Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 16(2)2024 Jan 21.
Article in English | MEDLINE | ID: mdl-38275898

ABSTRACT

CIC-DUX4-rearranged sarcoma (CDS) is a rare and aggressive soft tissue tumor that occurs most frequently in young adults. The key oncogenic driver of this disease is the expression of the CIC-DUX4 fusion protein as a result of chromosomal rearrangements. CIC-DUX4 displays chromatin binding properties, and is therefore believed to function as an aberrant transcription factor. However, the chromatin remodeling events induced by CIC-DUX4 are not well understood, limiting our ability to identify new mechanism-based therapeutic strategies for these patients. Here, we generated a genome-wide profile of CIC-DUX4 DNA occupancy and associated chromatin states in human CDS cell models and primary tumors. Combining chromatin profiling, proximity ligation assays, as well as genetic and pharmacological perturbations, we show that CIC-DUX4 operates as a potent transcriptional activator at its binding sites. This property is in contrast with the repressive function of the wild-type CIC protein, and is mainly mediated through the direct interaction of CIC-DUX4 with the acetyltransferase p300. In keeping with this, we show p300 to be essential for CDS tumor cell proliferation; additionally, we find its pharmacological inhibition to significantly impact tumor growth in vitro and in vivo. Taken together, our study elucidates the mechanisms underpinning CIC-DUX4-mediated transcriptional regulation.

2.
Nat Commun ; 13(1): 2267, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35477713

ABSTRACT

Oncogenic fusion proteins generated by chromosomal translocations play major roles in cancer. Among them, fusions between EWSR1 and transcription factors generate oncogenes with powerful chromatin regulatory activities, capable of establishing complex gene expression programs in permissive precursor cells. Here we define the epigenetic and 3D connectivity landscape of Clear Cell Sarcoma, an aggressive cancer driven by the EWSR1-ATF1 fusion gene. We find that EWSR1-ATF1 displays a distinct DNA binding pattern that requires the EWSR1 domain and promotes ATF1 retargeting to new distal sites, leading to chromatin activation and the establishment of a 3D network that controls oncogenic and differentiation signatures observed in primary CCS tumors. Conversely, EWSR1-ATF1 depletion results in a marked reconfiguration of 3D connectivity, including the emergence of regulatory circuits that promote neural crest-related developmental programs. Taken together, our study elucidates the epigenetic mechanisms utilized by EWSR1-ATF1 to establish regulatory networks in CCS, and points to precursor cells in the neural crest lineage as candidate cells of origin for these tumors.


Subject(s)
Sarcoma, Clear Cell , Soft Tissue Neoplasms , Carcinogenesis/genetics , Chromatin/genetics , Humans , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogenes , RNA-Binding Protein EWS/genetics , Sarcoma, Clear Cell/genetics , Sarcoma, Clear Cell/pathology , Soft Tissue Neoplasms/genetics
3.
Nat Med ; 27(2): 289-300, 2021 02.
Article in English | MEDLINE | ID: mdl-33495604

ABSTRACT

Synovial sarcoma (SyS) is an aggressive neoplasm driven by the SS18-SSX fusion, and is characterized by low T cell infiltration. Here, we studied the cancer-immune interplay in SyS using an integrative approach that combines single-cell RNA sequencing (scRNA-seq), spatial profiling and genetic and pharmacological perturbations. scRNA-seq of 16,872 cells from 12 human SyS tumors uncovered a malignant subpopulation that marks immune-deprived niches in situ and is predictive of poor clinical outcomes in two independent cohorts. Functional analyses revealed that this malignant cell state is controlled by the SS18-SSX fusion, is repressed by cytokines secreted by macrophages and T cells, and can be synergistically targeted with a combination of HDAC and CDK4/CDK6 inhibitors. This drug combination enhanced malignant-cell immunogenicity in SyS models, leading to induced T cell reactivity and T cell-mediated killing. Our study provides a blueprint for investigating heterogeneity in fusion-driven malignancies and demonstrates an interplay between immune evasion and oncogenic processes that can be co-targeted in SyS and potentially in other malignancies.


