Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Acta Neuropathol Commun ; 12(1): 42, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38500181

ABSTRACT

Central nervous system (CNS) embryonal tumors are a heterogeneous group of high-grade malignancies, and the increasing clinical use of methylation profiling and next-generation sequencing has led to the identification of molecularly distinct subtypes. One proposed tumor type, CNS tumor with BRD4::LEUTX fusion, has been described. As only a few CNS tumors with BRD4::LEUTX fusions have been described, we herein characterize a cohort of 9 such cases (4 new, 5 previously published) to further describe their clinicopathologic and molecular features. We demonstrate that CNS embryonal tumor with BRD4::LEUTX fusion comprises a well-defined methylation class/cluster. We find that patients are young (4 years or younger), with large tumors at variable locations, and frequently with evidence of leptomeningeal/cerebrospinal fluid (CSF) dissemination. Histologically, tumors were highly cellular with high-grade embryonal features. Immunohistochemically, 5/5 cases showed synaptophysin and 4/5 showed OLIG2 expression, thus overlapping with CNS neuroblastoma, FOXR2-activated. DNA copy number profiles were generally flat; however, two tumors had chromosome 1q gains. No recurring genomic changes, besides the presence of the fusion, were found. The LEUTX portion of the fusion transcript was constant in all cases assessed, while the BRD4 portion varied but included a domain with proto-oncogenic activity in all cases. Two patients with clinical follow up available had tumors with excellent response to chemotherapy. Two of our patients were alive without evidence of recurrence or progression after gross total resection and chemotherapy at 16 and 33 months. One patient relapsed, and the last of our four patients died of disease one month after diagnosis. Overall, this case series provides additional evidence for this as a distinct tumor type defined by the presence of a specific fusion as well as a distinct DNA methylation signature. Studies on larger series are required to further characterize these tumors.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Neoplasms, Germ Cell and Embryonal , Humans , Brain Neoplasms/pathology , Nuclear Proteins/genetics , Transcription Factors/genetics , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Neoplasms, Germ Cell and Embryonal/genetics , Bromodomain Containing Proteins , Cell Cycle Proteins , Forkhead Transcription Factors , Homeodomain Proteins
4.
Acta Neuropathol ; 143(3): 403-414, 2022 03.
Article in English | MEDLINE | ID: mdl-35103816

ABSTRACT

Tumors of the central nervous system (CNS) often display a wide morphologic spectrum that has, until recently, been the sole basis for tumor classification. The introduction of the integrated histomolecular diagnostic approach in CNS tumors has facilitated a classification system that is increasingly data-driven and with improved alignment to clinical outcome. Here, we report a previously uncharacterized glioma type (n = 31) using unsupervised clustering analysis of DNA methylation array data from approximately 14,000 CNS tumor samples. Histologic examination revealed circumscribed growth and morphologic similarities to pleomorphic xanthoastrocytoma (PXA), astroblastoma, ependymoma, polymorphous neuroepithelial tumor of the young (PLNTY), and IDH-wildtype glioblastoma (GBM). Median age (46.5 years) was significantly older than other circumscribed gliomas and younger than GBM. Dimensionality reduction with uniform manifold approximation and projection (UMAP) and hierarchical clustering confirmed a methylation signature distinct from known tumor types and methylation classes. DNA sequencing revealed recurrent mutations in TP53 (57%), RB1 (26%), NF1 (26%), and NF2 (14%). BRAF V600E mutations were detected in 3/27 sequenced cases (12%). Copy number analysis showed increased whole chromosome aneuploidy with recurrent loss of chromosome 13 (28/31 cases, 90%). CDKN2A/B deletion (2/31, 6%) and MGMT promoter methylation (1/31, 3%) were notably rare events. Most tumors showed features of a high-grade glioma, yet survival data showed significantly better overall survival compared to GBM (p < 0.0001). In summary, we describe a previously uncharacterized glioma of adults identified by a distinct DNA methylation signature and recurrent loss of chromosome 13.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Monosomy , Mutation , Tumor Suppressor Protein p53 , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Chromosomes, Human, Pair 13 , Humans , Middle Aged , Mutation/genetics , Tumor Suppressor Protein p53/genetics
5.
Acta Neuropathol Commun ; 10(1): 19, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35144680

