocation leading to the generation of EWS–ETS (or very rarely FUS–ETS) fusion proteins that are capable of transforming cells. These oncoproteins act as aberrant transcription factors due to the fusion of an ETS DNA binding domain to a highly potent EWS (or FUS) transactivation domain. Accordingly, many EWS–ETS target genes have been identified whose dysregulation could contribute to the development of tumor formation. Furthermore, EWS–ETS oncoproteins may impact on RNA splicing or affect other proteins through disturbing their ability to form functional complexes. The molecular knowledge gained so far from studying EWS–ETS oncoproteins has not only broadened our understanding of Ewing tumors but also improved the diagnosis of these highly undifferentiated tumors. In addition, several potential prognostic markers have been uncovered and novel therapies are suggested that may improve the still dismal survival rate of Ewing tumor patients.

Introduction

Ewing's sarcoma is an aggressive and frequently metastatic small round blue-cell tumor that derives its name from the description of an endothelioma of bone by James Ewing in 1921 (Ewing, 1921). Although the vast majority of Ewing's sarcomas arises in bone, especially in the pelvis, femur, fibula and tibia, in ∼ 15% of Ewing's sarcoma patients the primary tumor site is within soft tissue. Approximately half of all patients are between 10 and 20 years of age at the time of first diagnosis, making this highly undifferentiated tumor the second most common primary malignant bone tumor in children and adolescents (Grier, 1997, Denny, 1998, Kovar, 1998, de Alava and Gerald, 2000, Paulussen et al., 2001). The frequency of occurrence in children is in the range of 1–3 per million per year in the Western hemisphere; interestingly, Ewing's sarcoma is slightly more frequent in males than females as well as more frequent in people of European than Chinese or African origin (Fraumeni and Glass, 1970, Young and Miller, 1975, Li et al., 1980, Stiller and Parkin, 1996, Zucman-Rossi et al., 1997, Hense et al., 1999, Cotterill et al., 2000).

The diagnosis of Ewing's sarcoma has been difficult due to the absence of distinguishing morphological features. Although most Ewing's sarcomas express high levels of the transmembrane glycoprotein, MIC2/CD99 (Kovar et al., 1990, Perlman et al., 1994), this marker is also highly expressed in several other tumors, including small round blue-cell tumors such as rhabdomyosarcomas. Thus, MIC2/CD99 staining lacks specificity and therefore Ewing's sarcoma was often diagnosed in the past by excluding other tumors. This situation changed in the early 1990s with the discovery that Ewing's sarcomas are characterized by a chromosomal translocation involving the EWSR1 (Ewing's sarcoma breakpoint region 1) gene and an ETS (E26 transformation-specific) gene (Delattre et al., 1992, Arvand and Denny, 2001, Ladanyi, 2002). As a result of this translocation, fusion proteins composed of the N-terminus of the EWS (Ewing's sarcoma) protein and C-terminal portions of an ETS protein are expressed that are thought to be causative for the development of Ewing's sarcoma.

This molecular signature, the expression of an EWS–ETS fusion protein, was not only observed in classical Ewing's sarcoma, but also in other small round blue-cell tumors, including peripheral primitive neuroectodermal, Askin's and paravertebral small cell tumors. Thus, these tumors are now combined into the family of Ewing tumors (Kovar, 1998, de Alava and Gerald, 2000) and their diagnosis is helped by fluorescence in-situ hybridization and RT-PCR directed at detecting the underlying chromosomal translocations and resulting EWS–ETS fusion transcripts (Peter et al., 2001, Qian et al., 2005). Despite this recent progress, much remains to be learned about Ewing tumors. For instance, their cell of origin is still undefined, although many lines of evidence, including the expression of typical neuronal enzymes, the ability of some Ewing tumor cell lines to differentiate upon stimulation into primitive dendrites and a similar gene expression profile as neural crest-derived cells, support a neuroectodermal origin (Cavazzana et al., 1987, O'Regan et al., 1995, Staege et al., 2004). Furthermore, we do not know what other events apart from the chromosomal translocation generating an EWS–ETS fusion have to take place in order for a Ewing tumor to develop. And most significantly, the molecular knowledge gained so far has not yet translated into better therapies in the fight against Ewing tumors.

Section snippets

EWS–ETS oncoproteins

The realization that chromosomal translocations can be the cause of neoplasias may have provided a rationale for characterizing chromosomal abnormalities in Ewing tumors. Indeed, a recurrent t(11;22)(q24;q12) translocation has been noted in Ewing tumors for more than two decades (Aurias et al., 1983, Turc-Carel et al., 1983, Becroft et al., 1984, de Chadarevian et al., 1984, Whang-Peng et al., 1984, Whang-Peng et al., 1986, Whang-Peng et al., 1987, Vigfusson et al., 1986). In 1992, for the

EWS–ETS target genes

EWS–ETS fusion proteins are transcription factors in which a highly potent transactivation domain is fused to an ETS DNA binding domain. As such, it is expected that EWS–ETS fusion proteins upregulate many target genes in Ewing tumors and thereby lead to the development of this disease. However, it is also possible that EWS–ETS fusion proteins repress certain target genes, similar to other transcription factors that promoter–specifically up–or downregulate gene transcription; also, one has to

EWS–ETS proteins: beyond transcription regulation

Although most studies have focused on their role as aberrant transcription factors, EWS–ETS fusion proteins may have functions that are unrelated to transcription. In particular, EWS–ETS proteins may affect RNA splicing. Indeed, EWS–FLI1 has been shown to interact with the splicing factor U1C and is capable of altering splice site selection and modulating splicing efficiency (Knoop and Baker, 2000, Knoop and Baker, 2001, Yang et al., 2000). Of note, EWS–FLI1 can also interact with at least two

Functions of EWS, FUS and ETS proteins

Due to the nature of the chromosomal translocations in Ewing tumors, one of the EWS (or FUS) and one of the FLI1 (or ERG, FEV, ETV1, or ETV4) alleles will be inactivated, leading to hemizygosity. Thus, it is conceivable that dosage reduction of one of these genes contributes to the development of Ewing tumors. Furthermore, it has been shown that EWS–FLI1 can form complexes with EWS and FLI1 (Spahn et al., 2003), thereby potentially modulating their function. Finally, proteins that interact with

Therapeutic strategies

Ewing tumors are nowadays aggressively treated with surgery, radiation and chemotherapy (Meyers and Levy, 2000, Weber, 2002, Rodriguez-Galindo et al., 2003). Despite the introduction of enhanced chemotherapeutic regimens during the last decades that drastically improved therapy, the 5-year survival rate of Ewing tumor patients is still dismal at ∼ 60% and less than half of that for patients that present with metastases at diagnosis. Furthermore, long-term survival rates are probably

Conclusion

The discovery of EWS–ETS fusion proteins more than a decade ago has been instrumental for the vast expansion of our knowledge about Ewing tumors. In particular, diagnosis has benefited through the ability to detect EWS–ETS fusion proteins by molecular means, and prognosis will be helped in the future by profiling Ewing tumors through gene chip microarrays and possibly also proteomics. But therapy still awaits improvement, which may come through more insight into the molecular mechanisms of

Acknowledgement

This work was supported by Grant CA085257 from the National Cancer Institute.

References (232)

  • A. Goel et al.

    Concerted activation of ETS protein ER81 by p160 coactivators, the acetyltransferase p300 and the receptor tyrosine kinase HER2/Neu

    J. Biol. Chem.

    (2004)
  • H.E. Grier

    The Ewing family of tumors. Ewing's sarcoma and primitive neuroectodermal tumors

    Pediatr. Clin. North Am.

    (1997)
  • M. Hallier et al.

    The transcription factor Spi-1/PU.1 interacts with the potential splicing factor TLS

    J. Biol. Chem.

    (1998)
  • D. Hanahan et al.

    The hallmarks of cancer

    Cell

    (2000)
  • A. Hart

    Fli-1 is required for murine vascular and megakaryocytic development and is hemizygously deleted in patients with thrombocytopenia

    Immunity

    (2000)
  • E. Hiyama et al.

    Telomerase as tumor marker

    Cancer Lett.

    (2003)
  • I. Irminger-Finger et al.

    BRCA1-dependent and independent functions of BARD1

    Int. J. Biochem. Cell Biol.

    (2002)
  • R. Janknecht

    On the road to immortality: hTERT upregulation in cancer cells

    FEBS Lett.

    (2004)
  • R. Janknecht et al.

    Gene regulation by Ets proteins

    Biochim. Biophys. Acta

    (1993)
  • L.L. Knoop et al.

    The splicing factor U1C represses EWS/FLI-mediated transactivation

    J. Biol. Chem.

    (2000)
  • L.L. Knoop et al.

    EWS/FLI alters 5′-splice site selection

    J. Biol. Chem.

    (2001)
  • X.T. Kong

    Consistent detection of TLS/FUS–ERG chimeric transcripts in acute myeloid leukemia with t(16;21)(p11;q22) and identification of a novel transcript

    Blood

    (1997)
  • S.I. Ahmed et al.

    Vascular endothelial growth factor (VEGF) inhibition by small molecules

    J. Chemother.

    (2004)
  • A. Arvand et al.

    Biology of EWS/ETS fusions in Ewing's family tumors

    Oncogene

    (2001)
  • A. Arvand et al.

    The COOH-terminal domain of FLI-1 is necessary for full tumorigenesis and transcriptional modulation by EWS/FLI-1

    Cancer Res.

    (2001)
  • C. Attwooll et al.

    Identification of a novel fusion gene involving hTAFII68 and CHN from a t(9;17)(q22;q11.2) translocation in an extraskeletal myxoid chondrosarcoma

    Oncogene

    (1999)
  • A. Aurias et al.

    Chromosomal translocations in Ewing's sarcoma

    N. Engl. J. Med.

    (1983)
  • C. Baer

    Profiling and functional annotation of mRNA gene expression in pediatric rhabdomyosarcoma and Ewing's sarcoma

    Int. J. Cancer

    (2004)
  • R.A. Bailly

    DNA-binding and transcriptional activation properties of the EWS–FLI-1 fusion protein resulting from the t(11;22) translocation in Ewing sarcoma

    Mol. Cell. Biol.

    (1994)
  • Y. Ben-David et al.

    Erythroleukemia induction by Friend murine leukemia virus: insertional activation of a new member of the ets gene family, Fli-1, closely linked to c-ets-1

    Genes Dev.

    (1991)
  • A. Bertolotti et al.

    hTAF(II)68, a novel RNA/ssDNA-binding protein with homology to the pro-oncoproteins TLS/FUS and EWS is associated with both TFIID and RNA polymerase II

    EMBO J.

    (1996)
  • A. Bertolotti et al.

    EWS, but not EWS–FLI-1, is associated with both TFIID and RNA polymerase II: interactions between two members of the TET family, EWS and hTAFII68, and subunits of TFIID and RNA polymerase II complexes

    Mol. Cell. Biol.

    (1998)
  • A. Bertolotti et al.

    The N-terminal domain of human TAFII68 displays transactivation and oncogenic properties

    Oncogene

    (1999)
  • A.R. Black et al.

    Sp1 and kruppel-like factor family of transcription factors in cell growth regulation and cancer

    J. Cell. Physiol.

    (2001)
  • D.G. Bosc et al.

    Regulation of Her2/neu promoter activity by the ETS transcription factor, ER81

    J. Cell. Biochem.

    (2002)
  • D.G. Bosc et al.

    HER2/Neu-mediated activation of the ETS transcription factor ER81 and its target gene MMP-1

    Oncogene

    (2001)
  • B.S. Braun et al.

    Identification of target genes for the Ewing's sarcoma EWS/FLI fusion protein by representational difference analysis

    Mol. Cell. Biol.

    (1995)
  • C.E. Brinckerhoff et al.

    Matrix metalloproteinases: a tail of a frog that became a prince

    Nat. Rev., Mol. Cell Biol.

    (2002)
  • R.I. Brody

    Molecular analysis of the fusion of EWS to an orphan nuclear receptor gene in extraskeletal myxoid chondrosarcoma

    Am. J. Pathol.

    (1997)
  • S.A. Burchill

    Ewing's sarcoma: diagnostic, prognostic, and therapeutic implications of molecular abnormalities

    J. Clin. Pathol.

    (2003)
  • P. Carmeliet et al.

    Angiogenesis in cancer and other diseases

    Nature

    (2000)
  • A.O. Cavazzana et al.

    Experimental evidence for a neural origin of Ewing's sarcoma of bone

    Am. J. Pathol.

    (1987)
  • J. Clark

    Fusion of the EWS gene to CHN, a member of the steroid/thyroid receptor gene superfamily, in a human myxoid chondrosarcoma

    Oncogene

    (1996)
  • S.J. Cotterill et al.

    Incidence and survival for cancer in children and young adults in the North of England, 1968–1995: a report from the Northern region young persons' malignant disease registry

    Br. J. Cancer

    (2000)
  • L.M. Coussens et al.

    Matrix metalloproteinase inhibitors and cancer: trials and tribulations

    Science

    (2002)
  • A. Crozat et al.

    Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma

    Nature

    (1993)
  • S. Dalal

    Vascular endothelial growth factor: a therapeutic target for tumors of the Ewing's sarcoma family

    Clin. Cancer Res.

    (2005)
  • L. Dauphinot

    Analysis of the expression of cell cycle regulators in Ewing cell lines: EWS–FLI-1 modulates p57KIP2 and c-Myc expression

    Oncogene

    (2001)
  • E. de Alava et al.

    Molecular biology of the Ewing's sarcoma/primitive neuroectodermal tumor family

    J. Clin. Oncol.

    (2000)
  • E. de Alava

    EWS–FLI1 fusion transcript structure is an independent determinant of prognosis in Ewing's sarcoma

    J. Clin. Oncol.

    (1998)
  • Cited by (0)

    View full text