Subject(s)
Carcinogenesis/genetics , Molecular Targeted Therapy , Oncogene Proteins, Fusion/genetics , Sarcoma, Synovial/drug therapy , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Histone Deacetylases/therapeutic use , Humans , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogenes/genetics , RNA-Seq , Sarcoma, Synovial/genetics , Sarcoma, Synovial/pathology , Single-Cell Analysis
4.
Life Sci Alliance ; 4(2)2021 02.
Article in English | MEDLINE | ID: mdl-33361335

ABSTRACT

Synovial sarcoma (SyS) is an aggressive mesenchymal malignancy invariably associated with the chromosomal translocation t(X:18; p11:q11), which results in the in-frame fusion of the BAF complex gene SS18 to one of three SSX genes. Fusion of SS18 to SSX generates an aberrant transcriptional regulator, which, in permissive cells, drives tumor development by initiating major chromatin remodeling events that disrupt the balance between BAF-mediated gene activation and polycomb-dependent repression. Here, we developed SyS organoids and performed genome-wide epigenomic profiling of these models and mesenchymal precursors to define SyS-specific chromatin remodeling mechanisms and dependencies. We show that SS18-SSX induces broad BAF domains at its binding sites, which oppose polycomb repressor complex (PRC) 2 activity, while facilitating recruitment of a non-canonical (nc)PRC1 variant. Along with the uncoupling of polycomb complexes, we observed H3K27me3 eviction, H2AK119ub deposition and the establishment of de novo active regulatory elements that drive SyS identity. These alterations are completely reversible upon SS18-SSX depletion and are associated with vulnerability to USP7 loss, a core member of ncPRC1.1. Using the power of primary tumor organoids, our work helps define the mechanisms of epigenetic dysregulation on which SyS cells are dependent.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Sarcoma, Synovial/genetics , Binding Sites , Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Histones/metabolism , Humans , Multiprotein Complexes/metabolism , Organoids , Protein Binding , Protein Transport , Sarcoma, Synovial/metabolism , Transcriptome
5.
Swiss Med Wkly ; 148: w14667, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30506527

ABSTRACT

Synovial sarcoma is a highly aggressive soft tissue malignancy that often affects adolescents and young adults. It is associated with a unique chromosomal translocation that results in the formation and expression of the fusion gene SS18-SSX, which underlies its pathogenesis. Although SS18-SSX provides a potentially unique therapeutic target, all attempts to neutralise it have been unsuccessful thus far. When complete surgical removal of the tumour fails, therapy is limited to largely ineffective cytotoxic drug regimens. Nevertheless, recent discoveries about the mechanisms of SS18-SSX protein function have provided insight into potential alternative therapeutic strategies. SS18-SSX displays oncogenic activity through protein-protein interactions and participation in chromatin remodelling complexes. This review summarises our current understanding of the function of SS18-SSX and the mechanisms by which it alters the epigenetic landscape of permissive cells to induce transformation and the subsequent development of synovial sarcoma.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Sarcoma, Synovial/genetics , Translocation, Genetic/genetics , Chromatin/genetics , Humans , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Sarcoma, Synovial/pathology
6.
Sci Rep ; 6: 22113, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26905812

ABSTRACT

Expression of the SS18/SYT-SSX fusion protein is believed to underlie the pathogenesis of synovial sarcoma (SS). Recent evidence suggests that deregulation of the Wnt pathway may play an important role in SS but the mechanisms whereby SS18-SSX might affect Wnt signaling remain to be elucidated. Here, we show that SS18/SSX tightly regulates the elevated expression of the key Wnt target AXIN2 in primary SS. SS18-SSX is shown to interact with TCF/LEF, TLE and HDAC but not ß-catenin in vivo and to induce Wnt target gene expression by forming a complex containing promoter-bound TCF/LEF and HDAC but lacking ß-catenin. Our observations provide a tumor-specific mechanistic basis for Wnt target gene induction in SS that can occur in the absence of Wnt ligand stimulation.


Subject(s)
Gene Expression Profiling/methods , Oncogene Proteins, Fusion/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Animals , Axin Protein/genetics , Axin Protein/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , Co-Repressor Proteins , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mice , Microscopy, Confocal , Oncogene Proteins, Fusion/metabolism , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Synovial/genetics , Sarcoma, Synovial/metabolism , Sarcoma, Synovial/pathology , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/metabolism
7.
Genes Dev ; 24(9): 916-32, 2010 May.
Article in English | MEDLINE | ID: mdl-20382729

ABSTRACT

Cancer stem cells (CSCs) display plasticity and self-renewal properties reminiscent of normal tissue stem cells, but the events responsible for their emergence remain obscure. We recently identified CSCs in Ewing sarcoma family tumors (ESFTs) and showed that they retain mesenchymal stem cell (MSC) plasticity. In the present study, we addressed the mechanisms that underlie ESFT CSC development. We show that the EWS-FLI-1 fusion gene, associated with 85%-90% of ESFTs and believed to initiate their pathogenesis, induces expression of the embryonic stem cell (ESC) genes OCT4, SOX2, and NANOG in human pediatric MSCs (hpMSCs) but not in their adult counterparts. Moreover, under appropriate culture conditions, hpMSCs expressing EWS-FLI-1 generate a cell subpopulation displaying ESFT CSC features in vitro. We further demonstrate that induction of the ESFT CSC phenotype is the result of the combined effect of EWS-FLI-1 on its target gene expression and repression of microRNA-145 (miRNA145) promoter activity. Finally, we provide evidence that EWS-FLI-1 and miRNA-145 function in a mutually repressive feedback loop and identify their common target gene, SOX2, in addition to miRNA145 itself, as key players in ESFT cell differentiation and tumorigenicity. Our observations provide insight for the first time into the mechanisms whereby a single oncogene can reprogram primary cells to display a CSC phenotype.


Subject(s)
Cellular Reprogramming , Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , SOXB1 Transcription Factors/metabolism , Adolescent , Adult , Cell Differentiation , Cell Line, Tumor , Child , Homeodomain Proteins/metabolism , Humans , Nanog Homeobox Protein , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Phenotype , Sarcoma, Ewing/physiopathology , Tumor Cells, Cultured
8.
Cancer Res ; 69(24): 9211-8, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19934320

ABSTRACT

Overexpression of the polycomb group protein enhancer of zeste homologue 2 (EZH2) occurs in diverse malignancies, including prostate cancer, breast cancer, and glioblastoma multiforme (GBM). Based on its ability to modulate transcription of key genes implicated in cell cycle control, DNA repair, and cell differentiation, EZH2 is believed to play a crucial role in tissue-specific stem cell maintenance and tumor development. Here, we show that targeted pharmacologic disruption of EZH2 by the S-adenosylhomocysteine hydrolase inhibitor 3-deazaneplanocin A (DZNep), or its specific downregulation by short hairpin RNA (shRNA), strongly impairs GBM cancer stem cell (CSC) self-renewal in vitro and tumor-initiating capacity in vivo. Using genome-wide expression analysis of DZNep-treated GBM CSCs, we found the expression of c-myc, recently reported to be essential for GBM CSCs, to be strongly repressed upon EZH2 depletion. Specific shRNA-mediated downregulation of EZH2 in combination with chromatin immunoprecipitation experiments revealed that c-myc is a direct target of EZH2 in GBM CSCs. Taken together, our observations provide evidence that direct transcriptional regulation of c-myc by EZH2 may constitute a novel mechanism underlying GBM CSC maintenance and suggest that EZH2 may be a valuable new therapeutic target for GBM management.


Subject(s)
DNA-Binding Proteins/biosynthesis , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Transcription Factors/biosynthesis , Animals , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Down-Regulation , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, myc , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 2 , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
9.
PLoS One ; 4(11): e7904, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19936258

ABSTRACT

BACKGROUND: A characteristic SYT-SSX fusion gene resulting from the chromosomal translocation t(X;18)(p11;q11) is detectable in almost all synovial sarcomas, a malignant soft tissue tumor widely believed to originate from as yet unidentified pluripotent stem cells. The resulting fusion protein has no DNA binding motifs but possesses protein-protein interaction domains that are believed to mediate association with chromatin remodeling complexes. Despite recent advances in the identification of molecules that interact with SYT-SSX and with the corresponding wild type SYT and SSX proteins, the mechanisms whereby the SYT-SSX might contribute to neoplastic transformation remain unclear. Epigenetic deregulation has been suggested to be one possible mechanism. METHODOLOGY/PRINCIPAL FINDINGS: We addressed the effect of SYT/SSX expression on the transcriptome of four independent isolates of primary human bone marrow mesenchymal stem cells (hMSC). We observed transcriptional changes similar to the gene expression signature of synovial sarcoma, principally involving genes whose regulation is linked to epigenetic factors, including imprinted genes, genes with transcription start sites within a CpG island and chromatin related genes. Single population analysis revealed hMSC isolate-specific transcriptional changes involving genes that are important for biological functions of stem cells as well as genes that are considered to be molecular markers of synovial sarcoma including IGF2, EPHRINS, and BCL2. Methylation status analysis of sequences at the H19/IGF2 imprinted locus indicated that distinct epigenetic features characterize hMSC populations and condition the transcriptional effects of SYT-SSX expression. CONCLUSIONS/SIGNIFICANCE: Our observations suggest that epigenetic features may define the cellular microenvironment in which SYT-SSX displays its functional effects.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Adolescent , Alleles , Child , Chromatin/metabolism , CpG Islands , DNA/genetics , Gene Expression Profiling , Humans , Sarcoma, Synovial/metabolism , Transcription, Genetic , Translocation, Genetic
10.
Cancer Res ; 69(5): 1776-81, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19208848

ABSTRACT

Cancer stem cells that display tumor-initiating properties have recently been identified in several distinct types of malignancies, holding promise for more effective therapeutic strategies. However, evidence of such cells in sarcomas, which include some of the most aggressive and therapy-resistant tumors, has not been shown to date. Here, we identify and characterize cancer stem cells in Ewing's sarcoma family tumors (ESFT), a highly aggressive pediatric malignancy believed to be of mesenchymal stem cell (MSC) origin. Using magnetic bead cell separation of primary ESFT, we have isolated a subpopulation of CD133+ tumor cells that display the capacity to initiate and sustain tumor growth through serial transplantation in nonobese diabetic/severe combined immunodeficiency mice, re-establishing at each in vivo passage the parental tumor phenotype and hierarchical cell organization. Consistent with the plasticity of MSCs, in vitro differentiation assays showed that the CD133+ cell population retained the ability to differentiate along adipogenic, osteogenic, and chondrogenic lineages. Quantitative real-time PCR analysis of genes implicated in stem cell maintenance revealed that CD133+ ESFT cells express significantly higher levels of OCT4 and NANOG than their CD133- counterparts. Taken together, our observations provide the first identification of ESFT cancer stem cells and demonstration of their MSC properties, a critical step towards a better biological understanding and rational therapeutic targeting of these tumors.


Subject(s)
Bone Neoplasms/pathology , Immunomagnetic Separation/methods , Neoplastic Stem Cells/pathology , Sarcoma, Ewing/pathology , AC133 Antigen , Animals , Antigens, CD/analysis , Cell Line, Tumor , Glycoproteins/analysis , Humans , Mice , Peptides/analysis
11.
PLoS One ; 3(7): e2634, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18648544

ABSTRACT

BACKGROUND: The EWS-FLI-1 fusion protein is associated with 85-90% of Ewing's sarcoma family tumors (ESFT), the remaining 10-15% of cases expressing chimeric genes encoding EWS or FUS fused to one of several ets transcription factor family members, including ERG-1, FEV, ETV1 and ETV6. ESFT are dependent on insulin-like growth factor-1 (IGF-1) for growth and survival and recent evidence suggests that mesenchymal progenitor/stem cells constitute a candidate ESFT origin. METHODOLOGY/PRINCIPAL FINDINGS: To address the functional relatedness between ESFT-associated fusion proteins, we compared mouse progenitor cell (MPC) permissiveness for EWS-FLI-1, EWS-ERG and FUS-ERG expression and assessed the corresponding expression profile changes. Whereas all MPC isolates tested could stably express EWS-FLI-1, only some sustained stable EWS-ERG expression and none could express FUS-ERG for more than 3-5 days. Only 14% and 4% of the total number of genes that were respectively induced and repressed in MPCs by the three fusion proteins were shared. However, all three fusion proteins, but neither FLI-1 nor ERG-1 alone, activated the IGF1 promoter and induced IGF1 expression. CONCLUSION/SIGNIFICANCE: Whereas expression of different ESFT-associated fusion proteins may require distinct cellular microenvironments and induce transcriptome changes of limited similarity, IGF1 induction may provide one common mechanism for their implication in ESFT pathogenesis.


Subject(s)
Gene Expression Regulation , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/cytology , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/chemistry , RNA-Binding Protein FUS/metabolism , Transcription Factors/metabolism , Animals , Gene Expression Profiling , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS
12.
Cancer Res ; 68(7): 2176-85, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381423

ABSTRACT

Ewing's sarcoma family tumors (ESFT) express the EWS-FLI-1 fusion gene generated by the chromosomal translocation t(11;22)(q24;q12). Expression of the EWS-FLI-1 fusion protein in a permissive cellular environment is believed to play a key role in ESFT pathogenesis. However, EWS-FLI-1 induces growth arrest or apoptosis in differentiated primary cells, and the identity of permissive primary human cells that can support its expression and function has until now remained elusive. Here we show that expression of EWS-FLI-1 in human mesenchymal stem cells (hMSC) is not only stably maintained without inhibiting proliferation but also induces a gene expression profile bearing striking similarity to that of ESFT, including genes that are among the highest ESFT discriminators. Expression of EWS-FLI-1 in hMSCs may recapitulate the initial steps of Ewing's sarcoma development, allowing identification of genes that play an important role early in its pathogenesis. Among relevant candidate transcripts induced by EWS-FLI-1 in hMSCs, we found the polycomb group gene EZH2, which we show to play a critical role in Ewing's sarcoma growth. These observations are consistent with our recent findings using mouse mesenchymal progenitor cells and provide compelling evidence that hMSCs are candidate cells of origin of ESFT.