ABSTRACT

The tumor micro-environment (TME) plays an important role in various cancers, including gliomas. We estimated immune cell type-specific gene expression profiles in 3 large clinically annotated glioma datasets using CIBERSORTx and LM22/LM10 blood-based immune signatures and found that the proportions and estimated gene expression patterns of specific immune cells significantly varied according to IDH mutation status. When IDH-WT and IDH-MUT tumors were considered separately, cluster-of-cluster analyses of immune cell gene expression identified groups with distinct survival outcomes. We confirmed and extended these findings by applying a signature matrix derived from single-cell RNA-sequencing data derived from 19 glioma tumor samples to the bulk profiling data, validating findings from the LM22/LM10 results. To link immune cell signatures with outcomes in checkpoint therapy, we then showed a significant association of monocytic lineage cell gene expression clusters with patient survival and with mesenchymal gene expression scores. Integrating immune cell-based gene expression with previously described malignant cell states in glioma demonstrated that macrophage M0 abundance significantly correlated with mesenchymal state in IDH-WT gliomas, with evidence of a previously implicated role of the Oncostatin-M receptor and macrophages in the mesenchymal state. Among IDH-WT tumors that were enriched for the mesenchymal cell state, the estimated M0 macrophage expression signature coordinately also trended to a mesenchymal signature. We also examined IDH-MUT tumors stratified by 1p/19q status, showing that a mesenchymal gene expression signature the M0 macrophage fraction was enriched in IDH-MUT, non-codeleted tumors. Overall, these results highlight the biological and clinical significance of the immune cell environment related to IDH mutation status, patient prognosis and the mesenchymal state in diffuse gliomas.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/pathology , Glioma/immunology , Glioma/pathology , Tumor Microenvironment/immunology , Brain Neoplasms/genetics , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Macrophages/immunology , Mutation , Prognosis , Transcriptome
6.
Neuro Oncol ; 24(4): 571-581, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34555175

ABSTRACT

BACKGROUND: Accurate CNS tumor diagnosis can be challenging, and methylation profiling can serve as an adjunct to classify diagnostically difficult cases. METHODS: An integrated diagnostic approach was employed for a consecutive series of 1258 surgical neuropathology samples obtained primarily in a consultation practice over 2-year period. DNA methylation profiling and classification using the DKFZ/Heidelberg CNS tumor classifier was performed, as well as unsupervised analyses of methylation data. Ancillary testing, where relevant, was performed. RESULTS: Among the received cases in consultation, a high-confidence methylation classifier score (>0.84) was reached in 66.4% of cases. The classifier impacted the diagnosis in 46.7% of these high-confidence classifier score cases, including a substantially new diagnosis in 26.9% cases. Among the 289 cases received with only a descriptive diagnosis, methylation was able to resolve approximately half (144, 49.8%) with high-confidence scores. Additional methods were able to resolve diagnostic uncertainty in 41.6% of the low-score cases. Tumor purity was significantly associated with classifier score (P = 1.15e-11). Deconvolution demonstrated that suspected glioblastomas (GBMs) matching as control/inflammatory brain tissue could be resolved into GBM methylation profiles, which provided a proof-of-concept approach to resolve tumor classification in the setting of low tumor purity. CONCLUSIONS: This work assesses the impact of a methylation classifier and additional methods in a consultative practice by defining the proportions with concordant vs change in diagnosis in a set of diagnostically challenging CNS tumors. We address approaches to low-confidence scores and confounding issues of low tumor purity.


Subject(s)
Central Nervous System Neoplasms , Glioblastoma , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/genetics , DNA Methylation , Glioblastoma/diagnosis , Glioblastoma/genetics , Humans
7.
J Thorac Oncol ; 15(5): 843-859, 2020 05.
Article in English | MEDLINE | ID: mdl-32004714

