Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Front Oncol ; 13: 1099550, 2023.
Article in English | MEDLINE | ID: mdl-36793594

ABSTRACT

Ewing sarcoma is a cancer of children and young adults characterized by the critical translocation-associated fusion oncoprotein EWSR1::FLI1. EWSR1::FLI1 targets characteristic genetic loci where it mediates aberrant chromatin and the establishment of de novo enhancers. Ewing sarcoma thus provides a model to interrogate mechanisms underlying chromatin dysregulation in tumorigenesis. Previously, we developed a high-throughput chromatin-based screening platform based on the de novo enhancers and demonstrated its utility in identifying small molecules capable of altering chromatin accessibility. Here, we report the identification of MS0621, a molecule with previously uncharacterized mechanism of action, as a small molecule modulator of chromatin state at sites of aberrant chromatin accessibility at EWSR1::FLI1-bound loci. MS0621 suppresses cellular proliferation of Ewing sarcoma cell lines by cell cycle arrest. Proteomic studies demonstrate that MS0621 associates with EWSR1::FLI1, RNA binding and splicing proteins, as well as chromatin regulatory proteins. Surprisingly, interactions with chromatin and many RNA-binding proteins, including EWSR1::FLI1 and its known interactors, were RNA-independent. Our findings suggest that MS0621 affects EWSR1::FLI1-mediated chromatin activity by interacting with and altering the activity of RNA splicing machinery and chromatin modulating factors. Genetic modulation of these proteins similarly inhibits proliferation and alters chromatin in Ewing sarcoma cells. The use of an oncogene-associated chromatin signature as a target allows for a direct approach to screen for unrecognized modulators of epigenetic machinery and provides a framework for using chromatin-based assays for future therapeutic discovery efforts.

2.
J Biol Chem ; 293(35): 13592-13603, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29986887

ABSTRACT

Chromatin remodelers use bromodomains (BDs) to recognize histones. Polybromo 1 (PBRM1 or BAF180) is hypothesized to function as the nucleosome-recognition subunit of the PBAF chromatin-remodeling complex and is frequently mutated in clear cell renal cell carcinoma (ccRCC). Previous studies have applied in vitro methods to explore the binding specificities of the six individual PBRM1 BDs. However, BD targeting to histones and the influence of neighboring BD on nucleosome recognition have not been well characterized. Here, using histone microarrays and intact nucleosomes to investigate the histone-binding characteristics of the six PBRM1 BDs individually and combined, we demonstrate that BD2 and BD4 of PBRM1 mediate binding to acetylated histone peptides and to modified recombinant and cellular nucleosomes. Moreover, we show that neighboring BDs variably modulate these chromatin interactions, with BD1 and BD5 enhancing nucleosome interactions of BD2 and BD4, respectively, whereas BD3 attenuated these interactions. We also found that binding pocket missense mutations in BD4 observed in ccRCC disrupt PBRM1-chromatin interactions and that these mutations in BD4, but not similar mutations in BD2, in the context of full-length PBRM1, accelerate ccRCC cell proliferation. Taken together, our biochemical and mutational analyses have identified BD4 as being critically important for maintaining proper PBRM1 function and demonstrate that BD4 mutations increase ccRCC cell growth. Because of the link between PBRM1 status and sensitivity to immune checkpoint inhibitor treatment, these data also suggest the relevance of BD4 as a potential clinical target.


Subject(s)
Carcinoma, Renal Cell/metabolism , Chromatin/metabolism , Histones/metabolism , Kidney Neoplasms/metabolism , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Carcinoma, Renal Cell/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins , Histones/chemistry , Humans , Kidney Neoplasms/genetics , Models, Molecular , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Protein Domains , Sequence Alignment , Transcription Factors/chemistry , Transcription Factors/genetics
3.
Proc Natl Acad Sci U S A ; 113(11): 3018-23, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929321

ABSTRACT

Mutations in chromatin-modifying proteins and transcription factors are commonly associated with a wide variety of cancers. Through gain- or loss-of-function, these mutations may result in characteristic alterations of accessible chromatin, indicative of shifts in the landscape of regulatory elements genome-wide. The identification of compounds that reverse a specific chromatin signature could lead to chemical probes or potential therapies. To explore whether chromatin accessibility could serve as a platform for small molecule screening, we adapted formaldehyde-assisted isolation of regulatory elements (FAIRE), a chemical method to enrich for nucleosome-depleted genomic regions, as a high-throughput, automated assay. After demonstrating the validity and robustness of this approach, we applied this method to screen an epigenetically targeted small molecule library by evaluating regions of aberrant nucleosome depletion mediated by EWSR1-FLI1, the chimeric transcription factor critical for the bone and soft tissue tumor Ewing sarcoma. As a class, histone deacetylase inhibitors were greatly overrepresented among active compounds. These compounds resulted in diminished accessibility at targeted sites by disrupting transcription of EWSR1-FLI1. Capitalizing on precise differences in chromatin accessibility for drug discovery efforts offers significant advantages because it does not depend on the a priori selection of a single molecular target and may detect novel biologically relevant pathways.


Subject(s)
Chromatin/drug effects , High-Throughput Screening Assays/methods , Oncogene Proteins, Fusion/antagonists & inhibitors , Transcription, Genetic/drug effects , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor , Chromatin/ultrastructure , Drug Design , Drug Evaluation, Preclinical , Histone Deacetylase Inhibitors/isolation & purification , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Molecular Targeted Therapy , Nucleosomes/ultrastructure , Oncogene Proteins, Fusion/genetics , Panobinostat , Phenylbutyrates/pharmacology , Sarcoma, Ewing/pathology , Small Molecule Libraries , Vorinostat
4.
Mol Cancer Res ; 12(11): 1610-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24994750

ABSTRACT

UNLABELLED: Recent evidence implicates the insulin-like growth factor (IGF) pathway in development of Ewing sarcoma, a highly malignant bone and soft-tissue tumor that primarily affects children and young adults. Despite promising results from preclinical studies of therapies that target this pathway, early-phase clinical trials have shown that a significant fraction of patients do not benefit, suggesting that cellular factors determine tumor sensitivity. Using FAIRE-seq, a chromosomal deletion of the PTEN locus in a Ewing sarcoma cell line was identified. In primary tumors, PTEN deficiency was observed in a large subset of cases, although not mediated by large chromosomal deletions. PTEN loss resulted in hyperactivation of the AKT signaling pathway. PTEN rescue led to decreased proliferation, inhibition of colony formation, and increased apoptosis. Strikingly, PTEN loss decreased sensitivity to IGF1R inhibitors but increased responsiveness to temsirolimus, a potent mTOR inhibitor, as marked by induction of autophagy. These results suggest that PTEN is lost in a significant fraction of primary tumors, and this deficiency may have therapeutic consequences by concurrently attenuating responsiveness to IGF1R inhibition while increasing activity of mTOR inhibitors. The identification of PTEN status in the tumors of patients with recurrent disease could help guide the selection of therapies. IMPLICATIONS: PTEN status in Ewing sarcoma affects cellular responses to IGFI and mTOR-directed therapy, thus justifying its consideration as a biomarker in future clinical trials.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , PTEN Phosphohydrolase/deficiency , TOR Serine-Threonine Kinases/antagonists & inhibitors , Autophagy/drug effects , Cell Line, Tumor , Gene Deletion , Human Umbilical Vein Endothelial Cells , Humans , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Sarcoma, Ewing/enzymology , Sarcoma, Ewing/pathology , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
5.
Genome Res ; 22(2): 259-70, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086061

ABSTRACT

Chromosomal translocations involving transcription factor genes have been identified in an increasingly wide range of cancers. Some translocations can create a protein "chimera" that is composed of parts from different proteins. How such chimeras cause cancer, and why they cause cancer in some cell types but not others, is not understood. One such chimera is EWS-FLI, the most frequently occurring translocation in Ewing Sarcoma, a malignant bone and soft tissue tumor of children and young adults. Using EWS-FLI and its parental transcription factor, FLI1, we created a unique experimental system to address questions regarding the genomic mechanisms by which chimeric transcription factors cause cancer. We found that in tumor cells, EWS-FLI targets regions of the genome distinct from FLI1, despite identical DNA-binding domains. In primary endothelial cells, however, EWS-FLI and FLI1 demonstrate similar targeting. To understand this mistargeting, we examined chromatin organization. Regions targeted by EWS-FLI are normally repressed and nucleosomal in primary endothelial cells. In tumor cells, however, bound regions are nucleosome depleted and harbor the chromatin signature of enhancers. We next demonstrated that through chimerism, EWS-FLI acquired the ability to alter chromatin. Expression of EWS-FLI results in nucleosome depletion at targeted sites, whereas silencing of EWS-FLI in tumor cells restored nucleosome occupancy. Thus, the EWS-FLI chimera acquired chromatin-altering activity, leading to mistargeting, chromatin disruption, and ultimately, transcriptional dysregulation.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Transcription, Genetic , Binding Sites/genetics , Cell Line, Tumor , Chimerism , Chromatin/metabolism , Endothelial Cells/metabolism , Enhancer Elements, Genetic , Gene Silencing , Humans , Microfilament Proteins/metabolism , Microsatellite Repeats , Neoplasms/genetics , Neoplasms/metabolism , Nucleotide Motifs , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Trans-Activators , Translocation, Genetic
6.
Cancer Res ; 70(2): 639-45, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068147

ABSTRACT

Clear cell sarcoma (CCS), a childhood tumor of the tendons and aponeuroses, is uniformly fatal once it has metastasized because of its profound therapeutic resistance. CCS is characterized by production of a chimeric transcription factor, EWS-ATF1, which is formed as the result of a disease-specific chromosomal translocation. EWS-ATF1 activates the melanocyte transcription factor MITF, which in turn activates transcription of c-Met, an oncogenic receptor tyrosine kinase recently shown to be activated in CCS. Based on this connection, we hypothesized that c-Met inhibition may offer a strategy to treat CCS, as an indirect tactic to defeat a transforming pathway downstream of EWS-ATF1. Here, we show that primary CCS and CCS-derived cell lines express c-Met, which is activated in an autocrine fashion by its ligand hepatocyte growth factor (HGF)/scatter factor in some CCS cell lines. c-Met expression is critical for CCS invasion, chemotaxis, and survival. Blocking c-Met activity with a small-molecule inhibitor (SU11274) or a neutralizing antibody to its ligand HGF (AMG 102) significantly reduced CCS cell growth in culture. Similarly, AMG 102 significantly suppressed in vivo tumor growth in an autocrine xenograft model of CCS. Collectively, these findings suggest the HGF:c-Met signaling axis as a candidate therapeutic target to improve clinical management of CCS.


Subject(s)
Antibodies, Monoclonal/pharmacology , Hepatocyte Growth Factor/metabolism , Indoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Sarcoma, Clear Cell/drug therapy , Sarcoma, Clear Cell/metabolism , Sulfonamides/pharmacology , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/immunology , Humans , Male , Mice , Mice, Nude , Oncogene Proteins, Fusion/biosynthesis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/biosynthesis , Signal Transduction , Transcription Factors/biosynthesis , Xenograft Model Antitumor Assays
7.
Curr Top Dev Biol ; 80: 311-35, 2008.
Article in English | MEDLINE | ID: mdl-17950378

ABSTRACT

Proper platelet function is essential for hemostasis. However, understanding platelet function is complicated by the fact that platelets are anucleate and therefore not amenable to direct genetic manipulations. To study platelet function, several laboratories have developed CHO cell lines expressing platelet proteins or used megakaryocyte-like cell lines. However, these cell culture models are unable to mimic critical platelet functions, most notably agonist-induced activation of integrin alphaIIbbeta3. Mature megakaryocytes, which are platelet precursors, express platelet-specific proteins, and the function of such proteins and signaling pathways appears conserved between the two cell types. Murine megakaryocytes have been successfully differentiated in cultures from bone marrow, fetal liver, and embryonic stem (ES) cells, while human megakaryocytes have been cultured from human cord blood, peripheral blood, and ES cells. The various sources of megakaryocyte progenitors provide choices to researchers, allowing them to access the most applicable systems. As examples, both bone marrow-derived and ES cell-derived murine megakaryocytes have been used to study proteins involved in integrin alphaIIbbeta3 regulation such as CIB1 and H-Ras. Therefore, megakaryocytes have provided an invaluable resource for better understanding the biology of platelets. In this chapter, we will describe: (1) approaches to obtain, generate, and characterize megakaryocytes, (2) molecular manipulation of these cells to elevate or decrease expression levels of specific proteins, and (3) current uses and future applications of megakaryocytes.


Subject(s)
Blood Platelets/physiology , Megakaryocytes/physiology , Animals , Blood Platelet Disorders/genetics , Blood Platelet Disorders/therapy , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/cytology , Genetic Vectors , Humans , Megakaryocytes/cytology , Mice , RNA Interference , Transduction, Genetic
8.
Circ Res ; 101(11): 1185-93, 2007 Nov 26.
Article in English | MEDLINE | ID: mdl-17975111

ABSTRACT

Pathological angiogenesis contributes to various ocular, malignant, and inflammatory disorders, emphasizing the need to understand this process on a molecular level. CIB1 (calcium- and integrin-binding protein), a 22-kDa EF-hand-containing protein, modulates the activity of p21-activated kinase 1 in fibroblasts. Because p21-activated kinase 1 also contributes to endothelial cell function, we hypothesized that CIB1 may have a role in angiogenesis. We found that endothelial cells depleted of CIB1 by either short hairpin RNA or homologous recombination have reduced migration, proliferation, and tubule formation. Moreover, loss of CIB1 in these cells decreases p21-activated kinase 1 activation, downstream extracellular signal-regulated kinase 1/2 activation, and matrix metalloproteinase 2 expression, all of which are known to contribute to angiogenesis. Consistent with these findings, tissues derived from CIB1-deficient (CIB1-/-) mice have reduced growth factor-induced microvessel sprouting in ex vivo organ cultures and in vivo Matrigel plugs. Furthermore, in response to ischemia, CIB1-/- mice demonstrate decreased pathological retinal and adaptive hindlimb angiogenesis. Ischemic CIB1-/- hindlimbs also demonstrate increased tissue damage and significantly reduced p21-activated kinase 1 activation. These data therefore reveal a critical role for CIB1 in ischemia-induced pathological and adaptive angiogenesis.


Subject(s)
Calcium-Binding Proteins/physiology , Endothelial Cells/cytology , Ischemia/physiopathology , Neovascularization, Pathologic , Neovascularization, Physiologic , Angiogenic Proteins/analysis , Animals , Calcium-Binding Proteins/deficiency , Endothelial Cells/physiology , Hindlimb/blood supply , Hindlimb/pathology , Mice , Mice, Knockout , p21-Activated Kinases/metabolism
9.
Mol Cell Biol ; 26(22): 8507-14, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16982698

ABSTRACT

CIB1 is a 22-kDa calcium binding, regulatory protein with approximately 50% homology to calmodulin and calcineurin B. CIB1 is widely expressed and binds to a number of effectors, such as integrin alphaIIb, PAK1, and polo-like kinases, in different tissues. However, the in vivo functions of CIB1 are not well understood. To elucidate the function of CIB1 in whole animals, we used homologous recombination in embryonic stem cells to generate Cib1(-/-) mice. Although Cib1(-/-) mice grow normally, the males are sterile due to disruption of the haploid phase of spermatogenesis. This is associated with reduced testis size and numbers of germ cells in seminiferous tubules, increased germ cell apoptosis, and the loss of elongated spermatids and sperm. Cib1(-/-) testes also show increased mRNA and protein expression of the cell cycle regulator Cdc2/Cdk1. In addition, mouse embryonic fibroblasts (MEFs) derived from Cib1(-/-) mice exhibit a much slower growth rate compared to Cib1(+/+) MEFs, suggesting that CIB1 regulates the cell cycle, differentiation of spermatogenic germ cells, and/or differentiation of supporting Sertoli cells.


Subject(s)
Calcium-Binding Proteins/physiology , Infertility, Male/chemically induced , Spermatogenesis , Testis/metabolism , Acetic Acid/chemistry , Animals , Apoptosis , CDC2 Protein Kinase/metabolism , Cell Proliferation , Epididymis/pathology , Fibroblasts/physiology , Formaldehyde/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Picrates/chemistry , Recombination, Genetic , Spermatids/physiology , Spermatocytes/physiology , Testis/pathology
10.
J Cell Biol ; 172(2): 169-75, 2006 Jan 16.
Article in English | MEDLINE | ID: mdl-16418530

ABSTRACT

In response to agonist stimulation, the alphaIIbbeta3 integrin on platelets is converted to an active conformation that binds fibrinogen and mediates platelet aggregation. This process contributes to both normal hemostasis and thrombosis. Activation of alphaIIbbeta3 is believed to occur in part via engagement of the beta3 cytoplasmic tail with talin; however, the role of the alphaIIb tail and its potential binding partners in regulating alphaIIbbeta3 activation is less clear. We report that calcium and integrin binding protein 1 (CIB1), which interacts directly with the alphaIIb tail, is an endogenous inhibitor of alphaIIbbeta3 activation; overexpression of CIB1 in megakaryocytes blocks agonist-induced alphaIIbbeta3 activation, whereas reduction of endogenous CIB1 via RNA interference enhances activation. CIB1 appears to inhibit integrin activation by competing with talin for binding to alphaIIbbeta3, thus providing a model for tightly controlled regulation of alphaIIbbeta3 activation.


Subject(s)
Calcium-Binding Proteins/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex , Animals , Blood Platelets/metabolism , Calcium-Binding Proteins/genetics , Fibrinogen/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , RNA Interference , Talin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL