Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Oncogene ; 34(30): 4005-10, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-25284589

ABSTRACT

Cellular transformation is associated with altered glutamine (Gln) metabolism. Tumor cells utilize Gln in the tricarboxylic acid (TCA) cycle to maintain sufficient pools of biosynthetic precursors to support rapid growth and proliferation. However, Gln metabolism also generates NADPH, and Gln-derived glutamate is used for synthesis of glutathione (GSH). As both NADPH and GSH are antioxidants, Gln may also contribute to redox balance in transformed cells. The Hace1 E3 ligase is a tumor suppressor inactivated in diverse human cancers. Hace1 targets the Rac1 GTPase for degradation at Rac1-dependent NADPH oxidase complexes, blocking superoxide generation by the latter. Consequently, loss of Hace1 increases reactive oxygen species (ROS) levels in vitro and in vivo. Given the link between Hace1 loss and increased ROS, we investigated whether genetic inactivation of Hace1 alters Gln metabolism. We demonstrate that mouse embryonic fibroblasts (MEFs) derived from Hace1(-/-) mice are highly sensitive to Gln withdrawal, leading to enhanced cell death compared with wild-type (wt) MEFs, and Gln depletion or chemical inhibition of Gln uptake blocks soft agar colony formation by Hace1(-/-) MEFs. Hace1(-/-) MEFs exhibit increased Gln uptake and ammonia secretion, and metabolic labeling using (13)C-Gln revealed that Hace1 loss increases incorporation of Gln carbons into the TCA cycle intermediates. Gln starvation markedly increases ROS levels in Hace1(-/-) but not in wt MEFs, and treatment with the antioxidant N-acetyl cysteine or the TCA cycle intermediate oxaloacetate efficiently rescues Gln starvation-induced ROS elevation and cell death in Hace1(-/-) MEFs. Finally, Gln starvation increases superoxide levels in Hace1(-/-) MEFs, and NADPH oxidase inhibitors block the induction of superoxide and cell death by Gln starvation. Together, these results suggest that increased ROS production due to Hace1 loss leads to Gln addiction as a mechanism to cope with increased ROS-induced oxidative stress.


Subject(s)
Glutamine/physiology , Reactive Oxygen Species/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis , Cells, Cultured , Mice, Knockout , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
Oncogene ; 32(13): 1735-42, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-22614015

ABSTRACT

The small GTPase Rac1 is a key regulator of cell motility. Multiple mechanisms regulate Rac1 activity including its ubiquitylation and subsequent degradation. Here, we identify the tumour suppressor HACE1 (HECT domain and Ankyrin repeat Containing E3 ubiquitin-protein ligase 1) as an E3 ubiquitin ligase responsible for Rac1 degradation following activation by a migration stimulus. We show that HACE1 and Rac1 interaction is enhanced by hepatocyte growth factor (HGF) signalling, a Rac activator and potent stimulus of cell migration. Furthermore, HACE1 catalyses the poly-ubiquitylation of Rac1 at lysine 147 following its activation by HGF, resulting in its proteasomal degradation. This negative feedback mechanism likely restricts cell motility. Consistent with this, HACE1 depletion is accompanied by increased total Rac1 levels and accumulation of Rac1 in membrane ruffles. Moreover, HACE1-depletion enhances cell migration independently of growth factor stimulation, which may have significance for malignant conversion. A non-ubiquitylatable Rac1 rescues the migration defect of Rac1-null cells to a greater extent than wild-type Rac1. These findings identify HACE1 as an antagonist of cell migration through its ability to degrade active Rac1.


Subject(s)
Cell Movement/genetics , Proteolysis , Ubiquitin-Protein Ligases/physiology , rac1 GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Dogs , HEK293 Cells , Humans , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Polymorphism, Single Nucleotide/physiology , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , rac1 GTP-Binding Protein/genetics
3.
Oncogene ; 31(10): 1334-40, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-21804605

