Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Am Med Inform Assoc ; 25(3): 300-308, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29346583

ABSTRACT

OBJECTIVE: Finding relevant datasets is important for promoting data reuse in the biomedical domain, but it is challenging given the volume and complexity of biomedical data. Here we describe the development of an open source biomedical data discovery system called DataMed, with the goal of promoting the building of additional data indexes in the biomedical domain. MATERIALS AND METHODS: DataMed, which can efficiently index and search diverse types of biomedical datasets across repositories, is developed through the National Institutes of Health-funded biomedical and healthCAre Data Discovery Index Ecosystem (bioCADDIE) consortium. It consists of 2 main components: (1) a data ingestion pipeline that collects and transforms original metadata information to a unified metadata model, called DatA Tag Suite (DATS), and (2) a search engine that finds relevant datasets based on user-entered queries. In addition to describing its architecture and techniques, we evaluated individual components within DataMed, including the accuracy of the ingestion pipeline, the prevalence of the DATS model across repositories, and the overall performance of the dataset retrieval engine. RESULTS AND CONCLUSION: Our manual review shows that the ingestion pipeline could achieve an accuracy of 90% and core elements of DATS had varied frequency across repositories. On a manually curated benchmark dataset, the DataMed search engine achieved an inferred average precision of 0.2033 and a precision at 10 (P@10, the number of relevant results in the top 10 search results) of 0.6022, by implementing advanced natural language processing and terminology services. Currently, we have made the DataMed system publically available as an open source package for the biomedical community.

3.
Sci Data ; 4: 170059, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28585923

ABSTRACT

Today's science increasingly requires effective ways to find and access existing datasets that are distributed across a range of repositories. For researchers in the life sciences, discoverability of datasets may soon become as essential as identifying the latest publications via PubMed. Through an international collaborative effort funded by the National Institutes of Health (NIH)'s Big Data to Knowledge (BD2K) initiative, we have designed and implemented the DAta Tag Suite (DATS) model to support the DataMed data discovery index. DataMed's goal is to be for data what PubMed has been for the scientific literature. Akin to the Journal Article Tag Suite (JATS) used in PubMed, the DATS model enables submission of metadata on datasets to DataMed. DATS has a core set of elements, which are generic and applicable to any type of dataset, and an extended set that can accommodate more specialized data types. DATS is a platform-independent model also available as an annotated serialization in schema.org, which in turn is widely used by major search engines like Google, Microsoft, Yahoo and Yandex.

5.
J Transl Med ; 14: 46, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26861698

ABSTRACT

BACKGROUND: The majority of glioblastomas have aberrant receptor tyrosine kinase (RTK)/RAS/phosphoinositide 3 kinase (PI3K) signaling pathways and malignant glioma cells are thought to be addicted to these signaling pathways for their survival and proliferation. However, recent studies suggest that monotherapies or inappropriate combination therapies using the molecular targeted drugs have limited efficacy possibly because of tumor heterogeneities, signaling redundancy and crosstalk in intracellular signaling network, indicating necessity of rationale and methods for efficient personalized combination treatments. Here, we evaluated the growth of colonies obtained from glioma tumor-initiating cells (GICs) derived from glioma sphere culture (GSC) in agarose and examined the effects of combination treatments on GICs using targeted drugs that affect the signaling pathways to which most glioma cells are addicted. METHODS: Human GICs were cultured in agarose and treated with inhibitors of RTKs, non-receptor kinases or transcription factors. The colony number and volume were analyzed using a colony counter, and Chou-Talalay combination indices were evaluated. Autophagy and apoptosis were also analyzed. Phosphorylation of proteins was evaluated by reverse phase protein array and immunoblotting. RESULTS: Increases of colony number and volume in agarose correlated with the Gompertz function. GICs showed diverse drug sensitivity, but inhibitions of RTK and RAF/MEK or PI3K by combinations such as EGFR inhibitor and MEK inhibitor, sorafenib and U0126, erlotinib and BKM120, and EGFR inhibitor and sorafenib showed synergy in different subtypes of GICs. Combination of erlotinib and sorafenib, synergistic in GSC11, induced apoptosis and autophagic cell death associated with suppressed Akt and ERK signaling pathways and decreased nuclear PKM2 and ß-catenin in vitro, and tended to improve survival of nude mice bearing GSC11 brain tumor. Reverse phase protein array analysis of the synergistic treatment indicated involvement of not only MEK and PI3K signaling pathways but also others associated with glucose metabolism, fatty acid metabolism, gene transcription, histone methylation, iron transport, stress response, cell cycle, and apoptosis. CONCLUSION: Inhibiting RTK and RAF/MEK or PI3K could induce synergistic cytotoxicity but personalization is necessary. Examining colonies in agarose initiated by GICs from each patient may be useful for drug sensitivity testing in personalized cancer therapy.


