Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 474: 1-14, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31911079

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft tissue malignancy in childhood and adolescence. Patients with the most aggressive histological variant have an unfavorable prognosis due to a high metastasis incidence. Lysyl oxidase-like 2 (LOXL2) is a lysyl oxidase, member of a family of extracellular matrix (ECM) crosslinking enzymes that recently have emerged as important regulators of tumor progression and metastasis. We report that LOXL2 is overexpressed in RMS, suggesting a potential role for LOXL2 in RMS oncogenic progression. Consistently, transient and stable LOXL2 knockdown decreased cell migratory and invasive capabilities in two ARMS cell lines. Furthermore, introduction of LOXL2 in RMS non-expressing cells using wild type or mutated (catalytically inactive) constructs resulted in increased cell migration, cell invasion and number and incidence of spontaneous lung metastasis in vivo, independently of its catalytic activity. To further study the molecular mechanism associated with LOXL2 expression, a pull-down assay on LOXL2-transfected cells was performed and analyzed by mass spectrometry. The intermediated filament protein vimentin was validated as a LOXL2-interactor. Thus, our results suggest an oncogenic role of LOXL2 in RMS by regulating cytoskeleton dynamics and cell motility capabilities.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Biomarkers, Tumor/metabolism , Carcinogenesis/pathology , Lung Neoplasms/secondary , Rhabdomyosarcoma, Alveolar/pathology , Amino Acid Oxidoreductases/genetics , Animals , Apoptosis , Biocatalysis , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Movement , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Int J Cancer ; 143(5): 1188-1201, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29582409

ABSTRACT

Ewing sarcoma (ES) is the second most common bone malignancy affecting children and young adults with poor prognosis due to high metastasis incidence. Our group previously described that EphA2, a tyrosine kinase receptor, promotes angiogenesis in Ewing sarcoma (ES) cells via ligand-dependent signaling. Now we wanted to explore EphA2 ligand-independent activity, controlled upon phosphorylation at S897 (p-EphA2S897 ), as it has been linked to metastasis in several malignancies. By reverse genetic engineering we explored the phenotypic changes after EphA2 removal or reintroduction. Gene expression microarray was used to identify key players in EphA2 signaling. Mice were employed to reproduce metastatic processes from orthotopically implanted engineered cells. We established a correlation between ES cells aggressiveness and p-EphA2S897 . Moreover, stable overexpression of EphA2 in low EphA2 expression ES cells enhanced proliferation and migration, but not a non-phosphorylable mutant (S987A). Consistently, silencing of EphA2 reduced tumorigenicity, migration and invasion in vitro, and lung metastasis incidence in experimental and spontaneous metastasis assays in vivo. A gene expression microarray revealed the implication of EphA2 in cell signaling, cellular movement and survival. ADAM19 knockdown by siRNA technology strongly reproduced the negative effects on cell migration observed after EphA2 silencing. Altogether, our results suggest that p-EphA2S897 correlates with aggressiveness in ES, so blocking its function may be a promising treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/pathology , Cell Movement , Lung Neoplasms/secondary , Receptor, EphA2/metabolism , Sarcoma, Ewing/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Proliferation , Female , Follow-Up Studies , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic , Phosphorylation , Prognosis , Receptor, EphA2/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Lett ; 386: 196-207, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27894957

ABSTRACT

Epigenetic modifications have been shown to be important in developmental tumors as Ewing sarcoma. We profiled the DNA methylation status of 15 primary tumors, 7 cell lines, 10 healthy tissues and 4 human mesenchymal stem cells lines samples using the Infinium Human Methylation 450K. Differential methylation analysis between Ewing sarcoma and reference samples revealed 1166 hypermethylated and 864 hypomethylated CpG sites (Bonferroni p < 0.05, δ-ß-value with absolute difference of >0.20) corresponding to 392 and 470 genes respectively. Gene Ontology analysis of genes differentially methylated in Ewing sarcoma samples showed a significant enrichment of developmental genes. Membrane and cell signal genes were also enriched, among those, 11 were related to caveola formation. We identified differential hypermethylation of CpGs located in the body and S-Shore of the PTRF gene in Ewing sarcoma that correlated with its repressed transcriptional state. Reintroduction of PTRF/Cavin-1 in Ewing sarcoma cells revealed a role of this protein as a tumor suppressor. Restoration of caveolae in the membrane of Ewing sarcoma cells, by exogenously reintroducing PTRF, disrupts the MDM2/p53 complex, which consequently results in the activation of p53 and the induction of apoptosis.


Subject(s)
Bone Neoplasms/genetics , Caveolin 1/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling/methods , Genes, Tumor Suppressor , RNA-Binding Proteins/genetics , Sarcoma, Ewing/genetics , Animals , Apoptosis , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Caveolin 1/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA-Binding Proteins/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Signal Transduction , Spain , Transfection , Tumor Burden , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Oncotarget ; 7(35): 56889-56903, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27487136

ABSTRACT

Ewing sarcoma (ES) is a bone and soft tissue sarcoma affecting mostly children and young adults. Caveolin-1 (CAV1) is a well-known target of EWS/FLI1, the main driver of ES, with an oncogenic role in ES. We have previously described how CAV1 is able to induce metastasis in ES via matrix metalloproteinase-9 (MMP-9). In the present study we showed how CAV1 silencing in ES reduced MEK1/2 and ERK1/2 phosphorylation. Accordingly, chemical inhibition of MEK1/2 resulted in reduction in MMP-9 expression and activity that correlated with reduced migration and invasion. IQ Motif Containing GTPase Activating Protein 1 (IQGAP1) silencing reduced MEK1/2 and ERK1/2 phosphorylation and MMP-9 expression. Furthermore, IQGAP1 silenced cells showed a marked decrease in their migratory and invasive capacity. We demonstrated that CAV1 and IQGAP1 localize in close proximity at the cellular edge, thus IQGAP1 could be the connecting node between CAV1 and MEK/ERK in ES metastatic phenotype. Analysis of the phosphorylation profile of CAV1-silenced cells showed a decrease of p-ribosomal protein S6 (RPS6). RPS6 can be phosphorylated by p90 ribosomal S6 kinases (RSK) proteins. CAV1-silenced cells showed reduced levels of p-RSK1 and treatment with U0126 provoked the same effect. Despite not affecting ERK1/2 and RPS6 phosphorylation status neither MMP-9 expression nor activity, RSK1 silencing resulted in a reduced migratory and invasive capacity in vitro and reduced incidence of metastases in vivo in a novel orthotopic model. The present work provides new insights into CAV1-driven metastatic process in ES unveiling novel key nodes.


Subject(s)
Caveolin 1/metabolism , MAP Kinase Signaling System , Matrix Metalloproteinase 9/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Animals , Cell Line, Tumor , Cell Movement , Extracellular Matrix/metabolism , Female , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Oncogene Proteins, Fusion/metabolism , Phosphorylation , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Ribosomal Protein S6/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , ras GTPase-Activating Proteins/metabolism
5.
Front Oncol ; 5: 82, 2015.
Article in English | MEDLINE | ID: mdl-25905041

ABSTRACT

Cell death can occur through different mechanisms, defined by their nature and physiological implications. Correct assessment of cell death is crucial for cancer therapy success. Sarcomas are a large and diverse group of neoplasias from mesenchymal origin. Among cell death types, apoptosis is by far the most studied in sarcomas. Albeit very promising in other fields, regulated necrosis and other cell death circumstances (as so-called "autophagic cell death" or "mitotic catastrophe") have not been yet properly addressed in sarcomas. Cell death is usually quantified in sarcomas by unspecific assays and in most cases the precise sequence of events remains poorly characterized. In this review, our main objective is to put into context the most recent sarcoma cell death findings in the more general landscape of different cell death modalities.

6.
Oncotarget ; 5(20): 9744-55, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25313138

ABSTRACT

Rhabdomyosarcoma is the most common soft tissue sarcoma of childhood and adolescence. Despite advances in therapy, patients with histological variant of rhabdomyosarcoma known as alveolar rhabdomyosarcoma (ARMS) have a 5-year survival of less than 30%. Caveolin-1 (CAV1), encoding the structural component of cellular caveolae, is a suggested tumor suppressor gene involved in cell signaling. In the present study we report that compared to other forms of rhabdomyosarcoma (RMS) CAV1 expression is either undetectable or very low in ARMS cell lines and tumor samples. DNA methylation analysis of the promoter region and azacytidine-induced re-expression suggest the involvement of epigenetic mechanisms in the silencing of CAV1. Reintroduction of CAV1 in three of these cell lines impairs their clonogenic capacity and promotes features of muscular differentiation. In vitro, CAV1-expressing cells show high expression of Caveolin-3 (CAV3), a muscular differentiation marker. Blockade of MAPK signaling is also observed. In vivo, CAV1-expressing xenografts show growth delay, features of muscular differentiation and increased cell death. In summary, our results suggest that CAV1 could function as a potent tumor suppressor in ARMS tumors. Inhibition of CAV1 function therefore, could contribute to aberrant cell proliferation, leading to ARMS development.


Subject(s)
Caveolin 1/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Rhabdomyosarcoma, Alveolar/pathology , Animals , Caveolin 1/genetics , Cell Death/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Down-Regulation , Epigenomics , Gene Expression Regulation, Neoplastic , Genetic Therapy , Heterografts , Humans , Mice , Mice, Nude , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/therapy , Signal Transduction , Transfection
7.
PLoS One ; 8(8): e71449, 2013.
Article in English | MEDLINE | ID: mdl-23951165

ABSTRACT

Angiogenesis is the result of the combined activity of the tumor microenvironment and signaling molecules. The angiogenic switch is represented as an imbalance between pro- and anti-angiogenic factors and is a rate-limiting step in the development of tumors. Eph receptor tyrosine kinases and their membrane-anchored ligands, known as ephrins, constitute the largest receptor tyrosine kinase (RTK) subfamily and are considered a major family of pro-angiogenic RTKs. Ewing sarcoma (EWS) is a highly aggressive bone and soft tissue tumor affecting children and young adults. As other solid tumors, EWS are reliant on a functional vascular network for the delivery of nutrients and oxygen and for the removal of waste. Based on the biological roles of EphA2 in promoting angiogenesis, we explored the functional role of this receptor and its relationship with caveolin-1 (CAV1) in EWS angiogenesis. We demonstrated that lack of CAV1 results in a significant reduction in micro vascular density (MVD) on 3 different in vivo models. In vitro, this phenomenon correlated with inactivation of EphA2 receptor, lack of AKT response and downregulation of bFGF. We also demonstrated that secreted bFGF from EWS cells acted as chemoattractant for endothelial cells. Furthermore, interaction between EphA2 and CAV1 was necessary for the right localization and signaling of the receptor to produce bFGF through AKT and promote migration of endothelial cells. Finally, introduction of a dominant-negative form of EphA2 into EWS cells mostly reproduced the effects occurred by CAV1 silencing, strongly suggesting that the axis EphA2-CAV1 participates in the promotion of endothelial cell migration toward the tumors favoring EWS angiogenesis.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Caveolin 1/metabolism , Fibroblast Growth Factor 2/biosynthesis , Neovascularization, Pathologic/metabolism , Receptor, EphA2/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Animals , Bone Neoplasms/genetics , Caveolin 1/genetics , Cell Line, Tumor , Cell Movement/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Female , Fibroblast Growth Factor 2/genetics , Gene Silencing , Heterografts , Humans , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphA2/genetics , Sarcoma, Ewing/genetics , Signal Transduction , Transcription, Genetic , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...