ABSTRACT

ETV6-NTRK3 (EN), a chimeric tyrosine kinase generated by t(12;15) translocations, is a dominantly acting oncoprotein in diverse tumor types. We previously showed that insulin-like growth factor 1 receptor (IGF1R) is essential for EN-mediated oncogenesis and that insulin receptor substrate 1 (IRS1) is constitutively tyrosine phosphorylated and bound by EN in transformed cells. Given that IRS1 is also an adapter for IGF1R, we hypothesized that IRS1 might localize EN to IGF1R at the membrane to activate phosphatidylinositol 3-kinase (PI3K)-Akt, which is critical for EN oncogenesis. In this study, we examined EN/IRS1/IGF1R complexes in detail. We find that both IRS1 and kinase active IGF1R are required for EN transformation, that tyrosine phosphorylated IRS1 is present in high molecular weight complexes with EN and IGF1R, and that EN colocalizes with IGF1R at the plasma membrane. Both IGF1R kinase activity and an intact cytoplasmic Y950 residue, the IRS1-docking site of IGF1R, are required, confirming the importance of the IGF1R/IRS1 interaction for EN oncogenesis. The dual specificity IGF1R and insulin receptor (INSR) inhibitor, BMS-536924, blocks EN transformation activity, cell survival and its interaction with IRS proteins, and induces a striking shift of EN proteins to smaller sized molecular complexes. We conclude that a tripartite complex of EN, IRS1 and IGF1R localizes EN to the membrane and that this is essential for EN-mediated transformation. These findings provide an explanation for the observed IGF1R dependency of EN transformation. Blocking IGF1R kinase activity may, therefore, provide a tractable therapeutic strategy for the many tumor types driven by the EN oncoprotein.


Subject(s)
Cell Membrane/metabolism , Cell Transformation, Neoplastic , Insulin Receptor Substrate Proteins/physiology , Oncogene Proteins, Fusion/physiology , Receptor, IGF Type 1/physiology , Animals , Interleukin-3/pharmacology , Mice , Phosphorylation
4.
Cell Death Differ ; 19(3): 501-10, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21941369

ABSTRACT

Suppression of anoikis after detachment of cancer cells from the extracellular matrix is a key step during metastasis. Here we show that, after detachment, mouse embryonic fibroblasts (MEFs) transformed by K-Ras(V12) or ETV6-NTRK3 (EN) activate a transcriptional response overrepresented by genes related to bioenergetic stress and the AMP-activated protein kinase (AMPK) energy-sensing pathway. Accordingly, AMPK is activated in both transformed and non-transformed cells after detachment, and AMPK deficiency restores anoikis to transformed MEFs. However, AMPK activation represses the mTOR complex-1 (mTORC1) pathway only in transformed cells, suggesting a key role for AMPK-mediated mTORC1 inhibition in the suppression of anoikis. Consistent with this, AMPK-/- MEFs transformed by EN or K-Ras show sustained mTORC1 activation after detachment and fail to suppress anoikis. Transformed TSC1-/- MEFs, which are incapable of suppressing mTORC1, also undergo anoikis after detachment, which is reversed by mTORC1 inhibitors. Furthermore, transformed AMPK-/- and TSC1-/- MEFs both have higher total protein synthesis rates than wild-type controls, and translation inhibition using cycloheximide partially restores their anoikis resistance, indicating a mechanism whereby mTORC1 inhibition suppresses anoikis. Finally, breast carcinoma cell lines show similar detachment-induced AMPK/mTORC1 activation and restoration of anoikis by AMPK inhibition. Our data implicate AMPK-mediated mTORC1 inhibition and suppression of protein synthesis as a means for bioenergetic conservation during detachment, thus promoting anoikis resistance.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anoikis/physiology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line, Transformed , Cell Line, Tumor , Embryo, Mammalian/cytology , Fibroblasts/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , NIH 3T3 Cells , Proteins/genetics , Proteins/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
5.
Cancer Res ; 61(24): 8909-16, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751416

ABSTRACT

There is increasing interest in the potential role of the NTRK family of neurotrophin receptors in human neoplasia. These receptor protein tyrosine kinases (PTKs) are well-known mediators of neuronal cell survival and differentiation, but altered NTRK signaling has also been implicated in mesenchymal, hematopoietic, and epithelial malignancies. We recently identified a novel gene fusion involving one of the neurotrophin receptor genes, NTRK3, in the pediatric solid tumor, congenital fibrosarcoma. In these tumors (and subsequently demonstrated in several other human malignancies), a t(12;15)(p13;q25) rearrangement fuses the 3' portion of the ETV6 gene with exons encoding the PTK domain of NTRK3. The resulting ETV6-NTRK3 fusion protein functions as a chimeric PTK with potent transforming activity. However, previous studies failed to detect interactions between ETV6-NTRK3 and molecules known to link wild-type NTRK3 to its two major effector pathways, namely the Ras-Raf1-Mek1-Erk1/2 mitogenic pathway or the phosphatidylinositol 3'-kinase pathway leading to activation of the AKT survival factor. Therefore, it remains unknown whether ETV6-NTRK3 transformation involves altered NTRK3 signaling. We now report that ETV6-NTRK3 expression in NIH3T3 cells leads to constitutive activation of Mek1 and Akt, as well as to constitutively high expression of cyclin D1. ETV6-NTRK3-induced soft agar colony formation was almost completely abolished by inhibition of either the Ras-Raf1-Mek1-Erk1/2 or the phosphatidylinositol 3'-kinase-Akt pathway. Moreover, this inhibition dramatically reduced expression of cyclin D1. Our results indicate that ETV6-NTRK3 transformation involves a link between known NTRK3 signaling pathways and aberrant cell cycle progression and that Mek1 and Akt activation act synergistically to mediate these effects.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, trkC/physiology , Recombinant Fusion Proteins/physiology , Repressor Proteins/physiology , Signal Transduction/physiology , 3T3 Cells/enzymology , 3T3 Cells/physiology , Animals , Cell Transformation, Neoplastic/drug effects , Cyclin D1/biosynthesis , Cyclin D1/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 1 , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-ets , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/biosynthesis , Receptor, trkC/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction/drug effects , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism , ETS Translocation Variant 6 Protein
6.
Cancer Genet Cytogenet ; 129(2): 165-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566349