Subject(s)
Glioma/drug therapy , Glioma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplastic Stem Cells/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , raf Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Inhibitory Concentration 50 , Male , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , raf Kinases/metabolism
6.
J Biomed Inform ; 58 Suppl: S189-S196, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26210361

ABSTRACT

OBJECTIVE: In recognition of potential barriers that may inhibit the widespread adoption of biomedical software, the 2014 i2b2 Challenge introduced a special track, Track 3 - Software Usability Assessment, in order to develop a better understanding of the adoption issues that might be associated with the state-of-the-art clinical NLP systems. This paper reports the ease of adoption assessment methods we developed for this track, and the results of evaluating five clinical NLP system submissions. MATERIALS AND METHODS: A team of human evaluators performed a series of scripted adoptability test tasks with each of the participating systems. The evaluation team consisted of four "expert evaluators" with training in computer science, and eight "end user evaluators" with mixed backgrounds in medicine, nursing, pharmacy, and health informatics. We assessed how easy it is to adopt the submitted systems along the following three dimensions: communication effectiveness (i.e., how effective a system is in communicating its designed objectives to intended audience), effort required to install, and effort required to use. We used a formal software usability testing tool, TURF, to record the evaluators' interactions with the systems and 'think-aloud' data revealing their thought processes when installing and using the systems and when resolving unexpected issues. RESULTS: Overall, the ease of adoption ratings that the five systems received are unsatisfactory. Installation of some of the systems proved to be rather difficult, and some systems failed to adequately communicate their designed objectives to intended adopters. Further, the average ratings provided by the end user evaluators on ease of use and ease of interpreting output are -0.35 and -0.53, respectively, indicating that this group of users generally deemed the systems extremely difficult to work with. While the ratings provided by the expert evaluators are higher, 0.6 and 0.45, respectively, these ratings are still low indicating that they also experienced considerable struggles. DISCUSSION: The results of the Track 3 evaluation show that the adoptability of the five participating clinical NLP systems has a great margin for improvement. Remedy strategies suggested by the evaluators included (1) more detailed and operation system specific use instructions; (2) provision of more pertinent onscreen feedback for easier diagnosis of problems; (3) including screen walk-throughs in use instructions so users know what to expect and what might have gone wrong; (4) avoiding jargon and acronyms in materials intended for end users; and (5) packaging prerequisites required within software distributions so that prospective adopters of the software do not have to obtain each of the third-party components on their own.


Subject(s)
Attitude to Computers , Data Mining/statistics & numerical data , Electronic Health Records/statistics & numerical data , Natural Language Processing , Pattern Recognition, Automated/methods , Software , Data Mining/methods , Humans , Middle Aged , User-Computer Interface
7.
J Biol Chem ; 290(18): 11749-61, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25795775

