Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Cell Proteomics ; 20: 100123, 2021.
Article in English | MEDLINE | ID: mdl-34298159

ABSTRACT

The mitogen-activated protein kinase pathway is one of the most frequently altered pathways in cancer. It is involved in the control of cell proliferation, invasion, and metabolism, and can cause resistance to therapy. A number of aggressive malignancies, including melanoma, colon cancer, and glioma, are driven by a constitutively activating missense mutation (V600E) in the v-Raf murine sarcoma viral oncogene homolog B (BRAF) component of the pathway. Mitogen-activated protein kinase kinase (MEK) inhibition is initially effective in targeting these cancers, but reflexive activation of mammalian target of rapamycin (mTOR) signaling contributes to frequent therapy resistance. We have previously demonstrated that combination treatment with the MEK inhibitor trametinib and the dual mammalian target of rapamycin complex 1/2 inhibitor TAK228 improves survival and decreases vascularization in a BRAFV600E mutant glioma model. To elucidate the mechanism of action of this combination therapy and understand the ensuing tumor response, we performed comprehensive unbiased proteomic and phosphoproteomic characterization of BRAFV600E mutant glioma xenografts after short-course treatment with trametinib and TAK228. We identified 13,313 proteins and 30,928 localized phosphosites, of which 12,526 proteins and 17,444 phosphosites were quantified across all samples (data available via ProteomeXchange; identifier PXD022329). We identified distinct response signatures for each monotherapy and combination therapy and validated that combination treatment inhibited activation of the mitogen-activated protein kinase and mTOR pathways. Combination therapy also increased apoptotic signaling, suppressed angiogenesis signaling, and broadly suppressed the activity of the cyclin-dependent kinases. In response to combination therapy, both epidermal growth factor receptor and class 1 histone deacetylase proteins were activated. This study reports a detailed (phospho)proteomic analysis of the response of BRAFV600E mutant glioma to combined MEK and mTOR pathway inhibition and identifies new targets for the development of rational combination therapies for BRAF-driven tumors.


Subject(s)
Benzoxazoles/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphoproteins/metabolism , Protein Kinase Inhibitors/therapeutic use , Pyridones/therapeutic use , Pyrimidines/therapeutic use , Pyrimidinones/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzoxazoles/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Female , Glioma/genetics , Glioma/metabolism , Humans , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Proteomics , Proto-Oncogene Proteins B-raf/genetics , Pyridones/pharmacology , Pyrimidines/pharmacology , Pyrimidinones/pharmacology
2.
Int J Mol Sci ; 22(8)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920124

ABSTRACT

Central nervous system tumor with BCL6-corepressor internal tandem duplication (CNS-BCOR ITD) is a malignant entity characterized by recurrent alterations in exon 15 encoding the essential binding domain for the polycomb repressive complex (PRC). In contrast to deletion or truncating mutations seen in other tumors, BCOR expression is upregulated in CNS-BCOR ITD, and a distinct oncogenic mechanism has been suggested. However, the effects of this change on the biology of neuroepithelial cells is poorly understood. In this study, we introduced either wildtype BCOR or BCOR-ITD into human and murine neural stem cells and analyzed them with quantitative RT-PCR and RNA-sequencing, as well as growth, clonogenicity, and invasion assays. In human cells, BCOR-ITD promoted derepression of PRC2-target genes compared to wildtype BCOR. A similar effect was found in clinical specimens from previous studies. However, no growth advantage was seen in the human neural stem cells expressing BCOR-ITD, and long-term models could not be established. In the murine cells, both wildtype BCOR and BCOR-ITD overexpression affected cellular differentiation and histone methylation, but only BCOR-ITD increased cellular growth, invasion, and migration. BCOR-ITD overexpression drives transcriptional changes, possibly due to altered PRC function, and contributes to the oncogenic transformation of neural precursors.


Subject(s)
Cell Proliferation/genetics , Central Nervous System Neoplasms/genetics , Polycomb-Group Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Animals , Cell Line, Tumor , Central Nervous System Neoplasms/pathology , Gene Duplication/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Tandem Repeat Sequences/genetics
3.
Cancer Cell ; 41(4): 660-677.e7, 2023 04 10.
Article in English | MEDLINE | ID: mdl-37001527

ABSTRACT

Pediatric solid and central nervous system tumors are the leading cause of cancer-related death among children. Identifying new targeted therapies necessitates the use of pediatric cancer models that faithfully recapitulate the patient's disease. However, the generation and characterization of pediatric cancer models has significantly lagged behind adult cancers, underscoring the urgent need to develop pediatric-focused cell line resources. Herein, we establish a single-site collection of 261 cell lines, including 224 pediatric cell lines representing 18 distinct extracranial and brain childhood tumor types. We subjected 182 cell lines to multi-omics analyses (DNA sequencing, RNA sequencing, DNA methylation), and in parallel performed pharmacological and genetic CRISPR-Cas9 loss-of-function screens to identify pediatric-specific treatment opportunities and biomarkers. Our work provides insight into specific pathway vulnerabilities in molecularly defined pediatric tumor classes and uncovers biomarker-linked therapeutic opportunities of clinical relevance. Cell line data and resources are provided in an open access portal.


Subject(s)
Brain Neoplasms , Child , Humans , Brain Neoplasms/pathology , Cell Line, Tumor
4.
J Clin Invest ; 132(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36377657

ABSTRACT

Pediatric high-grade gliomas (pHGGs) are aggressive diseases with poor outcomes. The diverse molecular heterogeneity in these rare tumors and inadequate tumor models have limited the development of effective therapies. In this issue of the JCI, Haase et al. produced a genetically engineered mouse model of H3.3-G34R-mutant pHGG to help identify vulnerabilities in DNA repair pathways. The authors designed a therapy that combined radiation with DNA damage response inhibitors to induce an adaptive immune response and extend survival. These findings suggest that combinations of small-molecule therapies with immunotherapies could drive a more durable response and improve mortality for patients with pHGG.


Subject(s)
Brain Neoplasms , Glioma , Mice , Animals , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/metabolism , Glioma/genetics , Glioma/therapy , Glioma/metabolism , Immune System/metabolism , Mutation
5.
Cell Death Dis ; 13(4): 410, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35484114

ABSTRACT

Atypical teratoid/rhabdoid tumors (AT/RT) are the most common malignant brain tumors of infancy and have a dismal 4-year event-free survival (EFS) of 37%. We have previously shown that mTOR activation contributes to AT/RT's aggressive growth and poor survival. Targeting the mTOR pathway with the dual mTORC1/2 inhibitor TAK-228 slows tumor growth and extends survival in mice bearing orthotopic xenografts. However, responses are primarily cytostatic with limited durability. The aim of this study is to understand the impact of mTOR inhibitors on AT/RT signaling pathways and design a rational combination therapy to drive a more durable response to this promising therapy. We performed RNASeq, gene expression studies, and protein analyses to identify pathways disrupted by TAK-228. We find that TAK-228 decreases the expression of the transcription factor NRF2 and compromises AT/RT cellular defenses against oxidative stress and apoptosis. The BH3 mimetic, Obatoclax, is a potent inducer of oxidative stress and apoptosis in AT/RT. These complementary mechanisms of action drive extensive synergies between TAK-228 and Obatoclax slowing AT/RT cell growth and inducing apoptosis and cell death. Combination therapy activates the integrative stress response as determined by increased expression of phosphorylated EIF2α, ATF4, and CHOP, and disrupts the protective NOXA.MCL-1.BIM axis, forcing stressed cells to undergo apoptosis. Combination therapy is well tolerated in mice bearing orthotopic xenografts of AT/RT, slows tumor growth, and extends median overall survival. This novel combination therapy could be added to standard upfront therapies or used as a salvage therapy for relapsed disease to improve outcomes in AT/RT.


Subject(s)
Rhabdoid Tumor , Animals , Humans , Indoles , Mechanistic Target of Rapamycin Complex 1 , Mice , Pyrroles/pharmacology , Pyrroles/therapeutic use , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/pathology , TOR Serine-Threonine Kinases
6.
Neuro Oncol ; 21(2): 252-263, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30239952

ABSTRACT

BACKGROUND: Pediatric low-grade glioma (pLGG) often initially responds to front-line therapies such as carboplatin, but more than 50% of treated tumors eventually progress and require additional therapy. With the discovery that pLGG often contains mammalian target of rapamycin (mTOR) activation, new treatment modalities and combinations are now possible for patients. The purpose of this study was to determine if carboplatin is synergistic with the mTOR complex 1 inhibitor everolimus in pLGG. METHODS: We treated 4 pLGG cell lines and 1 patient-derived xenograft line representing various pLGG genotypes, including neurofibromatosis type 1 loss, proto-oncogene B-Raf (BRAF)-KIAA1549 fusion, and BRAFV600E mutation, with carboplatin and/or everolimus and performed assays for growth, cell proliferation, and cell death. Immunohistochemistry as well as in vivo and in vitro metabolomics studies were also performed. RESULTS: Carboplatin synergized with everolimus in all of our 4 pLGG cell lines (combination index <1 at Fa 0.5). Combination therapy was superior at inhibiting tumor growth in vivo. Combination treatment increased levels of apoptosis as well as gamma-H2AX phosphorylation compared with either agent alone. Everolimus treatment suppressed the conversion of glutamine and glutamate into glutathione both in vitro and in vivo. Exogenous glutathione reversed the effects of carboplatin and everolimus. CONCLUSIONS: The combination of carboplatin and everolimus was effective at inducing cell death and slowing tumor growth in pLGG models. Everolimus decreased the amount of available glutathione inside the cell, preventing the detoxification of carboplatin and inducing increased DNA damage and apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , Drug Synergism , Everolimus/pharmacology , Glioma/drug therapy , Glutathione/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Adolescent , Animals , Apoptosis , Cell Proliferation , Glioma/metabolism , Glioma/pathology , Humans , Male , Mice , Mice, Nude , Proto-Oncogene Mas , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 25(19): 5925-5936, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31300448

ABSTRACT

PURPOSE: Atypical teratoid/rhabdoid tumors (AT/RT) are aggressive infantile brain tumors with poor survival. Recent advancements have highlighted significant molecular heterogeneity in AT/RT with an aggressive subgroup featuring overexpression of the MYC proto-oncogene. We perform the first comprehensive metabolic profiling of patient-derived AT/RT cell lines to identify therapeutic susceptibilities in high MYC-expressing AT/RT. EXPERIMENTAL DESIGN: Metabolites were extracted from AT/RT cell lines and separated in ultra-high performance liquid chromatography mass spectrometry. Glutamine metabolic inhibition with 6-diazo-5-oxo-L-norleucine (DON) was tested with growth and cell death assays and survival studies in orthotopic mouse models of AT/RT. Metabolic flux analysis was completed to identify combination therapies to act synergistically to improve survival in high MYC AT/RT. RESULTS: Unbiased metabolic profiling of AT/RT cell models identified a unique dependence of high MYC AT/RT on glutamine for survival. The glutamine analogue, DON, selectively targeted high MYC cell lines, slowing cell growth, inducing apoptosis, and extending survival in orthotopic mouse models of AT/RT. Metabolic flux experiments with isotopically labeled glutamine revealed DON inhibition of glutathione (GSH) synthesis. DON combined with carboplatin further slowed cell growth, induced apoptosis, and extended survival in orthotopic mouse models of high MYC AT/RT. CONCLUSIONS: Unbiased metabolic profiling of AT/RT identified susceptibility of high MYC AT/RT to glutamine metabolic inhibition with DON therapy. DON inhibited glutamine-dependent synthesis of GSH and synergized with carboplatin to extend survival in high MYC AT/RT. These findings can rapidly translate into new clinical trials to improve survival in high MYC AT/RT.