ABSTRACT

INTRODUCTION: BRCA1-associated protein-1 (BAP1), a nuclear deubiquitinase thought to be involved in DNA double-strand break repair, is frequently mutated in mesothelioma. Because poly(adenosine diphosphate-ribose) polymerase inhibitors (PARPIs) induce synthetic lethality in BRCA1/2 mutant cancers, we evaluated whether BAP1 inactivating mutations confer sensitivity to PARPIs in mesothelioma and if combination therapy with temozolomide (TMZ) would be beneficial. METHODS: A total of 10 patient-derived mesothelioma cell lines were generated and characterized for BAP1 mutation status, protein expression, nuclear localization, and sensitivity to the PARPIs, olaparib, and talazoparib, alone or in combination with TMZ. BAP1 deubiquitinase (DUB) activity was evaluated by ubiquitin with 7-amido-4-methylcoumarin assay. BAP1 knockout mesothelioma cell lines were generated by CRISPR-Cas9. Because Schlafen 11 (SLFN11) and O6-methylguanine-DNA methyltransferase also drive response to TMZ and PARPIs, we tested their expression and relationship with drug response. RESULTS: BAP1 mutations or copy-number alterations, or both were present in all 10 cell lines. Nonetheless, four cell lines exhibited intact DUB activity and two had nuclear BAP1 localization. Half maximal-inhibitory concentrations of olaparib and talazoparib ranged from 4.8 µM to greater than 50 µM and 0.039 µM to greater than 5 µM, respectively, classifying them into sensitive (two) or resistant (seven) cells, independent of their BAP1 status. Cell lines with BAP1 knockout resulted in the loss of BAP1 DUB activity but did not increase sensitivity to talazoparib. Response to PARPI tended to be associated with high SLFN11 expression, and combination with temozolomide increased sensitivity of cells with low or no MGMT expression. CONCLUSIONS: BAP1 status does not determine sensitivity to PARPIs in patient-derived mesothelioma cell lines. Combination of PARPI with TMZ may be beneficial for patients whose tumors have high SLFN11 and low or no MGMT expression.


Subject(s)
Lung Neoplasms , Mesothelioma , Cell Line, Tumor , Guanine/analogs & derivatives , Humans , Mesothelioma/drug therapy , Mesothelioma/genetics , O(6)-Methylguanine-DNA Methyltransferase , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
8.
Haematologica ; 105(4): 951-960, 2020 04.
Article in English | MEDLINE | ID: mdl-31439678

ABSTRACT

Histiocytic sarcoma is a rare malignant neoplasm that may occur de novo or in the context of a previous hematologic malignancy or mediastinal germ cell tumor. Here, we performed whole exome sequencing and RNA-sequencing (RNA-Seq) on 21 archival cases of primary histiocytic sarcoma. We identified a high number of genetic alterations within the RAS/RAF/MAPK pathway in 21 of 21 cases, with alterations in NF1 (6 of 21), MAP2K1 (5 of 21), PTPN11 (4 of 21), BRAF (4 of 21), KRAS (4 of 21), NRAS (1 of 21), and LZTR1 (1 of 21), including single cases with homozygous deletion of NF1, high-level amplification of PTPN11, and a novel TTYH3-BRAF fusion. Concurrent NF1 and PTPN11 mutations were present in 3 of 21 cases, and 5 of 7 cases with alterations in NF1 and/or PTPN11 had disease involving the gastrointestinal tract. Following unsupervised clustering of gene expression data, cases with NF1 and/or PTPN11 abnormalities formed a distinct tumor subgroup. A subset of NF1/PTPN11 wild-type cases had frequent mutations in B-cell lymphoma associated genes and/or clonal IG gene rearrangements. Our findings expand the current understanding of the molecular pathogenesis of this rare tumor and suggest the existence of a distinct subtype of primary histiocytic sarcoma characterized by NF1/PTPN11 alterations with predilection for the gastrointestinal tract.


Subject(s)
Histiocytic Sarcoma , Genomics , Histiocytic Sarcoma/diagnosis , Histiocytic Sarcoma/genetics , Homozygote , Humans , Mutation , Sequence Deletion
9.
Cell ; 165(2): 357-71, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058666