ABSTRACT

The RET proto-oncogene, a tyrosine kinase receptor, is widely known for its essential role in cell survival. Germ line missense mutations, which give rise to constitutively active oncogenic RET, were found to cause multiple endocrine neoplasia type 2, a dominant inherited cancer syndrome that affects neuroendocrine organs. However, the mechanisms by which RET promotes cell survival and prevents cell death remain elusive. We demonstrate that in addition to cytoplasmic localization, RET is localized in the nucleus and functions as a tyrosine-threonine dual specificity kinase. Knockdown of RET by shRNA in medullary thyroid cancer-derived cells stimulated expression of activating transcription factor 4 (ATF4), a master transcription factor for stress-induced apoptosis, through activation of its target proapoptotic genes NOXA and PUMA. RET knockdown also increased sensitivity to cisplatin-induced apoptosis. We observed that RET physically interacted with and phosphorylated ATF4 at tyrosine and threonine residues. Indeed, RET kinase activity was required to inhibit the ATF4-dependent activation of the NOXA gene because the site-specific substitution mutations that block threonine phosphorylation increased ATF4 stability and activated its targets NOXA and PUMA. Moreover, chromatin immunoprecipitation assays revealed that ATF4 occupancy increased at the NOXA promoter in TT cells treated with tyrosine kinase inhibitors or the ATF4 inducer eeyarestatin as well as in RET-depleted TT cells. Together these findings reveal RET as a novel dual kinase with nuclear localization and provide mechanisms by which RET represses the proapoptotic genes through direct interaction with and phosphorylation-dependent inactivation of ATF4 during the pathogenesis of medullary thyroid cancer.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Proto-Oncogene Proteins c-ret/metabolism , Activating Transcription Factor 4/chemistry , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cisplatin/pharmacology , Gene Expression Regulation/drug effects , Humans , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Threonine/metabolism , Transcription, Genetic/drug effects
8.
Neoplasia ; 15(1): 73-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23359207

ABSTRACT

The hepatocyte growth factor receptor (c-Met) and a constitutively active mutant of the epidermal growth factor receptor (ΔEGFR/EGFRvIII) are frequently overexpressed in glioblastoma (GBM) and promote tumorigenesis. The mechanisms underlying elevated hepatocyte growth factor (HGF) production in GBM are not understood. We found higher, coordinated mRNA expression levels of HGF and c-Met in mesenchymal (Mes) GBMs, a subtype associated with poor treatment response and shorter overall survival. In an HGF/c-Met-dependent GBM cell line, HGF expression declined upon silencing of c-Met using RNAi or by inhibiting its activity with SU11274. Silencing c-Met decreased anchorage-independent colony formation and increased the survival of mice bearing intracranial GBM xenografts. Consistent with these findings, c-Met activation by ΔEGFR also elevated HGF expression, and the inhibition of ΔEGFR with AG1478 reduced HGF levels. Interestingly, c-Met expression was required for ΔEGFR-mediated HGF production, anchorage-independent growth, and in vivo tumorigenicity, suggesting that these pathways are coupled. Using an unbiased mass spectrometry-based screen, we show that signal transducer and activator of transcription 3 (STAT3) Y705 is a downstream target of c-Met signaling. Suppression of STAT3 phosphorylation with WP1193 reduced HGF expression in ΔEGFR-expressing GBM cells, whereas constitutively active STAT3 partially rescued HGF expression and colony formation in c-Met knockdown cells expressing ΔEGFR. These results suggest that the c-Met/HGF signaling axis is enhanced by ΔEGFR through increased STAT3-dependent HGF expression and that targeting c-Met in Mes GBMs may be an important strategy for therapy.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , Hepatocyte Growth Factor/biosynthesis , Proto-Oncogene Proteins c-met/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyanoacrylates/metabolism , ErbB Receptors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , HEK293 Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Mice , Mice, Nude , Phosphorylation/genetics , Proto-Oncogene Proteins c-met/genetics , Pyridines/metabolism , RNA Interference , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Cells, Cultured
9.
J Biol Chem ; 288(5): 3428-38, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23250739

ABSTRACT

ΔEGFR is a potent glioblastoma oncogene which has been studied primarily as a plasma membrane kinase. Using intracranial xenograft studies in mice, we show that blocking ΔEGFR access to the nucleus attenuates its tumorigenicity and, conversely, that promoting nuclear accumulation enhances this, providing the first in vivo evidence that the nuclear actions of ΔEGFR contribute strongly to its oncogenic function. Nuclear actions of ΔEGFR include regulation of gene expression by participation in chromatin-bound complexes, and genome-wide mapping of these sequences by chromatin immunoprecipitation and massively parallel sequencing identified 2294 peaks. Bioinformatic analysis showed enrichment of the E-box motif in the dataset, and c-Myc and ΔEGFR were corecruited to the promoters of and transcriptionally activated a subset of nuclear ΔEGFR chromatin targets. Knockdown of c-Myc decreased the expression of these targets and diminished ΔEGFR-stimulated anchorage-independent colony formation. We conclude that transcriptional regulation of target genes by association with gene regulatory chromatin in cooperation with c-Myc by nuclear ΔEGFR makes a unique contribution to its oncogenicity and propose that this venue provides new targets for therapeutic intervention.


Subject(s)
Cell Nucleus/metabolism , Cell Transformation, Neoplastic/metabolism , ErbB Receptors/metabolism , Mutation/genetics , Proto-Oncogene Proteins c-myc/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Transformation, Neoplastic/pathology , Chromatin Immunoprecipitation , E-Box Elements/genetics , ErbB Receptors/chemistry , Genome, Human/genetics , Glioma/metabolism , Humans , Mice , Mice, Nude , Mutant Proteins/metabolism , Nuclear Export Signals , Nuclear Localization Signals/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Multimerization , Protein Transport , Transcription Factors/metabolism
10.
J Gene Med ; 12(12): 968-80, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21104971

ABSTRACT

BACKGROUND: Inhibition of tumor angiogenesis is a promising approach for cancer therapy and the Tie-2/angiopoietin pathway appears to play an important role. In the present study, we have developed strategies to explore the therapeutic potential of blocking the Tie-2/angiopoietin pathway by sTie-2. METHODS: Ehrlich ascites tumor (EAT) cells were stably transfected to overexpress a truncated form of sTie-2. Transfectants were characterized for their in vitro growth behavior and transplanted into nude mice. Furthermore, recombinant sTie-2 produced by the baculovirus expression system was used to sequester angiopoietins in the murine ascites carcinoma model. The effect of sTie-2 treatment alone or in combination with sFLT-1 on the weight of the animal, ascites cell number and volume was studied. RESULTS: EAT cells stably transfected with a truncated form of sTie-2 showed no change in cell proliferation in vitro and colony forming in soft agar compared to control cells. However, sTie-2 transfected EAT cells transplanted into nude mice reduced tumor burden and demonstrated a reduction in ascites formation and peritoneal angiogenesis. Recombinant sTie-2 showed angiogenic activity in the tube formation and wound healing assay in vitro. sTie-2 treatment alone or in combination with sFLT-1 in an ascites tumor mouse model resulted in reduced peritoneal angiogenesis, with a concomitant decrease in tumor cell number, volume of ascites and the number of invasive tumor cells, as assayed by CD31 staining. CONCLUSIONS: The findings of the present study demonstrate an important role for the Tie-2/angiopoietin pathway in the formation of tumor vasculature and suggest that sTie-2 might yield useful anticancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ehrlich Tumor/pathology , Carcinoma, Ehrlich Tumor/therapy , Neovascularization, Pathologic/prevention & control , Receptor, TIE-2/therapeutic use , Vascular Endothelial Growth Factor Receptor-1/therapeutic use , Angiopoietin-2 , Animals , Carcinoma, Ehrlich Tumor/metabolism , Cell Proliferation/drug effects , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Receptor, TIE-2/genetics , Solubility , Transfection/methods , Transplants , Treatment Outcome
11.
EMBO Rep ; 11(9): 691-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20651739

