Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Oncogene ; 36(43): 5995-6005, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28671673

ABSTRACT

Ewing sarcoma (EWS) is a paediatric bone cancer with high metastatic potential. Cellular plasticity resulting from dynamic cytoskeletal reorganization, typically regulated via the Rho pathway, is a prerequisite for metastasis initiation. Here, we interrogated the role of the Ewing sarcoma driver oncogene EWS-FLI1 in cytoskeletal reprogramming. We report that EWS-FLI1 strongly represses the activity of the Rho-F-actin signal pathway transcriptional effector MRTFB, affecting the expression of a large number of EWS-FLI1-anticorrelated genes including structural and regulatory cytoskeletal genes. Consistent with this finding, chromatin immunoprecipitation sequencing (ChIP-seq) revealed strong overlaps in myocardin-related transcription factor B (MRTFB) and EWS-FLI1 chromatin occupation, especially for EWS-FLI1-anticorrelated genes. Binding of the transcriptional co-activator Yes-associated protein (YAP)-1, enrichment of TEAD-binding motifs in these shared genomic binding regions and overlapping transcriptional footprints of MRTFB and TEAD factors led us to propose synergy between MRTFB and the YAP/TEAD complex in the regulation of EWS-FLI1-anticorrelated genes. We propose that EWS-FLI1 suppresses the Rho-actin pathway by perturbation of a MRTFB/YAP-1/TEAD transcriptional module, which directly affects the actin-autoregulatory feedback loop. As spontaneous fluctuations in EWS-FLI1 levels of Ewing sarcoma cells in vitro and in vivo, associated with a switch between a proliferative, non-migratory EWS-FLI1-high and a non-proliferative highly migratory EWS-FLI1-low state, were recently described, our data provide a mechanistic basis for the underlying EWS-FLI1-dependent reversible cytoskeletal reprogramming of Ewing sarcoma cells.


Subject(s)
Cellular Reprogramming/genetics , Cytoskeleton/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Actins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin/genetics , Gene Expression Regulation, Neoplastic , Humans , Sarcoma, Ewing/pathology , Signal Transduction/genetics
2.
Oncogene ; 35(30): 3944-54, 2016 07 28.
Article in English | MEDLINE | ID: mdl-26616853

ABSTRACT

Sarcomas are mesenchymal tumors characterized by blocked differentiation process. In Ewing sarcoma (EWS) both CD99 and EWS-FLI1 concur to oncogenesis and inhibition of differentiation. Here, we demonstrate that uncoupling CD99 from EWS-FLI1 by silencing the former, nuclear factor-κB (NF-κB) signaling is inhibited and the neural differentiation program is re-established. NF-κB inhibition passes through miR-34a-mediated repression of Notch pathway. CD99 counteracts EWS-FLI1 in controlling NF-κB signaling through the miR-34a, which is increased and secreted into exosomes released by CD99-silenced EWS cells. Delivery of exosomes from CD99-silenced cells was sufficient to induce neural differentiation in recipient EWS cells through miR-34a inhibition of Notch-NF-κB signaling. Notably, even the partial delivery of CD99 small interfering RNA may have a broad effect on the entire tumor cell population owing to the spread operated by their miR-34a-enriched exosomes, a feature opening to a new therapeutic option.


Subject(s)
12E7 Antigen/physiology , MicroRNAs/physiology , NF-kappa B/physiology , Receptors, Notch/physiology , Sarcoma, Ewing/pathology , Signal Transduction/physiology , Cell Differentiation , Humans , Oncogene Proteins, Fusion/physiology , Proto-Oncogene Protein c-fli-1/physiology , RNA, Small Interfering/genetics , RNA-Binding Protein EWS/physiology
3.
Oncogene ; 33(30): 3927-38, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-23995784

ABSTRACT

The Ewing sarcoma (ES) EWS-FLI1 chimeric oncoprotein is a prototypic aberrant ETS transcription factor with activating and repressive regulatory functions. We report that EWS-FLI1-repressed promoters are enriched in forkhead box (FOX) recognition motifs, and identify FOXO1 as a EWS-FLI1-suppressed regulator orchestrating a major subset of EWS-FLI1-repressed genes. In addition to FOXO1 regulation by direct promoter binding of EWS-FLI1, its subcellular localization and activity is regulated by cyclin-dependent kinase 2- and AKT-mediated phosphorylation downstream of EWS-FLI1. Restoration of nuclear FOXO1 expression in ES cells impaired proliferation and significantly reduced clonogenicity. Gene-expression profiling revealed a significant overlap between EWS-FLI1-repressed and FOXO1-activated genes. As a proof of principle for a potential therapeutic application of our findings, the treatment of ES cell lines with methylseleninic acid (MSA) reactivated endogenous FOXO1 in the presence of EWS-FLI1 in a dose- and time-dependent manner and induced massive cell death dependent on FOXO1. In an orthotopic xenograft mouse model, MSA increased FOXO1 expression in the tumor paralleled by a significant decrease in ES tumor growth. FOXO1 reactivation by small molecules may therefore serve as a promising strategy for a future ES-specific therapy.


Subject(s)
Bone Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/metabolism , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Binding Sites , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Consensus Sequence , Cyclin-Dependent Kinase 2/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Silencing , Humans , Mice , Oncogene Proteins, Fusion/genetics , Organoselenium Compounds/pharmacology , Phosphorylation , Promoter Regions, Genetic , Protein Processing, Post-Translational , Protein Transport , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Transcription, Genetic , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Br J Cancer ; 109(10): 2696-704, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24129240

ABSTRACT

BACKGROUND: Though p53 mutations are rare in ES, there is a strong indication that p53 mutant tumours form a particularly bad prognostic group. As such, novel treatment strategies are warranted that would specifically target and eradicate tumour cells containing mutant p53 in this subset of ES patients. METHODS: PRIMA-1(Met), also known as APR-246, is a small organic molecule that has been shown to restore tumour-suppressor function primarily to mutant p53 and also to induce cell death in various cancer types. In this study, we interrogated the ability of APR-246 to induce apoptosis and inhibit tumour growth in ES cells with different p53 mutations. RESULTS: APR-246 variably induced apoptosis, associated with Noxa, Puma or p21(WAF1) upregulation, in both mutant and wild-type p53 harbouring cells. The apoptosis-inducing capability of APR-246 was markedly reduced in ES cell lines transfected with p53 siRNA. Three ES cell lines established from the same patient at different stages of the disease and two cell lines of different patients with identical p53 mutations all exhibited different sensitivities to APR-246, indicating cellular context dependency. Comparative transcriptome analysis on the three cell lines established from the same patient identified differential expression levels of several TP53 and apoptosis-associated genes such as APOL6, PENK, PCDH7 and MST4 in the APR-246-sensitive cell line relative to the less APR-246-sensitive cell lines. CONCLUSION: This is the first study reporting the biological response of Ewing sarcoma cells to APR-246 exposure and shows gross variability in responses. Our study also proposes candidate genes whose expression might be associated with ES cells' sensitivity to APR-246. With APR-246 currently in early-phase clinical trials, our findings call for caution in considering it as a potential adjuvant to conventional ES-specific chemotherapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Quinuclidines/pharmacology , Sarcoma, Ewing/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Apoptosis/genetics , Bone Neoplasms/pathology , Gene Knockdown Techniques , Humans , Microarray Analysis , Mutation/physiology , RNA, Small Interfering/pharmacology , Sarcoma, Ewing/pathology , Transcriptome , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors
5.
Oncogene ; 30(18): 2173-80, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21217773

ABSTRACT

EWS-FLI1 is a chromosome translocation-derived chimeric transcription factor that has a central and rate-limiting role in the pathogenesis of Ewing's sarcoma. Although the EWS-FLI1 transcriptomic signature has been extensively characterized on the mRNA level, information on its impact on non-coding RNA expression is lacking. We have performed a genome-wide analysis of microRNAs affected by RNAi-mediated silencing of EWS-FLI1 in Ewing's sarcoma cell lines, and differentially expressed between primary Ewing's sarcoma and mesenchymal progenitor cells. Here, we report on the identification of hsa-mir-145 as the top EWS-FLI1-repressed microRNA. Upon knockdown of EWS-FLI1, hsa-mir-145 expression dramatically increases in all Ewing's sarcoma cell lines tested. Vice versa, ectopic expression of the microRNA in Ewing's sarcoma cell lines strongly reduced EWS-FLI1 protein, whereas transfection of an anti-mir to hsa-mir-145 increased the EWS-FLI1 levels. Reporter gene assays revealed that this modulation of EWS-FLI1 protein was mediated by the microRNA targeting the FLI1 3'-untranslated region. Mutual regulations of EWS-FLI1 and hsa-mir-145 were mirrored by an inverse correlation between their expression levels in four of the Ewing's sarcoma cell lines tested. Consistent with the role of EWS-FLI1 in Ewing's sarcoma growth regulation, forced hsa-mir-145 expression halted Ewing's sarcoma cell line growth. These results identify feedback regulation between EWS-FLI1 and hsa-mir-145 as an important component of the EWS-FLI1-mediated Ewing's sarcomagenesis that may open a new avenue to future microRNA-mediated therapy of this devastating malignant disease.


Subject(s)
MicroRNAs/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Base Sequence , DNA Primers , Humans
6.
Oncogene ; 28(9): 1280-4, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19151750

ABSTRACT

The oncogene EWS-FLI1 encodes a chimeric transcription factor expressed in Ewing's sarcoma family tumors (ESFTs). EWS-FLI1 target gene expression is thought to drive ESFT pathogenesis and, therefore, inhibition of EWS-FLI1 activity holds high therapeutic promise. As the activity of many transcription factors is regulated by post-translational modifications, we studied the presence of modifications on EWS-FLI1. The immuno-purified fusion-protein was recognized by an antibody specific for O-linked beta-N-acetylglucosaminylation, and bound readily to a phosphoprotein-specific dye. Inhibition of Ser/Thr-specific phophatases increased EWS-FLI1 molecular weight and reduced its O-GlcNAc content, suggesting that phosphorylation and O-GlcNAcylation of EWS-FLI1 interact dynamically. By mutation analysis, O-GlcNAcylation was delineated to Ser/Thr residues of the amino-terminal EWS transcriptional-activation domain. Metabolic inhibition of the hexosamine biosynthetic pathway abrogated O-GlcNAcylation of EWS-FLI1 and interfered specifically with transcriptional activation of the EWS-FLI1 target Id2. These results suggest that drugs modulating glycosylation of EWS-FLI1 interfere functionally with its activity and might, therefore, constitute promising additions to the current ESFT chemotherapy.


Subject(s)
Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , Sarcoma, Ewing/genetics , Transcription, Genetic , Acylation , Humans , RNA-Binding Protein EWS
7.
Oncogene ; 25(19): 2795-800, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16314831

ABSTRACT

High CD99 expression levels and rearrangements of the EWS gene with ETS transcription factor genes characterize the Ewing's sarcoma family of tumors (ESFT). CD99 is a cell surface glycoprotein whose engagement has been implicated in cell proliferation as well as upregulation and transport of several transmembrane proteins in hematopoietic cells. In ESFT, antibody ligation of CD99 induces fast homotypic cell aggregation and cell death although its functional role in these processes remains largely unknown. Here, using an RNAi approach, we studied for the first time the consequences of modulated CD99 expression in six different ESFT cell lines, representing the most frequent variant forms of EWS gene rearrangement. CD99 suppression resulted in growth inhibition and reduced migration of ESFT cells. Among genes whose expression changes in response to CD99 modulation, the potassium-channel modulatory factor KCMF1 was consistently upregulated. In a series of 22 primary ESFT, KCMF1 expression levels inversely correlated with CD99 abundancy. Cells forced to express ectopic KCMF1 showed a similar reduction in migratory ability as CD99 silenced ESFT cells. Our results suggest that in ESFT, high CD99 expression levels contribute to the malignant properties of ESFT by promoting growth and migration of tumor cells and identify KCMF1 as a potential metastasis suppressor gene downregulated by high constitutive CD99 expression in ESFT.


Subject(s)
Antigens, CD/physiology , Bone Neoplasms/pathology , Cell Adhesion Molecules/physiology , Sarcoma, Ewing/pathology , Ubiquitin-Protein Ligases/metabolism , 12E7 Antigen , Bone Neoplasms/metabolism , Cell Movement , Cell Proliferation , Colony-Forming Units Assay , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/pharmacology , Sarcoma, Ewing/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/genetics , Up-Regulation
8.
Cancer Res ; 61(16): 5992-7, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507040

ABSTRACT

EWS encodes a ubiquitously expressed RNA binding protein with largely unknown function. In Ewing sarcoma family tumors (EFT), one allele is rearranged with an ETS gene. This is the first description of an EFT with a complete EWS deficiency in the presence of two copies of a rearranged chromosome 22 carrying an interstitial EWS-FLI1 translocation. Absence of EWS protein suggested that it is dispensable for EFT growth. By sequencing of EWS cDNA from unrelated EFTs, we excluded inactivation of EWS as a general mechanism in EFT pathogenesis. Rather, EWS was found to be uniformly expressed in two splicing variants of similar abundancy, EWSalpha and EWSbeta, which differ in a single amino acid. Three EWS negative cell lines were established, which will serve as valuable models to study normal and aberrant EWS function upon reintroduction into the tumor cells.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/pathology , Ribonucleoproteins/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Alleles , Alternative Splicing , Cell Division/genetics , Cell Division/physiology , Child, Preschool , Chromosomes, Human, Pair 22/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Gene Silencing , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Ribonucleoproteins/physiology , Transcription Factors/genetics , Translocation, Genetic , Tumor Cells, Cultured
9.
Cancer Res ; 60(6): 1557-60, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10749123