Subject(s)
Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Oncogene Proteins, Fusion/biosynthesis , Proto-Oncogene Protein c-fli-1/biosynthesis , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Differentiation/physiology , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase , Humans , Immunocompromised Host , Mice , Oncogene Proteins, Fusion/genetics , Phenotype , Polycomb Repressive Complex 2 , Proteins/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA-Binding Protein EWS , Sarcoma, Ewing/genetics , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology
13.
Cancer Res ; 66(14): 7016-23, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849546

ABSTRACT

A subset of sarcomas is associated with specific chromosomal translocations that give rise to fusion genes believed to participate in transformation and oncogenesis. Identification of the primary cell environment that provides permissiveness for the oncogenic potential of these fusion genes is essential to understand sarcoma pathogenesis. We have recently shown that expression of the EWS-FLI-1 fusion protein in primary mesenchymal progenitor cells (MPCs) suffices to develop Ewing's sarcoma-like tumors in mice. Because most sarcomas bearing unique chromosomal translocations are believed to originate from common progenitor cells, and because MPCs populate most organs, we expressed the sarcoma-associated fusion proteins FUS/TLS-CHOP, EWS-ATF1, and SYT-SSX1 in MPCs and tested the tumorigenic potential of these cells in vivo. Whereas expression of EWS-ATF1 and SYT-SSX1 failed to transform MPCs, FUS-CHOP-expressing cells formed tumors resembling human myxoid liposarcoma. Transcription profile analysis of these tumors revealed induction of transcripts known to be associated with myxoid liposarcoma and novel candidate genes, including PDGFA, whose expression was confirmed in human tumor samples. MPC(FUS-CHOP) and the previously described MPC(EWS-FLI-1) tumors displayed distinct transcription profiles, consistent with the different target gene repertoires of their respective fusion proteins. Unexpectedly, a set of genes implicated in cell survival and adhesion displayed similar behavior in the two tumors, suggesting events that may be common to primary MPC transformation. Taken together, our observations suggest that expression of FUS-CHOP may be the initiating event in myxoid liposarcoma pathogenesis, and that MPCs may constitute one cell type from which these tumors originate.


Subject(s)
Cell Transformation, Neoplastic/genetics , Liposarcoma, Myxoid/metabolism , Liposarcoma, Myxoid/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Oncogene Proteins, Fusion/biosynthesis , RNA-Binding Protein FUS/biosynthesis , Transcription Factor CHOP/biosynthesis , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Liposarcoma, Myxoid/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, SCID , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein FUS/genetics , Transcription Factor CHOP/genetics , Transfection
14.
Cancer Res ; 65(24): 11459-68, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16357154

ABSTRACT

Ewing's sarcoma is a member of Ewing's family tumors (EFTs) and the second most common solid bone and soft tissue malignancy of children and young adults. It is associated in 85% of cases with the t(11;22)(q24:q12) chromosomal translocation that generates fusion of the 5' segment of the EWS gene with the 3' segment of the ETS family gene FLI-1. The EWS-FLI-1 fusion protein behaves as an aberrant transcriptional activator and is believed to contribute to EFT development. However, EWS-FLI-1 induces growth arrest and apoptosis in normal fibroblasts, and primary cells that are permissive for its putative oncogenic properties have not been discovered, hampering basic understanding of EFT biology. Here, we show that EWS-FLI-1 alone can transform primary bone marrow-derived mesenchymal progenitor cells and generate tumors that display hallmarks of Ewing's sarcoma, including a small round cell phenotype, expression of EFT-associated markers, insulin like growth factor-I dependence, and induction or repression of numerous EWS-FLI-1 target genes. These observations provide the first identification of candidate primary cells from which EFTs originate and suggest that EWS-FLI-1 expression may constitute the initiating event in EFT pathogenesis.


Subject(s)
Bone Marrow Cells/pathology , Cell Transformation, Neoplastic , Mesenchymal Stem Cells/pathology , Sarcoma, Ewing/pathology , Animals , Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/metabolism , Phenotype , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS , Sarcoma, Ewing/etiology , Sarcoma, Ewing/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...