Subject(s)
Diazooxonorleucine/pharmacology , Glutamine/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Rhabdoid Tumor/metabolism , Teratoma/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Glutamine/metabolism , Humans , Metabolome/drug effects , Mice , Mice, Nude , Proto-Oncogene Mas , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/pathology , Teratoma/drug therapy , Teratoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Neuro Oncol ; 19(10): 1361-1371, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28582547

ABSTRACT

BACKGROUND: Atypical teratoid/rhabdoid tumors (AT/RTs) are deadly pediatric brain tumors driven by LIN28. Mammalian target of rapamycin (mTOR) is activated in many deadly, drug-resistant cancers and governs important cellular functions such as metabolism and survival. LIN28 regulates mTOR in normal cells. We therefore hypothesized that mTOR is activated downstream of LIN28 in AT/RT, and the brain-penetrating mTOR complex 1 and 2 (mTORC1/2) kinase inhibitor TAK228 would reduce AT/RT tumorigenicity. METHODS: Activation of mTOR in AT/RT was determined by measuring pS6 and pAKT (Ser473) by immunohistochemistry on tissue microarray of 18 primary AT/RT tumors. In vitro growth assays (BrdU and MTS), death assays (CC3, c-PARP by western blot), and survival curves of AT/RT orthotopic xenograft models were used to measure the efficacy of TAK228 alone and in combination with cisplatin. RESULTS: Lentiviral short hairpin RNA-mediated knockdown of LIN28A led to decreased mTOR activation. Primary human AT/RT had high levels of pS6 and pAKT (Ser473) in 21% and 87% of tumors by immunohistochemistry. TAK228 slowed cell growth, induced apoptosis in vitro, and nearly doubled median survival of orthotopic xenograft models of AT/RT. TAK228 combined with cisplatin synergistically slowed cell growth and enhanced cisplatin-induced apoptosis. Suppression of AKT sensitized cells to cisplatin-induced apoptosis and forced activation of AKT protected cells. Combined treatment with TAK228 and cisplatin significantly extended survival of orthotopic xenograft models of AT/RT compared with each drug alone. CONCLUSIONS: TAK228 has efficacy in AT/RT as a single agent and synergizes with conventional chemotherapies by sensitizing tumors to cisplatin-induced apoptosis. These results suggest TAK228 may be an effective new treatment for AT/RT.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzoxazoles/pharmacology , Cell Proliferation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Rhabdoid Tumor/drug therapy , Teratoma/drug therapy , Animals , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Humans , Mice , TOR Serine-Threonine Kinases/drug effects , Xenograft Model Antitumor Assays/methods
10.
Cancer Lett ; 400: 110-116, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28450157

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is an invasive and treatment-refractory pediatric brain tumor. Primary DIPG tumors harbor a number of mutations including alterations in PTEN, AKT, and PI3K and exhibit activation of mammalian Target of Rapamycin Complex 1 and 2 (mTORC1/2). mTORC1/2 regulate protein translation, cell growth, survival, invasion, and metabolism. Pharmacological inhibition of mTORC1 is minimally effective in DIPG. However, the activity of dual TORC kinase inhibitors has not been examined in this tumor type. Nanomolar levels of the mTORC1/2 inhibitor TAK228 reduced expression of p-AKTS473 and p-S6S240/244 and suppressed the growth of DIPG lines JHH-DIPG1, SF7761, and SU-DIPG-XIII. TAK228 induced apoptosis in DIPG cells and cooperated with radiation to further block proliferation and enhance apoptosis. TAK228 monotherapy inhibited the tumorigenicity of a murine orthotopic model of DIPG, more than doubling median survival (p = 0.0017) versus vehicle. We conclude that dual mTOR inhibition is a promising potential candidate for DIPG treatment.


Subject(s)
Benzoxazoles/pharmacology , Brain Stem Neoplasms/therapy , Chemoradiotherapy , Glioma/therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Brain Stem Neoplasms/enzymology , Brain Stem Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Dose-Response Relationship, Drug , Glioma/enzymology , Glioma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice, Inbred NOD , Mice, SCID , Multiprotein Complexes/metabolism , Neoplasm Invasiveness , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , TOR Serine-Threonine Kinases/metabolism , Time Factors , Xenograft Model Antitumor Assays
12.
Genes Dev ; 20(17): 2397-409, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951254

ABSTRACT

Taspase1 was identified as the threonine endopeptidase that cleaves mixed-lineage leukemia (MLL) for proper Hox gene expression in vitro. To investigate its functions in vivo, we generated Taspase1(-/-) mice. Taspase1 deficiency results in noncleavage (nc) of MLL and MLL2 and homeotic transformations. Remarkably, our in vivo studies uncover an unexpected role of Taspase1 in the cell cycle. Taspase1(-/-) animals are smaller in size. Taspase1(-/-) mouse embryonic fibroblasts (MEFs) exhibit impaired proliferation, and acute deletion of Taspase1 leads to a marked reduction of thymocytes. Taspase1 deficiency incurs down-regulation of Cyclin Es, As, and Bs and up-regulation of p16(Ink4a) . We show that MLL and MLL2 directly target E2Fs for Cyclin expression. The uncleaved precursor MLL displays a reduced histone H3 methyl transferase activity in vitro. Accordingly, chromatin immunoprecipitation assays demonstrate a markedly decreased histone H3 K4 trimethylation at Cyclin E1 and E2 genes in Taspase1(-/-) cells. Furthermore, MLL(nc/nc;2nc/nc) MEFs are also impaired in proliferation. Our data are consistent with a model in which precursor MLLs, activated by Taspase1, target to Cyclins through E2Fs to methylate histone H3 at K4, leading to activation. Lastly, Taspase1(-/-) cells are resistant to oncogenic transformation, and Taspase1 is overexpressed in many cancer cell lines. Thus, Taspase1 may serve as a target for cancer therapeutics.


Subject(s)
Cell Cycle/physiology , Endopeptidases/physiology , Myeloid-Lymphoid Leukemia Protein/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Endopeptidases/biosynthesis , Endopeptidases/deficiency , Endopeptidases/genetics , Hydrolysis , Mice , Mice, Knockout , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/genetics
13.
J Biol Chem ; 280(35): 30888-98, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-15998636

ABSTRACT

Arginine methylation can affect both nucleocytoplasmic transport and protein-protein interactions of RNA-binding proteins. These effects are seen in cells that lack the yeast hnRNP methyltransferase (HMT1), raising the question of whether effects on specific proteins are direct or indirect. The presence of multiple arginines in individual methylated proteins also raises the question of whether overall methylation or methylation of a subset of arginines affects protein function. We have used the yeast mRNA-binding protein Npl3 to address these questions in vivo. Matrix-assisted laser desorption/ionization Fourier transform mass spectrometry was used to identify 17 methylated arginines in Npl3 purified from yeast: whereas 10 Arg-Gly-Gly (RGG) tripeptides were exclusively dimethylated, variable levels of methylation were found for 5 RGG and 2 RG motif arginines. We constructed a set of Npl3 proteins in which subsets of the RGG arginines were mutated to lysine. Expression of these mutant proteins as the sole form of Npl3 specifically affected growth of a strain that requires Hmt1. Although decreased growth generally correlated with increased numbers of Arg-to-Lys mutations, lysine substitutions in the N terminus of the RGG domain showed more severe effects. Npl3 with all 15 RGG arginines mutated to lysine exited the nucleus independent of Hmt1, indicating a direct effect of methylation on Npl3 transport. These mutations also resulted in a decreased, methylation-independent interaction of Npl3 with transcription elongation factor Tho2 and inhibited Npl3 self-association. These results support a model in which arginine methylation facilitates Npl3 export directly by weakening contacts with nuclear proteins.


Subject(s)
Active Transport, Cell Nucleus/physiology , Arginine/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Humans , Lysine/metabolism , Methylation , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Interaction Mapping , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spectroscopy, Fourier Transform Infrared , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL