Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Cancer Ther ; 20(10): 1868-1879, 2021 10.
Article in English | MEDLINE | ID: mdl-34315769

ABSTRACT

The EWSR1-FLI1 t(11;22)(q24;q12) translocation is the hallmark genomic alteration of Ewing sarcoma, a malignancy of the bone and surrounding tissue, predominantly affecting children and adolescents. Although significant progress has been made for the treatment of localized disease, patients with metastasis or who relapse after chemotherapy have less than a 30% five-year survival rate. EWS-FLI1 is currently not clinically druggable, driving the need for more effective targeted therapies. Treatment with the H3K27 demethylase inhibitor, GSK-J4, leads to an increase in H3K27me and a decrease in H3K27ac, a significant event in Ewing sarcoma because H3K27ac associates strongly with EWS-FLI1 binding at enhancers and promoters and subsequent activity of EWS-FLI1 target genes. We were able to identify targets of EWS-FLI1 tumorigenesis directly inhibited by GSK-J4. GSK-J4 disruption of EWS-FLI1-driven transcription was toxic to Ewing sarcoma cells and slowed tumor growth in patient-derived xenografts (PDX) of Ewing sarcoma. Responses were markedly exacerbated by cotreatment with a disruptor of RNA polymerase II activity, the CDK7 inhibitor THZ1. This combination together suppressed EWS-FLI1 target genes and viability of ex vivo PDX Ewing sarcoma cells in a synergistic manner. In PDX models of Ewing Sarcoma, the combination shrank tumors. We present a new therapeutic strategy to treat Ewing sarcoma by decreasing H3K27ac at EWS-FLI1-driven transcripts, exacerbated by blocking phosphorylation of the C-terminal domain of RNA polymerase II to further hinder the EWS-FLI1-driven transcriptome.


Subject(s)
Benzazepines/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Histones/antagonists & inhibitors , Oncogene Proteins, Fusion/antagonists & inhibitors , Phenylenediamines/pharmacology , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Pyrimidines/pharmacology , RNA-Binding Protein EWS/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Transcriptome , Animals , Apoptosis , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Cancer Res ; 81(8): 2071-2085, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33526511

ABSTRACT

The failure of once promising target-specific therapeutic strategies often arises from redundancies in gene expression pathways. Even with new melanoma treatments, many patients are not responsive or develop resistance, leading to disease progression in terms of growth and metastasis. We previously discovered that the transcription factors ETS1 and PAX3 drive melanoma growth and metastasis by promoting the expression of the MET receptor. Here, we find that there are multiple ETS family members expressed in melanoma and that these factors have redundant functions. The small molecule YK-4-279, initially developed to target the ETS gene-containing translocation product EWS-FLI1, significantly inhibited cellular growth, invasion, and ETS factor function in melanoma cell lines and a clinically relevant transgenic mouse model, BrafCA;Tyr-CreERT2;Ptenf/f. One of the antitumor effects of YK-4-279 in melanoma is achieved via interference of multiple ETS family members with PAX3 and the expression of the PAX3-ETS downstream gene MET. Expression of exogenous MET provided partial rescue of the effects of YK-4-279, further supporting that MET loss is a significant contributor to the antitumor effects of the drug. This is the first study identifying multiple overlapping functions of the ETS family promoting melanoma. In addition, targeting all factors, rather than individual members, demonstrated impactful deleterious consequences in melanoma progression. Given that multiple ETS factors are known to have oncogenic functions in other malignancies, these findings have a high therapeutic impact. SIGNIFICANCE: These findings identify YK-4-279 as a promising therapeutic agent against melanoma by targeting multiple ETS family members and blocking their ability to act as transcription factors.


Subject(s)
Indoles/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Disease Progression , Humans , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Transgenic , Neoplasm Invasiveness , Oncogene Proteins, Fusion/antagonists & inhibitors , PAX3 Transcription Factor/antagonists & inhibitors , PAX3 Transcription Factor/metabolism , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
3.
Arterioscler Thromb Vasc Biol ; 41(3): 1191-1204, 2021 03.
Article in English | MEDLINE | ID: mdl-33406853

ABSTRACT

OBJECTIVE: Noncoding RNAs are emerging as important players in gene regulation and cardiovascular diseases. Their roles in the pathogenesis of atherosclerosis are not fully understood. The purpose of this study was to determine the role played by a previously uncharacterized long noncoding RNA, RP11-728F11.4, in the development of atherosclerosis and the mechanisms by which it acts. Approach and Results: Expression microarray analysis revealed that atherosclerotic plaques had increased expression of RP11-728F11.4 as well as the cognate gene FXYD6 (FXYD domain containing ion transport regulator 6), which encodes a modulator of Na+/K+-ATPase. In vitro experiments showed that RP11-728F11.4 interacted with the RNA-binding protein EWSR1 (Ewings sarcoma RNA binding protein-1) and upregulated FXYD6 expression. Lentivirus-induced overexpression of RP11-728F11.4 in cultured monocytes-derived macrophages resulted in higher Na+/K+-ATPase activity, intracellular cholesterol accumulation, and increased proinflammatory cytokine production. The effects of RP11-728F11.4 were enhanced by siRNA-mediated knockdown of EWSR1 and reduced by downregulation of FXYD domain containing ion transport regulator 6. In vivo experiments in apoE knockout mice fed a Western diet demonstrated that RP11-728F11.4 increased proinflammatory cytokine production and augmented atherosclerotic lesions. CONCLUSIONS: RP11-728F11.4 promotes atherosclerosis, with an influence on cholesterol homeostasis and proinflammatory molecule production, thus representing a potential therapeutic target. Graphic Abstract: A graphic abstract is available for this article.


Subject(s)
Atherosclerosis/genetics , RNA, Long Noncoding/genetics , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cells, Cultured , Cholesterol/metabolism , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Female , Gene Knockdown Techniques , Humans , Ion Channels/genetics , Ion Channels/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Middle Aged , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , RNA, Long Noncoding/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Up-Regulation
4.
Clin Cancer Res ; 27(4): 1184-1194, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33229458

ABSTRACT

PURPOSE: Desmoplastic small round cell tumor (DSRCT) is a highly lethal intra-abdominal sarcoma of adolescents and young adults. DSRCT harbors a t(11;22)(p13:q12) that generates the EWSR1-WT1 chimeric transcription factor, the key oncogenic driver of DSRCT. EWSR1-WT1 rewires global gene expression networks and activates aberrant expression of targets that together mediate oncogenesis. EWSR1-WT1 also activates a neural gene expression program. EXPERIMENTAL DESIGN: Among these neural markers, we found prominent expression of neurotrophic tyrosine kinase receptor 3 (NTRK3), a druggable receptor tyrosine kinase. We investigated the regulation of NTRK3 by EWSR1-WT1 and its potential as a therapeutic target in vitro and in vivo, the latter using novel patient-derived models of DSRCT. RESULTS: We found that EWSR1-WT1 binds upstream of NTRK3 and activates its transcription. NTRK3 mRNA is highly expressed in DSRCT compared with other major chimeric transcription factor-driven sarcomas and most DSRCTs are strongly immunoreactive for NTRK3 protein. Remarkably, expression of NTRK3 kinase domain mRNA in DSRCT is also higher than in cancers with NTRK3 fusions. Abrogation of NTRK3 expression by RNAi silencing reduces growth of DSRCT cells and pharmacologic targeting of NTRK3 with entrectinib is effective in both in vitro and in vivo models of DSRCT. CONCLUSIONS: Our results indicate that EWSR1-WT1 directly activates NTRK3 expression in DSRCT cells, which are dependent on its expression and activity for growth. Pharmacologic inhibition of NTRK3 by entrectinib significantly reduces growth of DSRCT cells both in vitro and in vivo, providing a rationale for clinical evaluation of NTRK3 as a therapeutic target in DSRCT.


Subject(s)
Benzamides/therapeutic use , Desmoplastic Small Round Cell Tumor/drug therapy , Indazoles/therapeutic use , Oncogene Proteins, Fusion/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , Adolescent , Adult , Animals , Benzamides/pharmacology , Cell Line, Tumor , Child , Desmoplastic Small Round Cell Tumor/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Indazoles/pharmacology , Male , Mice , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein EWS/genetics , Receptor, trkC/genetics , Receptor, trkC/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism , Xenograft Model Antitumor Assays , Young Adult
5.
PLoS One ; 15(9): e0238999, 2020.
Article in English | MEDLINE | ID: mdl-32915889

ABSTRACT

Aberrant expression of the transcription factor ERG is a key driving event in approximately one-half of all of prostate cancers. Lacking an enzymatic pocket and mainly disordered, the structure of ERG is difficult to exploit for therapeutic design. We recently identified EWS as a specific interacting partner of ERG that is required for oncogenic function. In this study, we aimed to target this specific protein-protein interaction with small molecules. A high-throughput screening (HTS) strategy was implemented to identify potential protein-protein interaction inhibitors. Secondary assays verified the function of several hit compounds, and one lead compound inhibited ERG-mediated phenotypes in prostate cells. This is the first study aimed at targeting the ERG-EWS protein-protein interaction for the development of a small molecule-based prostate cancer therapy.


Subject(s)
High-Throughput Screening Assays/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Feasibility Studies , Humans , Male , Prostatic Neoplasms/genetics , Protein Interaction Domains and Motifs/drug effects , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Small Molecule Libraries , Transcriptional Regulator ERG/antagonists & inhibitors , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
6.
Mol Med Rep ; 21(3): 1667-1675, 2020 03.
Article in English | MEDLINE | ID: mdl-32016454

ABSTRACT

Despite Ewing sarcoma (ES) being the second most common pediatric malignancy of bone and soft tissue, few novel therapeutic approaches have been introduced over the past few decades. ES contains a pathognomonic chromosomal translocation that leads to a fusion protein between EWSR1 and an ets family member, most often FLI1. EWS­FLI1 is the most common type of fusion protein and is a well­vetted therapeutic target. A small molecule inhibitor of EWS­FLI1, YK­4­279 (YK) was developed with the intention to serve as a targeted therapy option for patients with ES. The present study investigated resistance mechanisms by developing an ES cell line specifically resistant to YK. The ES cell line A4573 was treated with YK to create resistant cells by long term continuous exposure. The results revealed that resistance in A4573 was robust and sustainable, with a >27­fold increase in IC50 lasting up to 16 weeks in the absence of the compound. Resistant ES cells were still sensitive to standard of care drugs, including doxorubicin, vincristine and etoposide, which may be valuable in future combination treatments in the clinic. Resistant ES cells revealed an increased expression of CD99. RNA sequencing and qPCR validation of resistant ES cells confirmed an increased expression of ANO1, BRSK2 and IGSF21, and a reduced expression of COL24A1, PRSS23 and RAB38 genes. A functional association between these genes and mechanism of resistance remains to be investigated. The present study created a cell line to investigate YK resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Indoles/pharmacology , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , 12E7 Antigen/genetics , 12E7 Antigen/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression , Humans
7.
Int J Mol Sci ; 20(15)2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31370265

ABSTRACT

Osteosarcoma and Ewing sarcoma are the most common malignant primary bone tumors mainly occurring in children, adolescents and young adults. Current standard therapy includes multidrug chemotherapy and/or radiation specifically for Ewing sarcoma, associated with tumor resection. However, patient survival has not evolved for the past decade and remains closely related to the response of tumor cells to chemotherapy, reaching around 75% at 5 years for patients with localized forms of osteosarcoma or Ewing sarcoma but less than 30% in metastatic diseases and patients resistant to initial chemotherapy. Despite Ewing sarcoma being characterized by specific EWSR1-ETS gene fusions resulting in oncogenic transcription factors, currently, no targeted therapy could be implemented. It seems even more difficult to develop a targeted therapeutic strategy in osteosarcoma which is characterized by high complexity and heterogeneity in genomic alterations. Nevertheless, the common point between these different bone tumors is their ability to deregulate bone homeostasis and remodeling and divert them to their benefit. Therefore, targeting different actors of the bone tumor microenvironment has been hypothesized to develop new therapeutic strategies. In this context, it is well known that the Wnt/ß-catenin signaling pathway plays a key role in cancer development, including osteosarcoma and Ewing sarcoma as well as in bone remodeling. Moreover, recent studies highlight the implication of the Wnt/ß-catenin pathway in angiogenesis and immuno-surveillance, two key mechanisms involved in metastatic dissemination. This review focuses on the role played by this signaling pathway in the development of primary bone tumors and the modulation of their specific microenvironment.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Osteosarcoma/drug therapy , Sarcoma, Ewing/drug therapy , Tumor Microenvironment/drug effects , Adolescent , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Bone and Bones , Child , Humans , Lymphatic Metastasis , Molecular Targeted Therapy/methods , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/mortality , Neovascularization, Pathologic/prevention & control , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/immunology , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/mortality , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/immunology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/immunology , Sarcoma, Ewing/mortality , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Wnt Signaling Pathway/drug effects , Young Adult , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/immunology
8.
Clin Cancer Res ; 25(11): 3417-3429, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30723142

ABSTRACT

PURPOSE: The successful clinical translation of compounds that target specific oncogenic transcription factors will require an understanding of the mechanism of target suppression to optimize the dose and schedule of administration. We have previously shown trabectedin reverses the gene signature of the EWS-FLI1 transcription factor. In this report, we establish the mechanism of suppression and use it to justify the reevaluation of this drug in the clinic in patients with Ewing sarcoma.Experimental Design: We demonstrate a novel epigenetic mechanism of trabectedin using biochemical fractionation and chromatin immunoprecipitation sequencing. We link the effect to drug schedule and EWS-FLI1 downstream target expression using confocal microscopy, qPCR, Western blot analysis, and cell viability assays. Finally, we quantitate target suppression within the three-dimensional architecture of the tumor in vivo using 18F-FLT imaging. RESULTS: Trabectedin evicts the SWI/SNF chromatin-remodeling complex from chromatin and redistributes EWS-FLI1 in the nucleus leading to a marked increase in H3K27me3 and H3K9me3 at EWS-FLI1 target genes. These effects only occur at high concentrations of trabectedin leading to suppression of EWS-FLI1 target genes and a loss of cell viability. In vivo, low-dose irinotecan is required to improve the magnitude, penetrance, and duration of target suppression in the three-dimensional architecture of the tumor leading to differentiation of the Ewing sarcoma xenograft into benign mesenchymal tissue. CONCLUSIONS: These data provide the justification to evaluate trabectedin in the clinic on a short infusion schedule in combination with low-dose irinotecan with 18F-FLT PET imaging in patients with Ewing sarcoma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Chromatin/genetics , Gene Expression Regulation, Neoplastic/drug effects , Oncogene Proteins, Fusion/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Trabectedin/pharmacology , Transcription Factors/genetics , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Oncogene Proteins, Fusion/blood , Oncogene Proteins, Fusion/genetics , Protein Binding , Proto-Oncogene Protein c-fli-1/blood , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/blood , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Xenograft Model Antitumor Assays
9.
J Med Chem ; 61(17): 8001-8016, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30114371

ABSTRACT

Mithramycin A (1) was identified as the top potential inhibitor of the aberrant ETS transcription factor EWS-FLI1, which causes Ewing sarcoma. Unfortunately, 1 has a narrow therapeutic window, compelling us to seek less toxic and more selective analogues. Here, we used MTMSA (2) to generate analogues via peptide coupling and fragment-based drug development strategies. Cytotoxicity assays in ETS and non-ETS dependent cell lines identified two dipeptide analogues, 60 and 61, with 19.1- and 15.6-fold selectivity, respectively, compared to 1.5-fold for 1. Importantly, the cytotoxicity of 60 and 61 is <100 nM in ETS cells. Molecular assays demonstrated the inhibitory capacity of these analogues against EWS-FLI1 mediated transcription in Ewing sarcoma. Structural analysis shows that positioning the tryptophan residue in a distal position improves selectivity, presumably via interaction with the ETS transcription factor. Thus, these analogues may present new ways to target transcription factors for clinical use.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Drug Development , Oncogene Proteins, Fusion/antagonists & inhibitors , Plicamycin/analogs & derivatives , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Antibiotics, Antineoplastic/chemistry , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Molecular Structure , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Tumor Cells, Cultured
10.
Mol Med Rep ; 17(3): 4291-4298, 2018 03.
Article in English | MEDLINE | ID: mdl-29328472

ABSTRACT

Ciclopirox (CPX) is a synthetic antifungal drug that is mainly used to treat dermatomycoses. The aim of the present study was to determine whether CPX could influence Ewing sarcoma progression. The present study suggested that CPX treatment may inhibit Ewing sarcoma (ES) progression through Ewing sarcoma breakpoint region 1­Friend leukemia integration 1 (EWS­FLI1), a common fusion transcript structure in patients with ES. To determine the underlying mechanisms of ES progression, cross analysis was conducted on three high­throughput genome or transcript me datasets from the Gene Expression Omnibus. The results indicated that CPX may inhibit ES growth by affecting vasculature development and DNA replication. A combination of genome­wide expression and binding profiles revealed several potential targets for CPX in ES, including collagen type I α2 chain, N­myc proto­oncogene and transforming growth factor ß1, which contained significantly enriched binding peaks of FLI1. In addition, network analysis, including a protein­protein interaction network and a transcription regulatory network, provided further detailed information about the roles of CPX in ES. This study may provide a novel solution for ES treatment and may also aid in improving its prognosis.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Genome, Human , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , Pyridones/therapeutic use , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/drug therapy , Antifungal Agents/therapeutic use , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Ciclopirox , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Databases, Genetic , Drug Repositioning , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Humans , Molecular Sequence Annotation , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/metabolism , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
11.
Sci Signal ; 10(499)2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28974650

ABSTRACT

Ewing's sarcoma (ES) is a rare and highly malignant cancer that grows in the bones or surrounding tissues mostly affecting adolescents and young adults. A chimeric fusion between the RNA binding protein EWS and the ETS family transcription factor FLI1 (EWS-FLI1), which is generated from a chromosomal translocation, is implicated in driving most ES cases by modulation of transcription and alternative splicing. The small-molecule YK-4-279 inhibits EWS-FLI1 function and induces apoptosis in ES cells. We aimed to identify both the underlying mechanism of the drug and potential combination therapies that might enhance its antitumor activity. We tested 69 anticancer drugs in combination with YK-4-279 and found that vinca alkaloids exhibited synergy with YK-4-279 in five ES cell lines. The combination of YK-4-279 and vincristine reduced tumor burden and increased survival in mice bearing ES xenografts. We determined that independent drug-induced events converged to cause this synergistic therapeutic effect. YK-4-279 rapidly induced G2-M arrest, increased the abundance of cyclin B1, and decreased EWS-FLI1-mediated generation of microtubule-associated proteins, which rendered cells more susceptible to microtubule depolymerization by vincristine. YK-4-279 reduced the expression of the EWS-FLI1 target gene encoding the ubiquitin ligase UBE2C, which, in part, contributed to the increase in cyclin B1. YK-4-279 also increased the abundance of proapoptotic isoforms of MCL1 and BCL2, presumably through inhibition of alternative splicing by EWS-FLI1, thus promoting cell death in response to vincristine. Thus, a combination of vincristine and YK-4-279 might be therapeutically effective in ES patients.


Subject(s)
Drug Resistance, Neoplasm/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Indoles/pharmacology , M Phase Cell Cycle Checkpoints/drug effects , Oncogene Proteins, Fusion/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Vincristine/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cyclin B1/genetics , Cyclin B1/metabolism , Drug Resistance, Neoplasm/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Humans , M Phase Cell Cycle Checkpoints/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
13.
Mol Cancer Ther ; 16(1): 88-101, 2017 01.
Article in English | MEDLINE | ID: mdl-28062706

ABSTRACT

Improvements in survival for Ewing sarcoma pediatric and adolescent patients have been modest over the past 20 years. Combinations of anticancer agents endure as an option to overcome resistance to single treatments caused by compensatory pathways. Moreover, combinations are thought to lessen any associated adverse side effects through reduced dosing, which is particularly important in childhood tumors. Using a parallel phenotypic combinatorial screening approach of cells derived from three pediatric tumor types, we identified Ewing sarcoma-specific interactions of a diverse set of targeted agents including approved drugs. We were able to retrieve highly synergistic drug combinations specific for Ewing sarcoma and identified signaling processes important for Ewing sarcoma cell proliferation determined by EWS-FLI1 We generated a molecular target profile of PKC412, a multikinase inhibitor with strong synergistic propensity in Ewing sarcoma, revealing its targets in critical Ewing sarcoma signaling routes. Using a multilevel experimental approach including quantitative phosphoproteomics, we analyzed the molecular rationale behind the disease-specific synergistic effect of simultaneous application of PKC412 and IGF1R inhibitors. The mechanism of the drug synergy between these inhibitors is different from the sum of the mechanisms of the single agents. The combination effectively inhibited pathway crosstalk and averted feedback loop repression, in EWS-FLI1-dependent manner. Mol Cancer Ther; 16(1); 88-101. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Drug Screening Assays, Antitumor , Molecular Targeted Therapy , Animals , Antigens, CD , Cell Line, Tumor , Computational Biology/methods , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Interactions , Humans , Oncogene Proteins, Fusion/antagonists & inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Receptor, IGF Type 1 , Receptor, Insulin/antagonists & inhibitors , Receptors, Somatomedin/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Signal Transduction/drug effects , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Xenograft Model Antitumor Assays
14.
Oncotarget ; 7(28): 43504-43517, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27259270

ABSTRACT

Ewing sarcoma is driven by characteristic chromosomal translocations between the EWSR1 gene with genes encoding ETS family transcription factors (EWS-ETS), most commonly FLI1. However, direct pharmacological inhibition of transcription factors like EWS-FLI1 remains largely unsuccessful. Active gene transcription requires orchestrated actions of many epigenetic regulators, such as the bromodomain and extra-terminal domain (BET) family proteins. Emerging BET bromodomain inhibitors have exhibited promising antineoplastic activities via suppression of oncogenic transcription factors in various cancers. We reasoned that EWS-FLI1-mediated transcription activation might be susceptible to BET inhibition. In this study, we demonstrated that small molecule BET bromodomain inhibitors repressed EWS-FLI1-driven gene signatures and downregulated important target genes. However, expression of EWS-FLI1 was not significantly affected. Repression of autocrine IGF1 by BET inhibitors led to significant inhibition of the IGF1R/AKT pathway critical to Ewing sarcoma cell proliferation and survival. Consistently, BET inhibitors impaired viability and clonogenic survival of Ewing sarcoma cell lines and blocked EWS-FLI1-induced transformation of mouse NIH3T3 fibroblast cells. Selective depletion of individual BET genes partially phenocopied the actions of BET inhibitors. Finally, the prototypical BET inhibitor, JQ1, significantly repressed Ewing sarcoma xenograft tumor growth. These findings suggest therapeutic potential of BET inhibitors in Ewing sarcoma and highlight an emerging paradigm of using epigenetic agents to treat cancers driven by fusion transcription factors.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Bone Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Insulin-Like Growth Factor I/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Proteins/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Triazoles/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Autocrine Communication/drug effects , Azepines/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Epigenesis, Genetic/drug effects , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Proteins/genetics , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Protein EWS/genetics , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Triazoles/therapeutic use , Xenograft Model Antitumor Assays
15.
Oncotarget ; 7(17): 24125-40, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27006472

ABSTRACT

Ewing Sarcoma is a rare bone and soft tissue malignancy affecting children and young adults. Chromosomal translocations in this cancer produce fusion oncogenes as characteristic molecular signatures of the disease. The most common case is the translocation t (11; 22) (q24;q12) which yields the EWS-Fli1 chimeric transcription factor. Finding a way to directly target EWS-Fli1 remains a central therapeutic approach to eradicate this aggressive cancer. Here we demonstrate that treating Ewing Sarcoma cells with JQ1(+), a BET bromodomain inhibitor, represses directly EWS-Fli1 transcription as well as its transcriptional program. Moreover, the Chromatin Immuno Precipitation experiments demonstrate for the first time that these results are a consequence of the depletion of BRD4, one of the BET bromodomains protein from the EWS-Fli1 promoter. In vitro, JQ1(+) treatment reduces the cell viability, impairs the cell clonogenic and the migratory abilities, and induces a G1-phase blockage as well as a time- and a dose-dependent apoptosis. Furthermore, in our in vivo model, we observed a tumor burden delay, an inhibition of the global vascularization and an increase of the mice overall survival. Taken together, our data indicate that inhibiting the BET bromodomains interferes with EWS-FLi1 transcription and could be a promising strategy in the Ewing tumors context.


Subject(s)
Azepines/pharmacology , Bone Neoplasms/pathology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Oncogene Proteins, Fusion/antagonists & inhibitors , Proteins/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Sarcoma, Ewing/pathology , Triazoles/pharmacology , Adolescent , Adult , Animals , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Male , Mice , Mice, Nude , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult
16.
Clin Cancer Res ; 22(16): 4105-18, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-26979396

ABSTRACT

PURPOSE: The goal of this study was to identify second-generation mithramycin analogues that better target the EWS-FLI1 transcription factor for Ewing sarcoma. We previously established mithramycin as an EWS-FLI1 inhibitor, but the compound's toxicity prevented its use at effective concentrations in patients. EXPERIMENTAL DESIGN: We screened a panel of mithralogs to establish their ability to inhibit EWS-FLI1 in Ewing sarcoma. We compared the IC50 with the MTD established in mice to determine the relationship between efficacy and toxicity. We confirmed the suppression of EWS-FLI1 at the promoter, mRNA, gene signature, and protein levels. We established an improved therapeutic window by using time-lapse microscopy to model the effects on cellular proliferation in Ewing sarcoma cells relative to HepG2 control cells. Finally, we established an improved therapeutic window using a xenograft model of Ewing sarcoma. RESULTS: EC-8105 was found to be the most potent analogue and was able to suppress EWS-FLI1 activity at concentrations nontoxic to other cell types. EC-8042 was substantially less toxic than mithramycin in multiple species but maintained suppression of EWS-FLI1 at similar concentrations. Both compounds markedly suppressed Ewing sarcoma xenograft growth and inhibited EWS-FLI1 in vivo CONCLUSIONS: These results provide a basis for the continued development of EC-8042 and EC-8105 as EWS-FLI1 inhibitors for the clinic. Clin Cancer Res; 22(16); 4105-18. ©2016 AACR.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/metabolism , Plicamycin/pharmacology , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Molecular Targeted Therapy , Promoter Regions, Genetic , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/mortality , Transcription Factors , Xenograft Model Antitumor Assays
17.
Oncotarget ; 7(14): 17616-30, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26848860

ABSTRACT

Ewing sarcoma is an aggressive primary pediatric bone tumor, often diagnosed in adolescents and young adults. A pathognomonic reciprocal chromosomal translocation results in a fusion gene coding for a protein which derives its N-terminus from a FUS/EWS/TAF15 (FET) protein family member, commonly EWS, and C-terminus containing the DNA-binding domain of an ETS transcription factor, commonly FLI1. Nearly 85% of cases express the EWS-FLI protein which functions as a transcription factor and drives oncogenesis. As the primary genomic lesion and a protein which is not expressed in normal cells, disrupting EWS-FLI function is an attractive therapeutic strategy for Ewing sarcoma. However, transcription factors are notoriously difficult targets for the development of small molecules. Improved understanding of the oncogenic mechanisms employed by EWS-FLI to hijack normal cellular programming has uncovered potential novel approaches to pharmacologically block EWS-FLI function. In this review we examine targeting the chromatin regulatory enzymes recruited to conspire in oncogenesis with a focus on the histone lysine specific demethylase 1 (LSD1). LSD1 inhibitors are being aggressively investigated in acute myeloid leukemia and the results of early clinical trials will help inform the future use of LSD1 inhibitors in sarcoma. High LSD1 expression is observed in Ewing sarcoma patient samples and mechanistic and preclinical data suggest LSD1 inhibition globally disrupts the function of EWS-ETS proteins.


Subject(s)
Bone Neoplasms/drug therapy , Histone Demethylases/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Soft Tissue Neoplasms/drug therapy , Adolescent , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Child , Humans , Molecular Targeted Therapy , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology
18.
Cell Rep ; 14(3): 598-610, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26776507

ABSTRACT

Ewing sarcoma cells depend on the EWS-FLI1 fusion transcription factor for cell survival. Using an assay of EWS-FLI1 activity and genome-wide RNAi screening, we have identified proteins required for the processing of the EWS-FLI1 pre-mRNA. We show that Ewing sarcoma cells harboring a genomic breakpoint that retains exon 8 of EWSR1 require the RNA-binding protein HNRNPH1 to express in-frame EWS-FLI1. We also demonstrate the sensitivity of EWS-FLI1 fusion transcripts to the loss of function of the U2 snRNP component, SF3B1. Disrupted splicing of the EWS-FLI1 transcript alters EWS-FLI1 protein expression and EWS-FLI1-driven expression. Our results show that the processing of the EWS-FLI1 fusion RNA is a potentially targetable vulnerability in Ewing sarcoma cells.


Subject(s)
Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Base Sequence , Binding Sites , Calmodulin-Binding Proteins/antagonists & inhibitors , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Cell Line, Tumor , Cell Survival , Exons , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Humans , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/genetics , RNA Interference , RNA Precursors/metabolism , RNA Splicing , RNA Splicing Factors , RNA, Small Interfering/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/antagonists & inhibitors , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Sarcoma, Ewing/pathology , Trans-Activators , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Oncotarget ; 6(35): 37678-94, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26462019

ABSTRACT

Ewing sarcoma is an aggressive tumor of bone and soft tissue affecting predominantly children and young adults. Tumor-specific chromosomal translocations create EWS-FLI1 and similar aberrant ETS fusion proteins that drive sarcoma development in patients. ETS family fusion proteins and over-expressed ETS proteins are also found in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients. Transgenic expression of EWS-FLI1 in mice promotes high penetrance erythroid leukemia with dense hepatic and splenic infiltrations. We identified a small molecule, YK-4-279, that directly binds to EWS-FLI1 and inhibits its oncogenic activity in Ewing sarcoma cell lines and xenograft mouse models. Herein, we tested in vivo therapeutic efficacy and potential side effects of YK-4-279 in the transgenic mouse model with EWS-FLI1 induced leukemia. A two-week course of treatment with YK-4-279 significantly reduced white blood cell count, nucleated erythroblasts in the peripheral blood, splenomegaly, and hepatomegaly of erythroleukemic mice. YK-4-279 inhibited EWS-FLI1 target gene expression in neoplastic cells. Treated animals showed significantly better overall survival compared to control mice that rapidly succumbed to leukemia. YK-4-279 treated mice did not show overt toxicity in liver, spleen, or bone marrow. In conclusion, this in vivo study highlights the efficacy of YK-4-279 to treat EWS-FLI1 expressing neoplasms and support its therapeutic potential for patients with Ewing sarcoma and other ETS-driven malignancies.


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Indoles/pharmacology , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/etiology , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/toxicity , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/toxicity , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/toxicity , Animals , Blotting, Western , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Immunoenzyme Techniques , Leukemia, Erythroblastic, Acute/pathology , Mice , Mice, Transgenic , Oncogene Proteins, Fusion/administration & dosage , Proto-Oncogene Protein c-fli-1/administration & dosage , RNA, Messenger/genetics , RNA-Binding Protein EWS/administration & dosage , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance
20.
Proc Natl Acad Sci U S A ; 112(19): 6074-9, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25918410

ABSTRACT

EWS (Ewing sarcoma) encodes an RNA/ssDNA binding protein that is frequently rearranged in a number of different cancers by chromosomal translocations. Physiologically, EWS has diverse and essential roles in various organ development and cellular processes. In this study, we uncovered a new role of EWS in mitochondrial homeostasis and energy metabolism. Loss of EWS leads to a significant decrease in mitochondria abundance and activity, which is caused by a rapid degradation of Peroxisome proliferator-activated receptor γ Coactivator (PGC-1α), a central regulator of mitochondria biogenesis, function, and cellular energy metabolism. EWS inactivation leads to increased ubiquitination and proteolysis of PGC-1α via proteasome pathway. Complementation of EWS in Ews-deficient cells restores PGC-1α and mitochondrial abundance. We found that expression of E3 ubiquitin ligase, FBXW7 (F-box/WD40 domain protein 7), is increased in the absence of Ews and depletion of Fbxw7 in Ews-null cells restores PGC-1α expression and mitochondrial density. Consistent with these findings, mitochondrial abundance and activity are significantly reduced in brown fat and skeletal muscles of Ews-deficient mice. Furthermore, expression of mitochondrial biogenesis, respiration and fatty acid ß-oxidation genes is significantly reduced in the liver of Ews-null mice. These results demonstrate a novel role of EWS in mitochondrial and cellular energy homeostasis by controlling PGC-1α protein stability, and further implicate altered mitochondrial and energy metabolism in cancers harboring the EWS translocation.


Subject(s)
Mitochondria/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , Transcription Factors/metabolism , Adipose Tissue, Brown/metabolism , Animals , DNA, Mitochondrial/metabolism , Energy Metabolism , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Fatty Acids/chemistry , Fatty Acids/metabolism , Gene Expression Profiling , HEK293 Cells , Homeostasis , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Muscle, Skeletal/metabolism , Oxygen/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Conformation , RNA-Binding Protein EWS/metabolism , Ubiquitin/chemistry , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...