ABSTRACT

We report a mechanism through which the transcription machinery directly controls topoisomerase 1 (TOP1) activity to adjust DNA topology throughout the transcription cycle. By comparing TOP1 occupancy using chromatin immunoprecipitation sequencing (ChIP-seq) versus TOP1 activity using topoisomerase 1 sequencing (TOP1-seq), a method reported here to map catalytically engaged TOP1, TOP1 bound at promoters was discovered to become fully active only after pause-release. This transition coupled the phosphorylation of the carboxyl-terminal-domain (CTD) of RNA polymerase II (RNAPII) with stimulation of TOP1 above its basal rate, enhancing its processivity. TOP1 stimulation is strongly dependent on the kinase activity of BRD4, a protein that phosphorylates Ser2-CTD and regulates RNAPII pause-release. Thus the coordinated action of BRD4 and TOP1 overcame the torsional stress opposing transcription as RNAPII commenced elongation but preserved negative supercoiling that assists promoter melting at start sites. This nexus between transcription and DNA topology promises to elicit new strategies to intercept pathological gene expression.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , DNA/chemistry , DNA Topoisomerases, Type I/genetics , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic , RNA Polymerase II/chemistry , RNA Polymerase II/isolation & purification , Transcription Elongation, Genetic , Transcription Factors/isolation & purification , Transcription Initiation Site
10.
Am J Pathol ; 186(3): 701-15, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26774856

ABSTRACT

The transcription factor far upstream element binding protein (FBP) binds and activates the MYC promoter when far upstream element is via TFIIH helicase activity early in the transcription cycle. The fundamental biology and pathology of FBP are complex. In some tumors FBP seems pro-oncogenic, whereas in others it is a tumor suppressor. We generated an FBP knockout (Fubp1(-/-)) mouse to study FBP deficiency. FBP is embryo lethal from embryonic day 10.5 to birth. A spectrum of pathology is associated with FBP loss; besides cerebral hyperplasia and pulmonary hypoplasia, pale livers, hypoplastic spleen, thymus, and bone marrow, cardiac hypertrophy, placental distress, and small size were all indicative of anemia. Immunophenotyping of hematopoietic cells in wild-type versus knockout livers revealed irregular trilineage anemia, with deficits in colony formation. Despite normal numbers of hematopoietic stem cells, transplantation of Fubp1(-/-) hematopoietic stem cells into irradiated mice entirely failed to reconstitute hematopoiesis. In competitive transplantation assays against wild-type donor bone marrow, Fubp1(-/-) hematopoietic stem cells functioned only sporadically at a low level. Although cultures of wild-type mouse embryo fibroblasts set Myc levels precisely, Myc levels of mouse varied wildly between fibroblasts harvested from different Fubp1(-/-) embryos, suggesting that FBP contributes to Myc set point fixation. FBP helps to hold multiple physiologic processes to close tolerances, at least in part by constraining Myc expression.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Expression Regulation , Hematopoiesis/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , DNA-Binding Proteins/genetics , Female , Fibroblasts/metabolism , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Pregnancy , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/genetics
11.
Epigenetics Chromatin ; 5(1): 10, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22734930

ABSTRACT

BACKGROUND: The challenge in extracting genome-wide chromatin features from limiting clinical samples poses a significant hurdle in identification of regulatory marks that impact the physiological or pathological state. Current methods that identify nuclease accessible chromatin are reliant on large amounts of purified nuclei as starting material. This complicates analysis of trace clinical tissue samples that are often stored frozen. We have developed an alternative nuclease based procedure to bypass nuclear preparation to interrogate nuclease accessible regions in frozen tissue samples. RESULTS: Here we introduce a novel technique that specifically identifies Tissue Accessible Chromatin (TACh). The TACh method uses pulverized frozen tissue as starting material and employs one of the two robust endonucleases, Benzonase or Cyansase, which are fully active under a range of stringent conditions such as high levels of detergent and DTT. As a proof of principle we applied TACh to frozen mouse liver tissue. Combined with massive parallel sequencing TACh identifies accessible regions that are associated with euchromatic features and accessibility at transcriptional start sites correlates positively with levels of gene transcription. Accessible chromatin identified by TACh overlaps to a large extend with accessible chromatin identified by DNase I using nuclei purified from freshly isolated liver tissue as starting material. The similarities are most pronounced at highly accessible regions, whereas identification of less accessible regions tends to be more divergence between nucleases. Interestingly, we show that some of the differences between DNase I and Benzonase relate to their intrinsic sequence biases and accordingly accessibility of CpG islands is probed more efficiently using TACh. CONCLUSION: The TACh methodology identifies accessible chromatin derived from frozen tissue samples. We propose that this simple, robust approach can be applied across a broad range of clinically relevant samples to allow demarcation of regulatory elements of considerable prognostic significance.