ABSTRACT

High expression of metastasis-associated protein 1 co-regulator (MTA1), a component of the nuclear remodelling and histone deacetylase complex, has been associated with human tumours. However, the precise role of MTA1 in tumorigenesis remains unknown. In this study, we show that induced levels of MTA1 are sufficient to transform Rat1 fibroblasts and that the transforming potential of MTA1 is dependent on its acetylation at Lys626. Underlying mechanisms of MTA1-mediated transformation include activation of the Ras-Raf pathway by MTA1 but not by acetylation-inactive MTA1; this was due to the repression of Galphai2 transcription, which negatively influences Ras activation. We observed that acetylated MTA1-histone deacetylase (HDAC) interaction was required for the recruitment of the MTA1-HDAC complex to the Galphai2 regulatory element and consequently for the repression of Galphai2 transcription and expression leading to activation of the Ras-Raf pathway. The findings presented in this study provide for the first time--to the best of our knowledge--evidence of acetylation-dependent oncogenic activity of a cancer-relevant gene product.


Subject(s)
Cell Transformation, Neoplastic , Histone Deacetylases/metabolism , Oncogenes , Repressor Proteins/metabolism , Acetylation , Animals , Cell Line , Cell Movement , Female , Fibroblasts/cytology , Fibroblasts/physiology , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Humans , Lysine/metabolism , Mice , Mice, Nude , Neoplasms, Experimental , Repressor Proteins/genetics , Trans-Activators , Transcription, Genetic , Transplantation, Heterologous , ras Proteins/genetics , ras Proteins/metabolism
12.
J Gene Med ; 11(5): 422-34, 2009 May.
Article in English | MEDLINE | ID: mdl-19266483

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) is known to play a major role in angiogenesis. A soluble form of Flt-1, a VEGF receptor, is potentially useful as an antagonist of VEGF, and accumulating evidence suggests the applicability of sFlt-1 in tumor suppression. In the present study, we have developed and tested strategies targeted specifically to VEGF for the treatment of ascites formation. METHODS: As an initial strategy, we produced recombinant sFLT-1 in the baculovirus expression system and used it as a trap to sequester VEGF in the murine ascites carcinoma model. The effect of the treatment on the weight of the animal, cell number, ascites volume and proliferating endothelial cells was studied. The second strategy involved, producing Ehrlich ascites tumor (EAT) cells stably transfected with vectors carrying cDNA encoding truncated form of Flt-1 and using these cells to inhibit ascites tumors in a nude mouse model. RESULTS: The sFLT-1 produced by the baculovirus system showed potent anti-angiogenic activity as assessed by rat cornea and tube formation assay. sFLT-1 treatment resulted in reduced peritoneal angiogenesis with a concomitant decrease in tumor cell number, volume of ascites, amount of free VEGF and the number of invasive tumor cells as assayed by CD31 staining. EAT cells stably transfected with truncated form of Flt-1 also effectively reduced the tumor burden in nude mice transplanted with these cells, and demonstrated a reduction in ascites formation and peritoneal angiogenesis. CONCLUSIONS: The inhibition of peritoneal angiogenesis and tumor growth by sequestering VEGF with either sFlt-1 gene expression by recombinant EAT cells or by direct sFLT-1 protein therapy is shown to comprise a potential therapy.


Subject(s)
Ascites/pathology , Ascites/therapy , Paracrine Communication , Transfection , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/therapeutic use , Animals , Baculoviridae , Cell Proliferation , Mice , Neovascularization, Pathologic , Rats , Recombinant Proteins/metabolism , Reproducibility of Results , Solubility
13.
Cancer Res ; 67(15): 7062-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671172

ABSTRACT

Metastasis-associated protein 1 (MTA1), a component of the nuclear remodeling complex and the founding homologue of the MTA family, has been implicated in metastasis, but definitive causative evidence in an animal model system is currently lacking. Here, we show that MTA1 overexpression in transgenic mice is accompanied by a high incidence of spontaneous B cell lymphomas including diffuse large B cell lymphomas (DLBCL). Lymphocytes and lymphoma cells from MTA1-TG mice are hyperproliferative. Lymphomas were transplantable and of clonal origin and were characterized by down-regulation of p27Kip1 as well as up-regulation of Bcl2 and cyclin D1. The significance of these murine studies was established by evidence showing a widespread up-regulation of MTA1 in DLBCL from humans. These findings reveal a previously unrecognized role for the MTA1 pathway in the development of spontaneous B cell lymphomas, and offer a potential therapeutic target in B cell lymphomas. These observations suggest that MTA1-TG mice represent a new model of spontaneous DLBCL associated with high tumor incidence and could be used for therapeutic intervention studies.


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Neoplastic/physiology , Lymphoma, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Transcription Factors/genetics , Animals , Blotting, Southern , Cell Proliferation , Female , Histone Deacetylases/genetics , Humans , Lymph Nodes/pathology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/etiology , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Tumor Cells, Cultured
14.
Cancer Res ; 67(15): 7132-8, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671180

ABSTRACT

Previously, we have shown that metastasis-associated protein 1 (MTA1) overexpression in transgenic mice was accompanied by high incidence of spontaneous B-cell lymphomas including diffuse large B-cell lymphomas (DLBCL). To understand the molecular basis of lymphoma in MTA1-transgenic (MTA1-TG) mice, we wished to identify a putative MTA1 target with a causal role in B-cell lymphogenesis. Using chromatin immunoprecipitation assays, we identified paired box gene 5 (Pax5), a molecule previously implicated in B-cell lymphogenesis, as a potential downstream effector of MTA1. Lymphomas from MTA1-TG mice also showed up-regulation of Pax5. We also found that MTA1 acetylated on Lys(626) interacted with p300 histone acetyltransferase, and that acetylated MTA1 was recruited to the Pax5 promoter to stimulate Pax5 transcription. Global gene profiling identified down-regulation of a set of genes, including those downstream of Pax5 and directly implicated in the B-cell lymphogenesis. Significance of these murine studies was established by evidence showing a widespread up-regulation of both MTA1 and Pax5 in DLBCL from humans. These observations provide in vivo genetic evidence for a role of MTA1 in lymphomagenesis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Lymphoma, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , PAX5 Transcription Factor/genetics , Transcription Factors/physiology , Animals , Blotting, Northern , Chromatin Immunoprecipitation , Gene Expression Profiling , Histone Deacetylase 1 , Histone Deacetylases/genetics , Humans , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Plasmids , Promoter Regions, Genetic , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Transcriptional Activation , Transfection , Tumor Cells, Cultured
15.
Mol Endocrinol ; 21(8): 1847-60, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17505058

ABSTRACT

We recently reported that the breast carcinoma amplified sequence-3 (BCAS3) gene is regulated by estrogen receptor (ER) alpha. However, the role of ERalpha coactivators in the regulation of BCAS3 expression remains unknown, and information regarding the function of the BCAS3 protein is lacking. Here, we define the contribution of ERalpha coactivators to BCAS3 regulation and identify BCAS3 itself as an ERalpha coactivator in breast cancer cells. We found that PELP1 (proline-, glutamic acid-, and leucine-rich protein-1), a newly described ERalpha coregulator, is recruited to BCAS3 chromatin and activates its expression. Analysis of the BCAS3 sequence for functional motifs and evidence from biochemical fractionation suggested that BCAS3 acts as a transcriptional coactivator. Results from chromatin immunoprecipitation, reporter assays, and expression studies further validated the coactivator function of BCAS3 for ERalpha. BCAS3 physically associated with histone H3 and histone acetyltransferase complex protein P/CAF (p300/CBP-associated factor) and possessed histone acetyltransferase activity. Unexpectedly, BCAS3 required PELP1 to function as a coactivator in ERalpha transactivation activity. In brief, these results highlight a mechanism whereby ERalpha activation triggers a positive feedback loop leading to signal amplification in the cell.


Subject(s)
Estrogens/physiology , Gene Expression Regulation, Neoplastic/physiology , Neoplasm Proteins/genetics , Trans-Activators/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Co-Repressor Proteins , Female , Histone Acetyltransferases/metabolism , Humans , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Transcription Factors/metabolism , Tumor Cells, Cultured , p300-CBP Transcription Factors
16.
Proc Natl Acad Sci U S A ; 104(14): 5866-71, 2007 Apr 03.
Article in English | MEDLINE | ID: mdl-17389360

ABSTRACT

Transcription, splicing, and translation are potentially coordinately regulatable in a temporospatial-dependent manner, although supporting experimental evidence for this notion is scarce. Yeast two-hybrid screening of a mammary gland cDNA library with human p21-activated kinase 1 (Pak1) as bait identified polyC-RNA-binding protein 1 (PCBP1), which controls translation from mRNAs containing the DICE (differentiation control element). Mitogenic stimulation of human cells phosphorylated PCBP1 on threonines 60 and 127 in a Pak1-sensitive manner. Pak1-dependent phosphorylation of PCBP1 released its binding and translational inhibition from a DICE-minigene. Overexpression of PCBP1 also inhibited the translation of the endogenous L1 cell adhesion molecule mRNA, which contains two DICE motifs in the 3' untranslated region. We also found that Pak1 activation led to an increased nuclear retention of PCBP1, recruitment to the eukaryotic translation initiation factor 4E (eIF4E) promoter, and stimulation of eIF4E expression in a Pak1-sensitive manner. Moreover, mitogenic stimulation promoted Pak1- and PCBP1-dependent alternative splicing and exon inclusion from a CD44 minigene. The alternative splicing functions of PCBP1 were in turn mediated by its intrinsic interaction with Caper alpha, a U2 snRNP auxiliary factor-related protein previously implicated in RNA splicing. These findings establish the principle that a single coregulator can function as a signal-dependent and coordinated regulator of transcription, splicing, and translation.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/physiology , Protein Biosynthesis , RNA Splicing , Signal Transduction , Transcription, Genetic , DNA, Complementary , DNA-Binding Proteins , Enzyme Activation , Eukaryotic Initiation Factor-4E/metabolism , Female , Gene Library , Humans , Hyaluronan Receptors/metabolism , Kinetics , Models, Biological , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins , Two-Hybrid System Techniques , p21-Activated Kinases
17.
Cell Cycle ; 5(13): 1407-10, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16855396

ABSTRACT

Lysine acetylation occurs in many protein targets, including core histones, transcription factors, and other proteins. Metastasis-associated protein 1 (MTA1) is implicated in the progression and metastasis of various epithelial tumors. Because MTA1 functions as a transcriptional coregulator, much of its role in cancer promoting processes are likely to involve its ability to regulate the transcription of downstream target genes that encode effector proteins. We recently showed that MTA1 could be post-translationally modified by acetylation, which modulates its function as a coregulator molecule. We also defined a chromatin target of MTA1, namely, breast cancer-amplified sequence 3 (BCAS3), in the context of which MTA1 behaves as a transcriptional coactivator in breast cancer cells. Because the phenotypic effect of BCAS3 overexpression in tumors has not been defined, we investigated the consequence of increased expression of BCAS3 in human breast tumors. Here, we report that BCAS3 overexpression in hormone receptor-positive premenopausal breast cancer seemed to be associated with impaired responses to tamoxifen. Our findings have implications for endocrine therapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Histone Deacetylases/metabolism , Neoplasm Proteins/metabolism , Premenopause , Repressor Proteins/metabolism , Tamoxifen/pharmacology , Acetylation , Animals , Breast Neoplasms/pathology , Female , Humans , Neoplasm Proteins/genetics , Survival Rate , Trans-Activators
18.
Nat Rev Cancer ; 6(6): 459-71, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16723992

ABSTRACT

The pivotal role of kinases in signal transduction and cellular regulation has lent them considerable appeal as pharmacological targets across a broad spectrum of cancers. p21-activated kinases (Paks) are serine/threonine kinases that function as downstream nodes for various oncogenic signalling pathways. Paks are well-known regulators of cytoskeletal remodelling and cell motility, but have recently also been shown to promote cell proliferation, regulate apoptosis and accelerate mitotic abnormalities, which results in tumour formation and cell invasiveness. Alterations in Pak expression have been detected in human tumours, which makes them an attractive new therapeutic target.


Subject(s)
Neoplasms/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis , Cell Movement , Cytoskeleton/metabolism , Humans , Models, Biological , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , p21-Activated Kinases
19.
Proc Natl Acad Sci U S A ; 103(17): 6670-5, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16617102

ABSTRACT

Here we define a function of metastasis-associated protein 1 (MTA1), a presumed corepressor of estrogen receptor alpha (ERalpha), as a transcriptional activator of Breast Cancer Amplified Sequence 3 (BCAS3), a gene amplified and overexpressed in breast cancers. We identified BCAS3 as a MTA1 chromatin target in a functional genomic screen. MTA1 stimulation of BCAS3 transcription required ERalpha and involved a functional ERE half-site in BCAS3. Furthermore, we discovered that MTA1 is acetylated on lysine 626, and that this acetylation is necessary for a productive transcriptional recruitment of RNA polymerase II complex to the BCAS3 enhancer sequence. BCAS3 expression was elevated in mammary tumors from MTA1 transgenic mice and 60% of the human breast tumors, and correlated with the coexpression of MTA1 as well as with tumor grade and proliferation of primary breast tumor samples. These findings reveal a previously unrecognized function of MTA1 in stimulating BCAS3 expression and suggest an important role for MTA1-BCAS3 pathway in promoting cancerous phenotypes in breast tumor cells.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Acetylation , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Enhancer Elements, Genetic , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/chemistry , Humans , Molecular Sequence Data , Neoplasm Proteins/chemistry , Repressor Proteins/chemistry , Trans-Activators/chemistry
20.
J Biol Chem ; 281(22): 15394-404, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16574651

ABSTRACT

Retinoid X receptor alpha (RXRalpha), functioning as either a homodimer or a heterodimer with peroxisome proliferator receptors, is known to be involved in manifesting antiproliferative effects in cells. Consequently, studies of RXRalpha functions and its coregulators have been in the focus for therapeutic approaches against cancer. Here we have discovered that 9-cis-retinoic acid (9-cis-RA), a RXRalpha-specific ligand, up-regulated the expression of transcriptional coregulatory protein PELP1 (proline-, glutamic acid-, and leucine-rich protein 1). PELP1 functioned as a coactivator of RXRalpha, increasing its transactivation function in response to 9-cis-RA as evident by the retinoid X receptor response element-luciferase assays. PELP1 was found to be a binding partner of RXRalpha, and the binding interactions were confirmed both in vitro and in vivo. An electrophoretic mobility shift assay showed greater formation and stability of RXRalpha homodimers on consensus oligonucleotides in PELP1-overexpressing clones in comparison to the pcDNA clones. The presence of PELP1 in these oligonucleotide-bound RXRalpha homodimers was proved by the supershift of the complex when incubated with PELP1-specific antibody. PELP1-overexpressing stable MCF-7 cells exhibited a significantly higher extent of 9-cis-RA-induced apoptosis than the control pcDNA clones. Silencing of PELP1 expression in parental MCF-7 cells and PELP1-overexpressing clones using PELP1-specific RNA-mediated interference compromised the susceptibility to 9-cis-RA-induced apoptosis. PELP1 could also function as a coactivator of the RXRalpha-peroxisome proliferator-activated receptor (PPARgamma) heterodimer as evident by the peroxisome proliferator-activated receptor response element-luciferase assay in response to both 9-cis-RA and PPARgamma-specific ligands. This was reinforced by the higher propensity of PELP1-overexpressing clones to undergo differentiation in response to PPARgamma-specific ligands. This study has revealed a novel facet of PELP1 functions and identified it to be an important potentiator of the antiproliferative effects of 9-cis-RA and PPARgamma-specific ligands.


Subject(s)
Retinoid X Receptor alpha/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Tretinoin/pharmacology , Alitretinoin , Apoptosis/drug effects , Base Sequence , Cell Differentiation/drug effects , Cell Line, Tumor , Co-Repressor Proteins , Dimerization , Humans , In Vitro Techniques , Ligands , PPAR gamma/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Retinoid X Receptor alpha/chemistry , Transcription Factors , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...