ABSTRACT

Rearrangement of the EWS gene with FLI1 is thought to occur early in the pathogenesis of Ewing's sarcoma family tumors (EFTs) because the chromosomal aberration is pathognomonic for this disease. Recently, adenovirus (Ad) 5 E1A protein has been reported to induce this gene rearrangement in a variety of cell types. This finding, if generally substantiated, not only suggests an etiological role for viral agents in the generation of oncogenic chromosomal aberrations but would also significantly impact the use of adenoviral vectors for gene therapy. In contrast, we now report on the absence of EWS-FLI1 chimeric products from short- and long-term cultures of stably Ad-transformed cells lines and from transiently E1A-expressing cell lines. In addition, we demonstrate the absence of E1A from EFTs. We conclude that there is no role for Ads in EFT pathogenesis. Consequently, evidence for a viral genesis of tumor-specific gene rearrangements is not available.


Subject(s)
Adenovirus E1A Proteins/physiology , Bone Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Sarcoma, Ewing/genetics , Transcription Factors/genetics , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Blotting, Northern , Blotting, Western , Bone Neoplasms/metabolism , Cell Line , Cell Line, Transformed , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Gene Rearrangement , HeLa Cells , Humans , Oncogene Proteins, Fusion/metabolism , Plasmids/genetics , Proto-Oncogene Protein c-fli-1 , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA-Binding Protein EWS , Sarcoma, Ewing/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured
10.
Lab Invest ; 80(12): 1833-44, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11140696

ABSTRACT

Type 1 and type 2 EWS-FLI1 fusion products result from variation in breakpoint locations arising from the t(11;22)(q24;q12) recurrent chromosomal translocation in Ewing's sarcoma family tumors (EFT). Previously, studies from our institution (updated in the present communication at a median follow-up of more than 6 years) and others suggested a prognostic difference for EFT patients with localized disease depending on the type of EWS-FLI1 fusion present in the tumor. It has been suggested that the observed clinical discrepancies result from different transactivation potentials of the various EWS-FLI1 fusion proteins. In an attempt to identify genes whose expression levels are differentially modulated by structurally different EWS-FLI1 transcription factors, we have used two related PCR-based subtractive approaches, cDNA representational difference analysis (cDNA-RDA) and linker-capture subtraction (LCS) to compare transcript representations in cDNA pools of type 1 versus type 2 EFT cell lines. About 800 clones obtained by the two approaches were analyzed by dot blot hybridization to cDNA pools. Eighty-six clones showing the highest variability in signal intensities on the dot blots were further hybridized to individual EFT cell line RNAs on Northern blots, and four of them were additionally studied by real-time quantitative PCR (RTQ-PCR). Although interindividual variations in gene expression patterns in the range of one- to several-fold were observed, no correlation to specific EWS-FLI1 fusion types could be identified. Among the genes differentially expressed in individual EFT cell lines are several previously implicated in tumor growth, invasion, and metastasis. Although our data may have revealed candidate genes whose composite expression pattern may be relevant for the biology of individual EFT, they do not support a role of distinct EWS-FLI1 fusion types for EFT prognosis based on different transactivation potentials.


Subject(s)
Bone Neoplasms/genetics , Sarcoma, Ewing/genetics , Translocation, Genetic , Blotting, Northern , Bone Neoplasms/mortality , Cell Fusion , Chromosome Mapping , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 22 , Clone Cells , Disease-Free Survival , Humans , Polymerase Chain Reaction , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/mortality , Survival Rate , Tumor Cells, Cultured
11.
Curr Opin Oncol ; 11(4): 275-84, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10416880

ABSTRACT

The ideal cancer therapy would accomodate the specific biology of a tumor and be based upon understanding the mechanisms of malignancy. This vision has been the driving force in cancer research. However, the story of success in clinical cancer management is a story of empirie largely independent from progress in basic research. For the Ewing family of tumors (EFT) comprising Ewing's sarcoma and peripheral primitive neuroectodermal tumor, significant insights into the molecular basis have appeared recently. Some of last year's discoveries may have taken us closer to the identification of the Achilles' heel in this disease. The first clue has been obtained to the mechanism of chromosomal translocation, which constitutes a rate-limiting step in EFT pathogenesis. Also, researchers have progressed in understanding the control of EFT cell proliferation, differentiation, and death. A major role in these processes has been attributed to the EWS-ets gene rearrangement. Specific growth factor circuits appear to be involved in deregulated tumor cell growth. By analogy to heterologous cellular systems, it is possible to postulate an important functional role for CD99(MIC2) as it contributes to the malignant phenotype of EFT. In vitro, as well as the first in vivo, experimental evidence suggests that tumor cell expansion and spread can be counteracted by breaking these physiological pathways. Still, we are far from a tailored biological therapy of EFT. Before this goal may be achieved, we must seek further improvements and diversification of today's standard and intensified treatment regimens.


Subject(s)
Bone Neoplasms/therapy , Sarcoma, Ewing/therapy , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Hematopoietic Stem Cell Transplantation , Humans , Oncogene Proteins, Fusion/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
12.
Oncogene ; 17(5): 603-10, 1998 Aug 06.
Article in English | MEDLINE | ID: mdl-9704926

ABSTRACT

As a result of the t(11;22)(q24;q12) chromosomal translocation characterizing the Ewing family of tumors (ET), the amino terminal portion of EWS, an RNA binding protein of unknown function, is fused to the DNA-binding domain of the ets transcription factor Fli1. The hybrid EWS-Fli1 protein acts as a strong transcriptional activator and, in contrast to wildtype Fli1, is a potent transforming agent. Similar rearrangements involving EWS or the highly homologous TLS with various transcription factors have been found in several types of human tumors. Employing yeast two-hybrid cloning we isolated the seventh largest subunit of human RNA polymerase II (hsRPB7) as a protein that specifically interacts with the amino terminus of EWS. This association was confirmed by in vitro immunocoprecipitation. In nuclear extracts, hsRPB7 was found to copurify with EWS-Fli1 but not with Fli1. Overexpression of recombinant hsRPB7 specifically increased gene activation by EWS-chimeric transcription factors. Replacement of the EWS portion by hsRPB7 in the oncogenic fusion protein restored the transactivating potential of the chimera. Our results suggest that the interaction of the amino terminus of EWS with hsRPB7 contributes to the transactivation function of EWS-Fli1 and, since hsRPB7 has characteristics of a regulatory subunit of RNA polymerase II, may influence promoter selectivity.


Subject(s)
Gelsolin , Oncogene Proteins, Fusion/metabolism , RNA Polymerase II/metabolism , Receptors, Cytoplasmic and Nuclear , Transcription Factors/metabolism , 3T3 Cells , Animals , Artificial Gene Fusion , Binding Sites , Cloning, Molecular , Gene Expression , Humans , Mice , Microfilament Proteins , Nucleic Acid Hybridization , Oncogene Proteins, Fusion/genetics , Proteins/genetics , Proto-Oncogene Protein c-fli-1 , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA-Binding Protein EWS , Saccharomyces cerevisiae , Sarcoma, Ewing , Trans-Activators , Transcription Factors/genetics , Transcriptional Activation , Tumor Cells, Cultured
13.
Nucleic Acids Res ; 26(9): 2252-3, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9547290

ABSTRACT

The yeast two-hybrid system is frequently used to identify protein-protein interactions. Confirming the specificity of candidate clones requires separation and isolation of yeast plasmids, propagation in bacteria and testing combinations of DNA-binding and activation domain hybrids in yeast. In order to simplify this procedure, we developed a rapid method based on PCR amplification of library insert DNAs and in vivo cloning into the activation domain hybrid vector. Reporter gene activity is assayed in parallel for combinations with different DNA-binding domain hybrids. Further characterization of inserts does not require plasmid isolation and intermediate hosts.


Subject(s)
Cloning, Molecular/methods , Protein Binding/genetics , Recombination, Genetic , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Antigens, Viral, Tumor/genetics , DNA-Binding Proteins , Evaluation Studies as Topic , False Positive Reactions , Genetic Vectors , Heterogeneous-Nuclear Ribonucleoproteins , Polymerase Chain Reaction , Ribonucleoproteins/genetics , Simian virus 40/genetics , Time Factors , Transcription Factors/genetics , Transcriptional Activation
14.
Gene ; 210(1): 71-8, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9524226

ABSTRACT

The ETS gene family encodes a group of proteins that function as transcription factors under physiological conditions and, if aberrantly expressed, can lead to cellular transformation. ETS transcription factors are characterized by a unique conserved DNA binding domain. A subset of these proteins is rearranged with EWS in Ewing tumors (ET). We recently described a spectrum of ETS genes coexpressed with EWS-FLI1 in an ETcell line to define proteins that potentially compete in target site selection. We now report on the cloning and characterization of a novel ETS family member, ELFR, displaying 92% homology to ELF-1 in its DNA binding domain while diverging in the rest of the protein. ELFR expression was found in a very tissue restricted pattern with the highest abundancy in placenta. We also report the chromosomal assignment of ELFR and ELF-1 to Xq26 and 13q13, respectively, by means of fluorescence in-situ hybridization (FISH).


Subject(s)
Chromosome Mapping , DNA-Binding Proteins/chemistry , Sarcoma, Ewing/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Chromosomes, Human, Pair 13/genetics , Cloning, Molecular , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Neoplasm/genetics , Genes, Reporter/genetics , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Transcription Factors/genetics , Tumor Cells, Cultured , X Chromosome/genetics
15.
Oncogene ; 15(18): 2225-32, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9393981

ABSTRACT

The pRB cell cycle regulatory cascade is frequently perturbed in neoplasia by overexpression of a component of the pRB-phosphorylating cyclin D1/CDK4 complex or by inactivation of pRB or the CDK4 inhibitors p16 and p15. We investigated the status and expression of p16, p15, CCND1, CDK4 and RB genes in the Ewing family of tumors. P16 loss was observed in 8 of 27 tumors (30%) and in 12 of 23 (52%) tumor cell lines from unrelated patients. There were no discrepancies in the p16 status between primary tumors and the corresponding cell lines and between cell lines established from consecutive tumor samples. p15 was codeleted in most cases but p15 mRNA was absent also in cell lines retaining the gene. In addition, posttranscriptional p16 inactivation was observed in two cases. Although no evidence for CDK4 or CCND1 amplification was obtained, expression of these genes varied considerably in the cell lines in a case specific manner. In wild-type p16 cell lines, pRB expression was lost in one case. Our data indicate that, despite the absence of cytogenetically detectable 9p21 chromosomal aberrations, p16 deletions constitute the most frequent secondary molecular aberration in Ewing tumors so far. These results are discussed in the context of the stage of disease and the clinical outcome of the patients. The potential prognostic impact of these findings remains to be further evaluated.


Subject(s)
Cell Cycle Proteins , Gene Deletion , Genes, p16 , Proto-Oncogene Proteins , Retinoblastoma Protein/physiology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Tumor Suppressor Proteins , Cell Cycle/physiology , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinases/biosynthesis , Gene Expression , Genes, p53 , Humans , Mutation , Prognosis , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Sarcoma, Ewing/metabolism , Transcription Factors/biosynthesis
16.
Br J Cancer ; 74(11): 1693-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8956779

ABSTRACT

The nm23-H1 gene is a putative metastasis-suppressor gene encoding a 17 kDa protein with nucleoside diphosphate kinase activity. Expression of nm23-H1/NDPK-A correlates inversely with the metastasising potential of some human tumours and experimental animal cells. No nm23 expression studies exist for human malignant lymphomas so far. In this study, we examined nm23-H1 expression by Northern and immunohistochemical analysis in 106 primary lymphoma samples from patients with Hodgkin's disease (HD) (n = 15), high-grade non-Hodgkin's lymphoma (NHL) from different lineages (n = 71) and low-grade NHL (n = 20). Both inter- and intra-subtype variations in nm23-H1/NDPK-A expression levels were demonstrated by all disease subtypes. Besides this heterogeneity, a general trend towards highly malignant samples expressing higher nm23-H1/NDPK-A, levels than the low-grade lymphomas was observed. Both adult and childhood HD and high-grade NHL samples exhibited significantly higher NDPK-A expression than the low-grade NHL found only in adults. High nm23-H1/NDPK-A levels in lymphoma samples did not always reflect proliferative activity of tumour cells as monitored by Ki-67 antigen staining. Fifty samples were further investigated for possible mutations in the nm23-H1 coding sequence by means of reverse transcriptase-polymerase chain reaction (RT-PCR) and single-strand conformation polymorphism (SSCP) analysis. No mutation was found by this screening. Our results suggest a role for nm23-H1 expression in the disease aggressiveness of lymphomas.


Subject(s)
Hodgkin Disease/metabolism , Lymphoma, Non-Hodgkin/metabolism , Monomeric GTP-Binding Proteins , Neoplasm Proteins/metabolism , Nucleoside-Diphosphate Kinase , RNA, Messenger/metabolism , Transcription Factors/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Neoplasm/immunology , Child , Child, Preschool , DNA Mutational Analysis , Female , Genes, Tumor Suppressor , Hodgkin Disease/genetics , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Humans , Infant , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , NM23 Nucleoside Diphosphate Kinases , Neoplasm Proteins/genetics , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Transcription Factors/genetics
17.
Cell Growth Differ ; 7(4): 429-37, 1996 Apr.
Article in English | MEDLINE | ID: mdl-9052984

ABSTRACT

Among primitive neuroectodermal tumors, Ewing tumors are characterized by a gene rearrangement recombining the 5'-EWS gene portion with one of two ETS-related proto-oncogenes, FLI-1 or ERG, in roughly 90 and 10% of cases, respectively. We attempted to antagonize EWS/FLI-1 function in Ewing tumor cell lines. As a control, a cell line derived from another small round cell tumor of neuroectodermal origin, neuroblastoma, was used. This cell line was found to express almost identical patterns of ETS proteins except for EWS/FLI-1 and a novel, ELF-related gene product. Stable expression of antisense EWS/FLI-1 cDNA resulted in decreased endogenous EWS/FLI-1 RNA levels and growth reduction of ET cells but not of neuroblastoma cells. DNA-binding FLI-1 derivatives localizing to the nucleus in which the 5'-EWS regulatory domain was replaced by bacterial beta-galactosidase dominantly modulated transcriptional transactivation from a degenerate ETS-binding motif. Transient transfection of these dominant-negative recombinants resulted in a significant decrease in the relative number of mitoses in four Ewing tumor cell lines tested but not in the neuroblastoma cell line. The presented evidence for modulation of tumor cell proliferation by EWS/FLI-1 antagonists suggests a causal role for EWS/FLI-1-mediated gene activation in the malignant transformation of the enigmatic Ewing tumor-precursor cell.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Immediate-Early Proteins , RNA, Antisense/pharmacology , Retroviridae Proteins, Oncogenic/genetics , Sarcoma, Ewing/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Blotting, Northern , Cell Division/drug effects , DNA, Complementary/metabolism , Early Growth Response Protein 1 , Gene Rearrangement , Humans , Neuroblastoma/genetics , Neuroblastoma/metabolism , Plasmids , Proto-Oncogene Protein c-ets-2 , Proto-Oncogene Protein c-fli-1 , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , RNA, Antisense/metabolism , Recombinant Proteins/pharmacology , Repressor Proteins/genetics , Sarcoma, Ewing/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , beta-Galactosidase/genetics
18.
Int J Cancer ; 64(2): 104-11, 1995 Apr 21.
Article in English | MEDLINE | ID: mdl-7542225

ABSTRACT

Expression of nm23-H1/NDPK-A has been reported to correlate inversely with metastasizing potential of rodent experimental cells and some human tumors. In the search for reliable molecular prognostic indicators for Ewing tumors (ET), a group of aggressive presumably neuroectodermal malignancies in children and adolescents, we studied nm23-H1/NDPK-A expression. Northern-blot and RT-PCR analyses were employed to semi-quantificatively measure nm23-H1 mRNA levels in ET cell lines and tissue extracts. A panel of monoclonal antibodies (MAbs) were used to evaluate protein abundance by Western blotting and immunohistochemistry. The nm23-H1/NDPK-A gene was also investigated on the DNA level to define possible genomic alterations. Our results revealed neither nm23-H1 allelic loss nor gene amplification and failed to show any significant variation in nm23-H1 mRNA or NDPK-A protein levels of primary or metastatic ET. NDPK-A protein levels were high and comparable to those of MCF-7 breast-cancer cells and to aggressive stage-IV neuroblastoma cell lines. nm23-H2/NDPK-B expression in ET was slightly more variable but generally lower than in MCF-7 cells. In the immunohistochemical analysis, however, discrepancies in the reactivity patterns with different antibodies were observed. Differential sensitivity to various fixation methods and heat treatment pointed to a structurally polymorphic NDPK-A protein. nm23-H1 expression studies using immunohistochemistry for prognostic counselling should thus be interpreted with caution.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase/analysis , Sarcoma, Ewing/chemistry , Transcription Factors/analysis , Adolescent , Antibodies, Monoclonal , Base Sequence , Blotting, Northern , Blotting, Southern , Blotting, Western , Child , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Molecular Sequence Data , NM23 Nucleoside Diphosphate Kinases , Polymerase Chain Reaction/methods , Prognosis , RNA-Directed DNA Polymerase , Sarcoma, Ewing/enzymology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/secondary
19.
Leukemia ; 8(11): 1929-39, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7967739

ABSTRACT

In malignant non-Hodgkin lymphomas (NHL), cytogenetic analysis may provide prognostic information including prediction of histologic evolution and responsiveness to therapy. In this study, we correlate clinical data and chromosomal aberrations in 70 adult patients with newly diagnosed NHL followed for a median of 20 months. Clonal aberrations were detected in 68/70 patients (97%). Besides t(2;5)(p23;q35), observed exclusively in three patients with anaplastic large cell lymphoma, Ki-1 positive, none of the characteristic aberrations observed was specific for a given histological subtype. Aberrations of chromosome 7 (n = 21) occurred in all histological subtypes together with aberrations of chromosome 3 and of the short arm of chromosome 17. They were clinically associated with a high serum lactate dehydrogenase level (LDH) and a trend to short survival. Anomalies of the long arm of chromosome 13 (n = 10) were found in patients with high grade B-cell lymphomas and bulky disease. In t(14;18)(q32;q21) bearing lymphomas (n = 27), distinct patterns of additional aberrations were observed in low grade and high grade lymphomas: trisomy 3 and trisomy 18 occurred concomitantly in high grade lymphomas (n = 6, p < 0.001) as well as aberrations of 1q, 5q, 6q and +der (18)(q21). In conclusion, cytogenetic analysis provides information about the complexity of genetic changes in NHL. These changes act not only as indicators of disease activity, but influence clinical outcome as demonstrated by their stringent correlation to the International Index and might reveal more general rules of tumor growth and spreading.


Subject(s)
Chromosome Aberrations/pathology , Lymphoma, Non-Hodgkin/pathology , Adolescent , Adult , Aged , Chromosome Aberrations/genetics , Chromosome Disorders , Chromosome Inversion , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 7 , Female , Humans , Karyotyping , L-Lactate Dehydrogenase/blood , Lymph Nodes/pathology , Lymphoma, Non-Hodgkin/genetics , Male , Middle Aged , Prognosis , Survival Analysis , Translocation, Genetic
20.
Klin Padiatr ; 206(4): 196-200, 1994.
Article in German | MEDLINE | ID: mdl-7967416

ABSTRACT

The family of Ewing tumors (ET) is characterised by a unique gene rearrangement which is represented by a translocation t(11;22) (q24;q12) or a deletion del 22q12 in most cytogenetically analysable cases. The recent cloning of the underlying gene fusion provides the basis for the diagnostic detection of minimal amounts of residual tumor cells at resection margins, in blood and bone marrow. In addition, the very first steps in ET tumorigenesis can be studied on a functional basis. In this study, a variety of fusion products were identified with a sensitivity of 10(-6) by means of RT-PCR. In 20 of 22 ET, a gene rearrangement was identified which resulted in the substitution of the effector domain of one of the closely related DNA-binding oncogenes, FLI-1 or ERG, by the transactivating domain of a new gene, EWS. Presumably, the oncogene and consequently its target genes are activated by this type of translocation. If the EWS domain was replaced with a transcriptionally irrelevant domain by transfection of a recombinant gene into ET cells, competition with the endogenous chimeric oncogene-product for DNA-binding was observed resulting in a partial growth inhibition. Activation of FLI-1 has been previously shown to occur as a primary event in Friend virus induced mouse erythroleukemia. During progression of this disease, inactivating p53 mutations have been observed frequently. In contrast, such aberrations were found to be extremely rare in ET.


Subject(s)
Bone Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Chromosome Aberrations , Gene Rearrangement/genetics , Immediate-Early Proteins , Proto-Oncogene Proteins , Sarcoma, Ewing/genetics , Transcription Factors , Adolescent , Bone Marrow/pathology , Bone Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Child , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 22 , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Female , Humans , Male , Neoplastic Cells, Circulating , Proto-Oncogene Protein c-fli-1 , Retroviridae Proteins, Oncogenic/genetics , Sarcoma, Ewing/pathology , Trans-Activators/genetics , Transcriptional Regulator ERG , Translocation, Genetic/genetics , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...