ABSTRACT

We report a novel cytogenetic finding in a favorable histology Wilms tumor occurring in a 4-month-old boy. Karyotypic analysis demonstrated a t(6;15)(q21;q21) in all tumor cells examined. This was confirmed using fluorescence in situ hybridization analysis. Molecular analysis of this rearrangement may provide clues to understanding the pathobiology of Wilms tumor.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 6/genetics , Translocation, Genetic/genetics , Wilms Tumor/genetics , Chromosome Painting , Humans , In Situ Hybridization, Fluorescence , Infant , Karyotyping , Male , Ploidies , Tumor Cells, Cultured , White People/genetics , Wilms Tumor/diagnosis , Wilms Tumor/therapy
8.
Hum Pathol ; 32(5): 513-20, 2001 May.
Article in English | MEDLINE | ID: mdl-11381370

ABSTRACT

Mixed epithelial and stromal tumor of the kidney is a recently recognized neoplasm that occurs almost exclusively in perimenopausal women. Because it frequently contains areas of smooth muscle in which epithelial structures are embedded, some have concluded that it is the adult form of congenital mesoblastic nephroma. Others have concluded that the morphology and epidemiology of mixed epithelial and stromal tumor indicate that it is unrelated to congenital mesoblastic nephroma. Although the genetic alterations of mixed epithelial and stromal tumor have not been previously elucidated, much is known about the genetic alterations of cellular congenital mesoblastic nephroma. The present study was undertaken to determine if mixed epithelial and stromal tumors have any of the genetic alterations recognized as typical of cellular congenital mesoblastic nephroma. RNA extraction was performed on formalin-fixed, paraffin-embedded tissue from 7 mixed epithelial and stromal tumors followed by reverse-transcription polymerase chain reaction to detect the ETV6-NTRK3 gene fusion. Fluorescent in situ hybridization with centromere-specific probes for chromosomes 8, 11, and 17 was performed to evaluate polyploidy of these chromosomes in 11 cases of mixed epithelial and stromal tumor. None of the mixed epithelial and stromal tumors showed any of these genetic alterations. We conclude that mixed epithelial and stromal tumor of the kidney lacks the genetic alterations typical of cellular congenital mesoblastic nephroma, is unrelated to it, and the appellation "adult mesoblastic nephroma" should not be used for these tumors.


Subject(s)
Epithelial Cells/pathology , Kidney Neoplasms/genetics , Nephroma, Mesoblastic/congenital , Nephroma, Mesoblastic/genetics , Repressor Proteins , Stromal Cells/pathology , Adult , Aged , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 8 , DNA-Binding Proteins/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Kidney Neoplasms/pathology , Menopause , Middle Aged , Oncogene Proteins, Fusion , Ploidies , Proto-Oncogene Proteins c-ets , Receptor, trkC/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Translocation, Genetic , ETS Translocation Variant 6 Protein
9.
Cancer Genet Cytogenet ; 126(2): 128-33, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11376805

ABSTRACT

Undifferentiated embryonal sarcoma (UES) is the third most common hepatic malignancy in children. Previous reports have described a broad range of complex cytogenetic abnormalities in individual cases of hepatic UES. Herein we report the cytogenetic findings of six cases of hepatic UES at our institution analyzed by conventional cytogenetic methods and comparative genomic hybridization (CGH). The CGH demonstrated several chromosomal gains and deletions in each case, but there was no specific abnormality seen in every case. Patterns of chromosomal changes included gains of chromosome 1q (four cases), 5p (four cases), 6q (four cases), 8p (three cases), and 12q (three cases), and losses of chromosome 9p (two cases), 11p (two cases), and chromosome 14 (three cases). The three cases in which CGH showed gains in the 12q region were studied specifically for amplifications of MDM2 and CDK4, two genes that have been shown to be amplified in other soft tissue sarcomas. However, Southern analysis showed no amplification of MDM2 or CDK4 in these three cases. Further analysis will be needed to determine the critical events in the pathogenesis of these malignant pediatric liver tumors.


Subject(s)
Chromosome Aberrations , Liver Neoplasms/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Nucleic Acid Hybridization , Sarcoma/genetics , Blotting, Southern , Humans , Liver Neoplasms/pathology , Neoplasms, Germ Cell and Embryonal/pathology , Sarcoma/pathology
10.
Oncogene ; 20(1): 48-57, 2001 Jan 04.
Article in English | MEDLINE | ID: mdl-11244503

ABSTRACT

Alveolar soft part sarcoma (ASPS) is an unusual tumor with highly characteristic histopathology and ultrastructure, controversial histogenesis, and enigmatic clinical behavior. Recent cytogenetic studies have identified a recurrent der(17) due to a non-reciprocal t(X;17)(p11.2;q25) in this sarcoma. To define the interval containing the Xp11.2 break, we first performed FISH on ASPS cases using YAC probes for OATL1 (Xp11.23) and OATL2 (Xp11.21), and cosmid probes from the intervening genomic region. This localized the breakpoint to a 160 kb interval. The prime candidate within this previously fully sequenced region was TFE3, a transcription factor gene known to be fused to translocation partners on 1 and X in some papillary renal cell carcinomas. Southern blotting using a TFE3 genomic probe identified non-germline bands in several ASPS cases, consistent with rearrangement and possible fusion of TFE3 with a gene on 17q25. Amplification of the 5' portion of cDNAs containing the 3' portion of TFE3 in two different ASPS cases identified a novel sequence, designated ASPL, fused in-frame to TFE3 exon 4 (type 1 fusion) or exon 3 (type 2 fusion). Reverse transcriptase PCR using a forward primer from ASPL and a TFE3 exon 4 reverse primer detected an ASPL-TFE3 fusion transcript in all ASPS cases (12/12: 9 type 1, 3 type 2), establishing the utility of this assay in the diagnosis of ASPS. Using appropriate primers, the reciprocal fusion transcript, TFE3-ASPL, was detected in only one of 12 cases, consistent with the non-reciprocal nature of the translocation in most cases, and supporting ASPL-TFE3 as its oncogenically significant fusion product. ASPL maps to chromosome 17, is ubiquitously expressed, and matches numerous ESTs (Unigene cluster Hs.84128) but no named genes. The ASPL cDNA open reading frame encodes a predicted protein of 476 amino acids that contains within its carboxy-terminal portion of a UBX-like domain that shows significant similarity to predicted proteins of unknown function in several model organisms. The ASPL-TFE3 fusion replaces the N-terminal portion of TFE3 by the fused ASPL sequences, while retaining the TFE3 DNA-binding domain, implicating transcriptional deregulation in the pathogenesis of this tumor, consistent with the biology of several other translocation-associated sarcomas. Oncogene (2001) 20, 48 - 57.


Subject(s)
Chromosomes, Human, Pair 17/genetics , DNA-Binding Proteins/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Sarcoma, Alveolar Soft Part/genetics , Transcription Factors/genetics , Translocation, Genetic , X Chromosome/genetics , Adolescent , Adult , Amino Acid Sequence , Axilla , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Blotting, Southern , Child , Chromosome Breakage , Chromosome Mapping , DNA, Complementary/isolation & purification , Extremities , Female , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins , Karyotyping , Male , Molecular Sequence Data , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/isolation & purification , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/isolation & purification , Organ Specificity/genetics , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, Protein , Tumor Cells, Cultured
12.
Am J Surg Pathol ; 24(7): 937-46, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10895816

ABSTRACT

Congenital fibrosarcoma (CFS) is a pediatric spindle cell tumor of the soft tissues that usually presents before the age of 2 years. Although these tumors display histologic features of malignancy and frequently recur, they have a relatively good prognosis and only rarely metastasize. CFS must therefore be differentiated from more aggressive spindle cell sarcomas that occur during childhood, particularly adult-type fibrosarcoma (ATFS), which can have an identical morphology. CFS must also be distinguished from benign but cellular fibroblastic lesions of the same age group, including infantile fibromatosis (IFB) and myofibromatosis (MFB). Unfortunately, standard pathologic examination often does not differentiate CFS from these other conditions. The authors recently identified a novel chromosomal translocation in CFS, t(12;15)(p13;q25), which gives rise to an ETV6-NTRK3 gene fusion. They subsequently developed reverse transcription-polymerase chain reaction (RT-PCR) assays that can detect ETV6-NTRK3 fusion transcripts in CFS frozen or paraffin-embedded tumor specimens. To confirm the use of this assay in the differential diagnosis of CFS, they have screened a larger series of childhood pediatric spindle cell lesions for ETV6-NTRK3 gene fusions, including 11 cases of CFS, 13 malignant spindle cell tumors (including ATFS), and 38 benign spindle cell tumors (including IFB and MFB). Of the 11 cases diagnosed as CFS, 10 showed the ETV6-NTRK3 gene fusion, whereas none of the 51 other malignant or benign spindle cell tumors demonstrated this fusion gene. They also compared their RT-PCR findings with those of conventional cytogenetics and with immunohistochemical detection of the ETV6-NTRK3 protein using antisera to NTRK3. They conclude that RT-PCR analysis is superior to these techniques for the detection of the ETV6-NTRK3 gene fusion in pediatric spindle cell tumors, and it is a reliable and specific modality for the diagnosis of CFS.


Subject(s)
Artificial Gene Fusion , DNA-Binding Proteins/genetics , Fibrosarcoma/genetics , Receptor, trkC/genetics , Repressor Proteins , Soft Tissue Neoplasms/genetics , Transcription Factors/genetics , Adolescent , Adult , Child , Child, Preschool , DNA, Neoplasm/analysis , DNA-Binding Proteins/analysis , Diagnosis, Differential , Female , Fibroma/diagnosis , Fibrosarcoma/chemistry , Fibrosarcoma/congenital , Fibrosarcoma/diagnosis , Humans , Immunoenzyme Techniques , Infant , Infant, Newborn , Karyotyping , Male , Myofibromatosis/diagnosis , Proto-Oncogene Proteins c-ets , Receptor, trkC/analysis , Reverse Transcriptase Polymerase Chain Reaction , Soft Tissue Neoplasms/chemistry , Soft Tissue Neoplasms/congenital , Soft Tissue Neoplasms/diagnosis , Transcription Factors/analysis , Translocation, Genetic , ETS Translocation Variant 6 Protein
13.
Cancer Res ; 60(6): 1536-40, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10749119

ABSTRACT

Ewing sarcoma-specific chromosomal translocations fuse the EWS gene to a subset of ets transcription factor family members, most commonly the FLI1 gene and less frequently ERG, ETV1, E1A-F, or FEV. These fusion proteins are thought to act as aberrant transcription factors that bind DNA through their ets DNA binding domain. Recently, we have shown (K-B. Hahm et al., Nat. Genet., 23: 222-227, 1999) that the transforming growth factor beta (TGF-beta) type II receptor (TGF-beta RII), a putative tumor suppressor gene, is a target of the EWS-FLI1 fusion protein. Here, we also examined effects of EWS-ETV1 and EWS-ERG on expression of the TGF-beta RII gene. We show that relative to the control, NIH-3T3 cell lines stably transfected with the EWS-FLI1, EWS-ERG, or EWS-ETV1 gene fusion express reduced levels of TGF-beta RII mRNA and protein, and that these cell lines have reduced TGF-beta sensitivity. Cotransfection of these fusion genes and the TGF-beta RII promoter suppresses TGF-beta RII promoter activity and also FLI1-, ERG-, or ETV1-induced promoter activity. These results indicate that transcriptional repression of TGF-beta RII is an important target of the EWS-FLI1, EWS-ERG, or EWS-ETV1 oncogene, and that EWS-ets fusion proteins may function as dominant negative forms of ets transcription factors.


Subject(s)
Bone Neoplasms/genetics , DNA-Binding Proteins , Oncogene Proteins, Fusion/genetics , Receptors, Transforming Growth Factor beta/genetics , Sarcoma, Ewing/genetics , Trans-Activators , 3T3 Cells , Animals , Gene Expression Regulation , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Oncogene Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases , Proto-Oncogene Protein c-fli-1 , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein EWS , Radioligand Assay , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Regulator ERG , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
14.
Oncogene ; 19(7): 906-15, 2000 Feb 17.
Article in English | MEDLINE | ID: mdl-10702799