12.
EMBO J ; 30(5): 846-58, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21285945

ABSTRACT

c-myc and p53 networks control proliferation, differentiation, and apoptosis and are responsive to, and cross-regulate a variety of stresses and metabolic and biosynthetic processes. At c-myc, the far upstream element binding protein (FBP) and FBP-interacting repressor (FIR) program transcription by looping to RNA polymerase II complexes engaged at the promoter. Another FBP partner, JTV1/AIMP2, a structural subunit of a multi-aminoacyl-tRNA synthetase (ARS) complex, has also been reported to stabilize p53 via an apparently independent mechanism. Here, we show that in response to oxidative stress, JTV1 dissociates from the ARS complex, translocates to the nucleus, associates with FBP and co-activates the transcription of a new FBP target, ubiquitin-specific peptidase 29 (USP29). A previously uncharacterized deubiquitinating enzyme, USP29 binds to, cleaves poly-ubiquitin chains from, and stabilizes p53. The accumulated p53 quickly induces apoptosis. Thus, FBP and JTV1 help to coordinate the molecular and cellular response to oxidative stress.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Endopeptidases/genetics , Oxidative Stress , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Amino Acyl-tRNA Synthetases/genetics , Apoptosis , Biomarkers/metabolism , Blotting, Western , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Endopeptidases/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Luciferases/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , Ubiquitin/metabolism , Ubiquitin-Specific Proteases
13.
Cancer Cell ; 18(6): 590-605, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21156283

ABSTRACT

Chromosome band 9p24 is frequently amplified in primary mediastinal B cell lymphoma (PMBL) and Hodgkin lymphoma (HL). To identify oncogenes in this amplicon, we screened an RNA interference library targeting amplicon genes and thereby identified JAK2 and the histone demethylase JMJD2C as essential genes in these lymphomas. Inhibition of JAK2 and JMJD2C cooperated in killing these lymphomas by decreasing tyrosine 41 phosphorylation and increasing lysine 9 trimethylation of histone H3, promoting heterochromatin formation. MYC, a major target of JAK2-mediated histone phosphorylation, was silenced after JAK2 and JMJD2C inhibition, with a corresponding increase in repressive chromatin. Hence, JAK2 and JMJD2C cooperatively remodel the PMBL and HL epigenome, offering a mechanistic rationale for the development of JAK2 and JMJD2C inhibitors in these diseases.


Subject(s)
Epigenesis, Genetic , Hodgkin Disease/genetics , Lymphoma, B-Cell/genetics , Mediastinal Neoplasms/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 9 , Histones/metabolism , Hodgkin Disease/pathology , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/physiology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Lymphoma, B-Cell/pathology , Phosphorylation
14.
Cell Oncol ; 32(1-2): 121-30, 2010.
Article in English | MEDLINE | ID: mdl-20208140

ABSTRACT

BACKGROUND: The general transcription factor II H (TFIIH), comprised of a core complex and an associated CAK-complex, functions in transcription, DNA repair and cell cycle control. Mutations of the two largest subunits, p89 (XPB) and p80 (XPD), cause the hereditary cancer-prone syndrome xeroderma pigmentosum. METHODS: The TFIIH subunit p89 was monitored during interphase and cell division by immunofluorescence staining, GFP-fusion constructs including deletions, live cell imaging and immuno-precipitations. RESULTS: Here we demonstrate that during cell division, from prophase until telophase, the TFIIH core subunit p89, but not other subunits of TFIIH, associates with the centrosomes and the adjacent parts of the mitotic spindle. With overall constant levels throughout mitosis, p89 re-localizes to the newly formed nuclei by the end of mitosis. Furthermore, p89 interacts with the centrosomal protein gamma-tubulin. Truncations of p89 result in an abnormal subcellular distribution during interphase and abolished centrosomal association during mitosis. CONCLUSIONS: Our observations suggest a so far unappreciated role for p89 in cell cycle regulation, and may be the structural basis for a long known, but hitherto unexplained interaction between p89 and tubulin.


Subject(s)
Centrosome/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Mitosis , DNA Helicases/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Protein Transport , Tubulin/metabolism
15.
Hepatology ; 50(4): 1121-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19637194

ABSTRACT

UNLABELLED: We identified the far upstream element binding protein 1 (FBP1), an activator of transcription of the proto-oncogene c-myc, in a functional yeast survival screen for tumor-related antiapoptotic proteins and demonstrated strong overexpression of FBP1 in human hepatocellular carcinoma (HCC). Knockdown of the protein in HCC cells resulted in increased sensitivity to apoptotic stimuli, reduced cell proliferation, and impaired tumor formation in a mouse xenograft transplantation model. Interestingly, analysis of gene regulation in these cells revealed that c-myc levels were not influenced by FBP1 in HCC cells. Instead, we identified the cell cycle inhibitor p21 as a direct target gene repressed by FBP1, and in addition, expression levels of the proapoptotic genes tumor necrosis factor alpha, tumor necrosis factor-related apoptosis-inducing ligand, Noxa, and Bik were elevated in the absence of FBP1. CONCLUSION: Our data establish FBP1 as an important oncoprotein overexpressed in HCC that induces tumor propagation through direct or indirect repression of cell cycle inhibitors and proapoptotic target genes.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Humans , Mice , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins , Signal Transduction/physiology , Transplantation, Heterologous
16.
Proc Natl Acad Sci U S A ; 105(47): 18296-301, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19015535

ABSTRACT

The far upstream element (FUSE) binding protein (FBP), a single-stranded nucleic acid binding protein, is recruited to the c-myc promoter after melting of FUSE by transcriptionally generated dynamic supercoils. Via interactions with TFIIH and FBP-interacting repressor (FIR), FBP modulates c-myc transcription. Here, we investigate the contributions of FBP's 4 K Homology (KH) domains to sequence selectivity. EMSA and missing contact point analysis revealed that FBP contacts 4 separate patches spanning a large segment of FUSE. A SELEX procedure using paired KH-domains defined the preferred subsequences for each KH domain. Unexpectedly, there was also a strong selection for the noncontacted residues between these subsequences, showing that the contact points must be optimally presented in a backbone that minimizes secondary structure. Strategic mutation of contact points defined in this study disabled FUSE activity in vivo. Because the biological specificity of FBP is tuned at several layers: (i) accessibility of the site; (ii) supercoil-driven melting; (iii) presentation of unhindered bases for recognition; and (iv) modular interaction of KH-domains with cognate bases, the FBP-FIR system and sequence-specific, single-strand DNA binding proteins in general are likely to prove versatile tools for adjusting gene expression.


Subject(s)
DNA Helicases/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , Consensus Sequence , DNA Helicases/genetics , DNA, Single-Stranded/chemistry , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Genes, myc , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Point Mutation , RNA-Binding Proteins , SELEX Aptamer Technique
17.
EMBO J ; 27(1): 277-89, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18059478

ABSTRACT

c-myc is essential for cell homeostasis and growth but lethal if improperly regulated. Transcription of this oncogene is governed by the counterbalancing forces of two proteins on TFIIH--the FUSE binding protein (FBP) and the FBP-interacting repressor (FIR). FBP and FIR recognize single-stranded DNA upstream of the P1 promoter, known as FUSE, and influence transcription by oppositely regulating TFIIH at the promoter site. Size exclusion chromatography coupled with light scattering reveals that an FIR dimer binds one molecule of single-stranded DNA. The crystal structure confirms that FIR binds FUSE as a dimer, and only the N-terminal RRM domain participates in nucleic acid recognition. Site-directed mutations of conserved residues in the first RRM domain reduce FIR's affinity for FUSE, while analogous mutations in the second RRM domain either destabilize the protein or have no effect on DNA binding. Oppositely oriented DNA on parallel binding sites of the FIR dimer results in spooling of a single strand of bound DNA, and suggests a mechanism for c-myc transcriptional control.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , DNA/chemistry , Dimerization , Drosophila Proteins/chemistry , Gene Expression Regulation/physiology , Humans , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Protein Binding/physiology , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , RNA Splicing Factors , RNA-Binding Proteins , Transcription Factor TFIIH/metabolism
18.
Mol Cell Biol ; 26(17): 6584-97, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914741

ABSTRACT

The three far-upstream element (FUSE) binding protein (FBP) family members have been ascribed different functions in gene regulation. They were therefore examined with various biochemical, molecular biological, and cell biological tests to evaluate whether their sequence differences reflect functional customization or neutral changes at unselected residues. Each FBP displayed a characteristic profile of intrinsic transcription activation and repression, binding with protein partners, and subcellular trafficking. Although some differences, such as weakened FBP3 nuclear localization, were predictable from primary sequence differences, the unexpected failure of FBP3 to bind the FBP-interacting repressor (FIR) was traced to seemingly conservative substitutions within a small patch of an N-terminal alpha-helix. The transactivation strength and the FIR-binding strength of the FBPs were in the opposite order. Despite their distinguishing features and differential activities, the FBPs traffic to shared subnuclear sites and regulate many common target genes, including c-myc. Though a variety of functions have been attributed to the FBPs, based upon their panel of shared and unique features, we propose that they constitute a molecular regulatory kit that tunes the expression of shared targets through a common mechanism.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , DNA/metabolism , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/metabolism , RNA Splicing Factors , RNA-Binding Proteins/chemistry , Repressor Proteins/metabolism , Subcellular Fractions , Trans-Activators/chemistry , Transcription Factors
19.
EMBO J ; 25(10): 2119-30, 2006 May 17.
Article in English | MEDLINE | ID: mdl-16628215

ABSTRACT

FarUpStream Element (FUSE) Binding Protein (FBP) binds the human c-myc FUSE in vitro only in single-stranded or supercoiled DNA. Because transcriptionally generated torsion melts FUSE in vitro even in linear DNA, and FBP/FBP Interacting Repressor (FIR) regulates transcription through TFIIH, these components have been speculated to be the mechanosensor (FUSE) and effectors (FBP/FIR) of a real-time mechanism controlling c-myc transcription. To ascertain whether the FUSE/FBP/FIR system operates according to this hypothesis in vivo, the flux of activators, repressors and chromatin remodeling complexes on the c-myc promoter was monitored throughout the serum-induced pulse of transcription. After transcription was switched on by conventional factors and chromatin regulators, FBP and FIR were recruited and established a dynamically remodeled loop with TFIIH at the P2 promoter. In XPB cells carrying mutant TFIIH, loop formation failed and the serum response was abnormal; RNAi depletion of FIR similarly disabled c-myc regulation. Engineering FUSE into episomal vectors predictably re-programmed metallothionein-promoter-driven reporter expression. The in vitro recruitment of FBP and FIR to dynamically stressed c-myc DNA paralleled the in vivo process.


Subject(s)
Gene Expression Regulation , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factor TFIIH/metabolism , Cell Line , Chromatin/chemistry , Chromatin/metabolism , DNA, Superhelical/chemistry , DNA, Superhelical/metabolism , Genes, Reporter , Humans , Metallothionein/genetics , Metallothionein/metabolism , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA Splicing Factors , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Transcription Factor TFIIH/genetics , Transcription, Genetic
20.
Mol Cells ; 20(2): 157-66, 2005 Oct 31.
Article in English | MEDLINE | ID: mdl-16267388

ABSTRACT

The c-myc proto-oncogene encodes a nuclear protein that is deregulated and/or mutated in most human cancers. Acting primarily as an activator and sometimes as a repressor, MYC protein controls the synthesis of up to 10-15% of genes. The key MYC targets contributing to oncogenesis are incompletely enumerated and it is not known whether pathology arises from the expression of physiologic targets at abnormal levels or from the pathologic response of new target genes that are not normally regulated by MYC. Regardless of which, available evidence indicates that the level of MYC expression is an important determinant of MYC biology. The c-myc promoter has architectural and functional features that contribute to uniform expression and help to prevent or mitigate conditions that might otherwise create noisy expression. Those features include the use of an expanded proximal promoter, the averaging of input from dozens of transcription factors, and real-time feedback using the supercoil-deformable Far UpStream Element (FUSE) as physical sensor of ongoing transcriptional activity, and the FUSE binding protein (FBP) as well as the FBP interacting repressor (FIR) as effectors to enforce normal transcription from the c-myc promoter.


Subject(s)
Genes, myc/physiology , Proto-Oncogene Proteins c-myc/biosynthesis , Animals , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...