ABSTRACT

The congenital fibrosarcoma t(12;15)(p13;q25) rearrangement splices the ETV6 (TEL) gene on chromosome 12p13 in frame with the NTRK3 (TRKC) neurotrophin-3 receptor gene on chromosome 15q25. Resultant ETV6-NTRK3 fusion transcripts encode the helix - loop - helix (HLH) dimerization domain of ETV6 fused to the protein tyrosine kinase (PTK) domain of NTRK3. We show here that ETV6-NTRK3 homodimerizes and is capable of forming heterodimers with wild-type ETV6. Moreover, ETV6-NTRK3 has PTK activity and is autophosphorylated on tyrosine residues. To determine if the fusion protein has transforming activity, NIH3T3 cells were infected with recombinant retroviral vectors carrying the full-length ETV6-NTRK3 cDNA. These cells exhibited a transformed phenotype, grew macroscopic colonies in soft agar, and formed tumors in severe combined immunodeficient (SCID) mice. We hypothesize that chimeric proteins mediate transformation by dysregulating NTRK3 signal transduction pathways via ligand-independent dimerization and PTK activation. To test this hypothesis, we expressed a series of ETV6-NTRK3 mutants in NIH3T3 cells and assessed their transformation activities. Deletion of the ETV6 HLH domain abolished dimer formation with either ETV6 or ETV6-NTRK3, and cells expressing this mutant protein were morphologically non-transformed and failed to grow in soft agar. An ATP-binding mutant failed to autophosphorylate and completely lacked transformation activity. Mutants of the three NTRK3 PTK activation-loop tyrosines had variable PTK activity but had limited to absent transformation activity. Of a series of signaling molecules well known to bind to wild-type NTRK3, only phospholipase-Cgamma (PLCgamma) associated with ETV6-NTRK3. However, a PTK active mutant unable to bind PLCgamma did not show defects in transformation activity. Our studies confirm that ETV6-NTRK3 is a transforming protein that requires both an intact dimerization domain and a functional PTK domain for transformation activity. Oncogene (2000) 19, 906 - 915.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Line, Transformed/enzymology , DNA-Binding Proteins/genetics , Receptor, trkC/genetics , Recombinant Fusion Proteins/genetics , Repressor Proteins , Transcription Factors/genetics , Translocation, Genetic , 3T3 Cells , Animals , Cell Line, Transformed/metabolism , GRB2 Adaptor Protein , Helix-Loop-Helix Motifs/genetics , Humans , Isoenzymes/metabolism , Mice , Mice, SCID , Molecular Sequence Data , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma , Protein Kinases/genetics , Protein Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-ets , Receptor, trkC/biosynthesis , Receptor, trkC/chemistry , Receptor, trkC/metabolism , Type C Phospholipases/metabolism , src Homology Domains/genetics , ETS Translocation Variant 6 Protein
15.
J Pediatr Surg ; 34(11): 1641-4, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10591560

ABSTRACT

PURPOSE: This study was undertaken to review the authors' clinical experience with undifferentiated embryonal sarcoma of the liver (UES) in children, focusing on the clinical presentation and results of treatment. METHODS: A retrospective analysis of all children who have undergone treatment for UES during the 15-year period from 1984 through 1998 was performed. RESULTS: Seven patients (4 boys and 3 girls) ranging in age from 20 months to 12 years at the time of diagnosis were identified. All presented with large abdominal masses and normal liver function test results. All underwent complete tumor resection; trisegmentectomy was required in 4 of these cases. All patients received postoperative chemotherapy. Two patients suffered tumor recurrence at 12 and 29 months; both of these patients died of their disease. Another patient died of complications related to chemotherapy. The other 4 patients are alive with no evidence of disease after 19 to 150 months' follow-up. CONCLUSIONS: Undifferentiated embryonal sarcoma of the liver presents as a large hepatic tumor. Operative resection is difficult, but combined with adjuvant chemotherapy offers the best hope for cure.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hepatectomy/methods , Liver Neoplasms/therapy , Neoplasms, Germ Cell and Embryonal/therapy , Child , Child, Preschool , Combined Modality Therapy , Disease-Free Survival , Female , Follow-Up Studies , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Neoplasms, Germ Cell and Embryonal/mortality , Neoplasms, Germ Cell and Embryonal/pathology , Retrospective Studies , Survival Rate , Treatment Outcome
16.
J Clin Oncol ; 17(6): 1809-14, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10561219

ABSTRACT

PURPOSE: There are a variety of solid tumors in which alternative chromosomal translocations generate related fusion products. In alveolar rhabdomyosarcoma and synovial sarcoma, these variant fusions have been found to have major clinical significance. We investigated whether the two alternative gene fusion products, EWS-FLI1 and EWS-ERG, define different clinical subsets within the Ewing's sarcoma family of tumors. PATIENTS AND METHODS: We selected 30 cases of Ewing's sarcoma with the EWS-ERG gene fusion and 106 cases with the EWS-FLI1 fusion. Clinical data were obtained for each case and compared with the molecular diagnostic findings. RESULTS: There were no significant clinical differences observed between the two groups in age of diagnosis, sex, metastasis at diagnosis, primary site, event-free survival, or overall survival. CONCLUSION: Differences in the C-terminal partner in the Ewing's sarcoma family gene fusions are not associated with significant phenotypic differences.


Subject(s)
Bone Neoplasms/genetics , DNA-Binding Proteins , Oncogene Proteins, Fusion/genetics , Oncogene Proteins/genetics , Sarcoma, Ewing/genetics , Trans-Activators , Transcription Factors/genetics , Adolescent , Adult , Bone Neoplasms/diagnosis , Bone Neoplasms/mortality , Bone Neoplasms/therapy , Disease-Free Survival , Female , Humans , Male , Prognosis , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/mortality , Sarcoma, Ewing/therapy , Survival Rate , Transcriptional Regulator ERG , Translocation, Genetic/genetics , Treatment Outcome
17.
Nat Genet ; 23(2): 222-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508522

ABSTRACT

Chromosomal translocations resulting in the expression of chimaeric transcription factors are frequently observed in tumour cells, and have been suggested to be a common mechanism in human carcinogenesis. Ewing sarcoma and related peripheral primitive neuroectodermal tumours share recurrent translocations that fuse the gene EWSR1 (formerly EWS) from 22q-12 to FLI1 and genes encoding other ETS transcription factors (which bind DNA through the conserved ETS domain). It has been shown that transduction of the gene EWSR1-FLI1 (encoding EWS-FLI1 protein) can transform NIH3T3 cells, and that mutants containing a deletion in either the EWS domain or the DNA-binding domain in FLI1 lose this ability. This indicates that the EWS-FLI1 fusion protein may act as an aberrant transcription factor, but the exact mechanism of oncogenesis remains unknown. Because ETS transcription factors regulate expression of TGFBR2 (encoding the TGF-beta type II receptor, TGF-beta RII; Refs 9,14), a putative tumour suppressor gene, we hypothesized that TGFBR2 may be a target of the EWS-FLI1 fusion protein. We show here that Ewing sarcoma [corrected] (ES) cell lines with the EWSR1-FLI1 fusion have reduced TGF-beta sensitivity, and that fusion-positive ES cells and primary tumours both express low or undetectable levels of TGFBR2 mRNA and protein product. Co-transfection of FLI1 and the TGFBR2 promoter induces promoter activity, whereas EWSR1-FLI1 leads to suppression of TGFBR2 promoter activity and FLI1-induced promoter activity. Introduction of EWSR1-FLI1 into cells lacking the EWSR1-FLI1 fusion suppresses TGF-beta RII expression, whereas antisense to EWSR1-FLI1 in ES cell lines positive for this gene fusion restores TGF-beta RII expression. Furthermore, introduction of normal TGF-beta RII into ES cell lines restores TGF-beta sensitivity and blocks tumorigenicity. Our results implicate TGF-beta RII as a direct target of EWS-FLI1.


Subject(s)
Oncogene Proteins, Fusion/physiology , Proto-Oncogene Proteins , Receptors, Transforming Growth Factor beta/genetics , Transcription Factors/physiology , Animals , Cell Line , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Nude , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases , Proto-Oncogene Protein c-fli-1 , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein EWS , Receptor, Transforming Growth Factor-beta Type II , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Sequence Deletion , Trans-Activators/genetics , Transcription Factors/genetics , Transfection , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
18.
Lab Invest ; 79(12): 1535-43, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10616204

ABSTRACT

Ewing's sarcoma is the least differentiated member of the peripheral primitive neuroectodermal (pPNET) tumor family. Chromosomal translocations involving the EWS gene and five different Ets family transcription factor genes create fusion genes encoding aberrant transcription factors and are implicated in the vast majority of Ewing's sarcoma cases. Here, NIH 3T3 fibroblasts were infected with control (tk-neo or RAS) and two different EWS/ETS-expressing retroviruses. In vitro studies of established polyclonal lines expressing the two EWS/ETS genes, either EWS/FLI1 or EWS/ETV1, showed induction of cytokeratin 15 gene expression. Both fusion genes also caused characteristic gross morphologic, histologic, and ultrastructural changes in NIH 3T3 cells when transformed cell lines were injected into CB-17-scid mice. Native NIH 3T3 cells with a spindled cell morphology were converted to polygonal cells with high nucleo-cytoplasmic ratios that continued to express abundant cytokeratin. Extracellular collagen deposition was abolished, rough endoplasmic reticulum was markedly diminished, and rudimentary cell-cell attachments appeared. Most strikingly, neurosecretory-type dense core granules like those seen in pPNET were now evident. This murine model, created in mesenchyme-derived NIH 3T3 cells, demonstrated new characteristics of both neuroectodermal and epithelial differentiation and resembled small round cell tumors microscopically.


Subject(s)
Cell Differentiation , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/genetics , Ribonucleoproteins/genetics , Transcription Factors/genetics , 3T3 Cells , Animals , Cell Line, Transformed , Ectoderm/cytology , Epithelial Cells/cytology , Heterogeneous-Nuclear Ribonucleoproteins , Immunohistochemistry , Mice , Mice, SCID , Neoplasm Invasiveness , Neuroectodermal Tumors, Primitive/genetics , Neuroectodermal Tumors, Primitive/pathology , Neuroectodermal Tumors, Primitive/ultrastructure , Proto-Oncogene Proteins c-ets , RNA-Binding Protein EWS , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Sarcoma, Ewing/ultrastructure
19.
Cancer Res ; 58(22): 5046-8, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9823307

ABSTRACT

Congenital mesoblastic nephroma (CMN) is an infantile spindle cell tumor of the kidney that is subdivided into "classical" and "cellular" forms based on the degree of cellularity and mitotic activity. The histogenesis of CMN remains obscure, but relationships to other pediatric renal neoplasms have been proposed. However, cellular CMN is virtually identical histologically to congenital fibrosarcoma (CFS), a malignant tumor of fibroblasts in children of the same age group. Moreover, cytogenetic studies have reported common trisomies in CFS and cellular CMN, particularly of chromosome 11. We show here that t(12;15)(p13;q25)-associated ETV6-NTRK3 gene fusions described in CFS are also present in cellular CMN. ETV6-NTRK3 chimeric transcripts were detected in 8 of 9 cellular CMNs and 2 of 2 mixed CMNs. In contrast, all of the four classical CMNs tested were negative, as were cases of Wilms' tumor and clear cell sarcoma of the kidney. Moreover, we found trisomy 11 only in cellular or mixed CMNs with the ETV6-NTRK3 gene fusion. Our studies indicate that classical and cellular CMN have different genetic features and support the concept that cellular CMN is histogenetically related to CFS. They also provide insight into potential mechanisms involved in the transformation of the classical into the cellular form of CMN.


Subject(s)
Chromosomes, Human, Pair 11/genetics , DNA-Binding Proteins/genetics , Fibrosarcoma/genetics , Kidney Neoplasms/genetics , Nephroma, Mesoblastic/genetics , Oncogene Proteins, Fusion/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/genetics , Repressor Proteins , Transcription Factors/genetics , Trisomy/genetics , Child, Preschool , Female , Fibrosarcoma/congenital , Humans , Infant , Infant, Newborn , Kidney Neoplasms/congenital , Male , Nephroma, Mesoblastic/congenital , Proto-Oncogene Proteins c-ets , Receptor, trkC , ETS Translocation Variant 6 Protein
20.
Nucleic Acids Res ; 26(12): 3059-65, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9611255

ABSTRACT

The identification of differential gene expressionbetween cells is a frequent goal in modern biological research. Here we demonstrate the coupling of representational difference analysis (RDA) of cDNA with microarray analysis of the output for high throughput screening. Two primary Ewing's sarcoma tissue samples with different biological behavior in vivo were compared by RDA: one which was metastatic and progressed rapidly; the other localized and successfully treated. A modified RDA protocol that minimizes the necessary starting material was employed. After a reduced number of subtractive rounds, the output of RDA was shotgun cloned into a plasmid vector. Inserts from individual colonies from the subtracted library were amplified with vector-specific primers and arrayed at high density on glass slides. The arrays were then hybridized with differentially fluorescently labeled starting amplicons from the two tissues and fluorescent signals were measured at each DNA spot. We show that the relative amounts of fluorescent signal correlate well with the abundance of fragments in the RDA amplicon and in the starting mRNA. In our system, we analyzed 192 products and 173 (90%) were appropriately detected as being >2-fold differentially expressed. Fifty unique, differentially expressed clones were identified. Therefore, the use of RDA essentially provides an enriched library of differentially expressed genes, while analysis of this library with microarrays allows rapid and reproducible screening of thousands of DNA molecules simultaneously. The coupling of these two techniques in this system resulted in a large pool of differentially expressed genes.


Subject(s)
DNA, Complementary/analysis , DNA, Neoplasm/analysis , Gene Expression Regulation, Neoplastic , Molecular Probe Techniques , Sarcoma, Ewing/chemistry , 3T3 Cells , Animals , Cloning, Molecular/methods , Gene Library , Genes, Neoplasm , Humans , Mice , Nucleic Acid Hybridization , Polymerase Chain Reaction/methods , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL