Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Rep Methods ; 2(11): 100340, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36452860

ABSTRACT

Tumor heterogeneity is a major challenge for oncology drug discovery and development. Understanding of the spatial tumor landscape is key to identifying new targets and impactful model systems. Here, we test the utility of spatial transcriptomics (ST) for oncology discovery by profiling 40 tissue sections and 80,024 capture spots across a diverse set of tissue types, sample formats, and RNA capture chemistries. We verify the accuracy and fidelity of ST by leveraging matched pathology analysis, which provides a ground truth for tissue section composition. We then use spatial data to demonstrate the capture of key tumor depth features, identifying hypoxia, necrosis, vasculature, and extracellular matrix variation. We also leverage spatial context to identify relative cell-type locations showing the anti-correlation of tumor and immune cells in syngeneic cancer models. Lastly, we demonstrate target identification approaches in clinical pancreatic adenocarcinoma samples, highlighting tumor intrinsic biomarkers and paracrine signaling.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Humans , Transcriptome/genetics , Pancreatic Neoplasms/diagnosis , Medical Oncology , Gene Expression Profiling , Biomarkers, Tumor/genetics
2.
Cell ; 170(3): 577-592.e10, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753431

ABSTRACT

Elucidation of the mutational landscape of human cancer has progressed rapidly and been accompanied by the development of therapeutics targeting mutant oncogenes. However, a comprehensive mapping of cancer dependencies has lagged behind and the discovery of therapeutic targets for counteracting tumor suppressor gene loss is needed. To identify vulnerabilities relevant to specific cancer subtypes, we conducted a large-scale RNAi screen in which viability effects of mRNA knockdown were assessed for 7,837 genes using an average of 20 shRNAs per gene in 398 cancer cell lines. We describe findings of this screen, outlining the classes of cancer dependency genes and their relationships to genetic, expression, and lineage features. In addition, we describe robust gene-interaction networks recapitulating both protein complexes and functional cooperation among complexes and pathways. This dataset along with a web portal is provided to the community to assist in the discovery and translation of new therapeutic approaches for cancer.


Subject(s)
Neoplasms/genetics , Neoplasms/pathology , RNA Interference , Cell Line, Tumor , Gene Library , Gene Regulatory Networks , Humans , Multiprotein Complexes/metabolism , Neoplasms/metabolism , Oncogenes , RNA, Small Interfering , Signal Transduction , Transcription Factors/metabolism
3.
Science ; 351(6278): 1208-13, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26912361

ABSTRACT

5-Methylthioadenosine phosphorylase (MTAP) is a key enzyme in the methionine salvage pathway. The MTAP gene is frequently deleted in human cancers because of its chromosomal proximity to the tumor suppressor gene CDKN2A. By interrogating data from a large-scale short hairpin RNA-mediated screen across 390 cancer cell line models, we found that the viability of MTAP-deficient cancer cells is impaired by depletion of the protein arginine methyltransferase PRMT5. MTAP-deleted cells accumulate the metabolite methylthioadenosine (MTA), which we found to inhibit PRMT5 methyltransferase activity. Deletion of MTAP in MTAP-proficient cells rendered them sensitive to PRMT5 depletion. Conversely, reconstitution of MTAP in an MTAP-deficient cell line rescued PRMT5 dependence. Thus, MTA accumulation in MTAP-deleted cancers creates a hypomorphic PRMT5 state that is selectively sensitized toward further PRMT5 inhibition. Inhibitors of PRMT5 that leverage this dysregulated metabolic state merit further investigation as a potential therapy for MTAP/CDKN2A-deleted tumors.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Methionine/metabolism , Neoplasms/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Cell Line, Tumor , Cell Survival , Cyclin-Dependent Kinase Inhibitor p16/genetics , Deoxyadenosines/metabolism , Gene Deletion , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Protein-Arginine N-Methyltransferases/genetics , Purine-Nucleoside Phosphorylase/genetics , RNA, Small Interfering/genetics , Thionucleosides/metabolism
4.
Proc Natl Acad Sci U S A ; 111(46): 16532-7, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25378704

ABSTRACT

We have developed a screening platform for the isolation of genetic entities involved in metastatic reactivation. Retroviral libraries of cDNAs from fully metastatic breast-cancer cells or pooled microRNAs were transduced into breast-cancer cells that become dormant upon infiltrating the lung. Upon inoculation in the tail vein of mice, the cells that had acquired the ability to undergo reactivation generated metastatic lesions. Integrated retroviral vectors were recovered from these lesions, sequenced, and subjected to a second round of validation. By using this strategy, we isolated canonical genes and microRNAs that mediate metastatic reactivation in the lung. To identify genes that oppose reactivation, we screened an expression library encoding shRNAs, and we identified target genes that encode potential enforcers of dormancy. Our screening strategy enables the identification and rapid biological validation of single genetic entities that are necessary to maintain dormancy or to induce reactivation. This technology should facilitate the elucidation of the molecular underpinnings of these processes.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Neoplasm , High-Throughput Screening Assays/methods , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/genetics , Animals , Carcinoma/secondary , Cell Line, Tumor , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Female , Gene Library , Genetic Vectors/genetics , Genetic Vectors/isolation & purification , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/isolation & purification , Neoplasm Metastasis/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasm Transplantation , Proviruses/genetics , Proviruses/isolation & purification , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , RNA, Small Interfering/genetics , Retroviridae/genetics , Retroviridae/isolation & purification , Transduction, Genetic , Virus Integration
5.
Sci Signal ; 7(352): ra111, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25406379

ABSTRACT

The posttranscriptional control of gene expression by microRNAs (miRNAs) is highly redundant, and compensatory effects limit the consequences of the inactivation of individual miRNAs. This implies that only a few miRNAs can function as effective tumor suppressors. It is also the basis of our strategy to define functionally relevant miRNA target genes that are not under redundant control by other miRNAs. We identified a functionally interconnected group of miRNAs that exhibited a reduced abundance in leukemia cells from patients with T cell acute lymphoblastic leukemia (T-ALL). To pinpoint relevant target genes, we applied a machine learning approach to eliminate genes that were subject to redundant miRNA-mediated control and to identify those genes that were exclusively targeted by tumor-suppressive miRNAs. This strategy revealed the convergence of a small group of tumor suppressor miRNAs on the Myb oncogene, as well as their effects on HBP1, which encodes a transcription factor. The expression of both genes was increased in T-ALL patient samples, and each gene promoted the progression of T-ALL in mice. Hence, our systematic analysis of tumor suppressor miRNA action identified a widespread mechanism of oncogene activation in T-ALL.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Genes, myb/genetics , MicroRNAs/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adoptive Transfer , Animals , Artificial Intelligence , Hematopoietic Stem Cell Transplantation , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Mice , MicroRNAs/metabolism , Models, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism
6.
Nature ; 513(7516): 65-70, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25079319

ABSTRACT

The translational control of oncoprotein expression is implicated in many cancers. Here we report an eIF4A RNA helicase-dependent mechanism of translational control that contributes to oncogenesis and underlies the anticancer effects of silvestrol and related compounds. For example, eIF4A promotes T-cell acute lymphoblastic leukaemia development in vivo and is required for leukaemia maintenance. Accordingly, inhibition of eIF4A with silvestrol has powerful therapeutic effects against murine and human leukaemic cells in vitro and in vivo. We use transcriptome-scale ribosome footprinting to identify the hallmarks of eIF4A-dependent transcripts. These include 5' untranslated region (UTR) sequences such as the 12-nucleotide guanine quartet (CGG)4 motif that can form RNA G-quadruplex structures. Notably, among the most eIF4A-dependent and silvestrol-sensitive transcripts are a number of oncogenes, superenhancer-associated transcription factors, and epigenetic regulators. Hence, the 5' UTRs of select cancer genes harbour a targetable requirement for the eIF4A RNA helicase.


Subject(s)
5' Untranslated Regions/genetics , Eukaryotic Initiation Factor-4A/metabolism , G-Quadruplexes , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Biosynthesis , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Base Sequence , Cell Line, Tumor , Epigenesis, Genetic , Female , Humans , Mice , Mice, Inbred C57BL , Nucleotide Motifs , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Biosynthesis/drug effects , Ribosomes/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Triterpenes/pharmacology
7.
Nat Methods ; 10(8): 768-73, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23817070

ABSTRACT

We report a technique to selectively and continuously label the proteomes of individual cell types in coculture, named cell type-specific labeling using amino acid precursors (CTAP). Through transgenic expression of exogenous amino acid biosynthesis enzymes, vertebrate cells overcome their dependence on supplemented essential amino acids and can be selectively labeled through metabolic incorporation of amino acids produced from heavy isotope-labeled precursors. When testing CTAP in several human and mouse cell lines, we could differentially label the proteomes of distinct cell populations in coculture and determine the relative expression of proteins by quantitative mass spectrometry. In addition, using CTAP we identified the cell of origin of extracellular proteins secreted from cells in coculture. We believe that this method, which allows linking of proteins to their cell source, will be useful in studies of cell-cell communication and potentially for discovery of biomarkers.


Subject(s)
Lysine/metabolism , Proteome/biosynthesis , Proteomics/methods , Animals , Base Sequence , Cell Line , Coculture Techniques/methods , Humans , Isotope Labeling/methods , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Organisms, Genetically Modified , Proteome/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Sequence Analysis, DNA , Tandem Mass Spectrometry
8.
Cell ; 147(3): 554-64, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036564

ABSTRACT

Insights into cancer genetics can lead to therapeutic opportunities. By cross-referencing chromosomal changes with an unbiased genetic screen we identify the ephrin receptor A7 (EPHA7) as a tumor suppressor in follicular lymphoma (FL). EPHA7 is a target of 6q deletions and inactivated in 72% of FLs. Knockdown of EPHA7 drives lymphoma development in a murine FL model. In analogy to its physiological function in brain development, a soluble splice variant of EPHA7 (EPHA7(TR)) interferes with another Eph-receptor and blocks oncogenic signals in lymphoma cells. Consistent with this drug-like activity, administration of the purified EPHA7(TR) protein produces antitumor effects against xenografted human lymphomas. Further, by fusing EPHA7(TR) to the anti-CD20 antibody (rituximab) we can directly target this tumor suppressor to lymphomas in vivo. Our study attests to the power of combining descriptive tumor genomics with functional screens and reveals EPHA7(TR) as tumor suppressor with immediate therapeutic potential.


Subject(s)
Genes, Tumor Suppressor , Lymphoma, Follicular/metabolism , Receptor, EphA7/metabolism , Animals , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Cell Line, Tumor , Chromosomes, Human, Pair 6 , Genomics , Humans , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/genetics , Male , Mice , Neoplasm Transplantation , RNA Interference , Rituximab , Transplantation, Heterologous
9.
Cell Cycle ; 10(17): 2845-9, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21857153

ABSTRACT

Individual microRNAs (miRNAs) have been implicated as oncogenes in experimental cancer models and their expression may affect clinical outcomes. To gain a more comprehensive view of miRNA action in leukemia, we analyzed miRNA expression patters in T-cell leukemia ALL (T-ALL) and cross-referenced the results with an unbiased genetic screen and computational analyses.1 We found that multiple microRNAs contribute to leukmogenesis and act as multi-targeted regulators of several tumor suppressor genes. The oncomirs form a network of overlapping and partially redundant interactions that stabilize the malignant phenotype though coordinate repression of cellular failsafe programs. The emerging network pattern of oncomir action is distinct from the notion of single oncogenic 'driver' mutation. We will discuss experimental, diagnostic and therapeutic implications of this concept of miRNA action in cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , MicroRNAs/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Disease Models, Animal , F-Box Proteins/genetics , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Gene Knockdown Techniques , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Nat Genet ; 43(7): 673-8, 2011 Jun 05.
Article in English | MEDLINE | ID: mdl-21642990

ABSTRACT

The importance of individual microRNAs (miRNAs) has been established in specific cancers. However, a comprehensive analysis of the contribution of miRNAs to the pathogenesis of any specific cancer is lacking. Here we show that in T-cell acute lymphoblastic leukemia (T-ALL), a small set of miRNAs is responsible for the cooperative suppression of several tumor suppressor genes. Cross-comparison of miRNA expression profiles in human T-ALL with the results of an unbiased miRNA library screen allowed us to identify five miRNAs (miR-19b, miR-20a, miR-26a, miR-92 and miR-223) that are capable of promoting T-ALL development in a mouse model and which account for the majority of miRNA expression in human T-ALL. Moreover, these miRNAs produce overlapping and cooperative effects on tumor suppressor genes implicated in the pathogenesis of T-ALL, including IKAROS (also known as IKZF1), PTEN, BIM, PHF6, NF1 and FBXW7. Thus, a comprehensive and unbiased analysis of miRNA action in T-ALL reveals a striking pattern of miRNA-tumor suppressor gene interactions in this cancer.


Subject(s)
Gene Regulatory Networks , Genes, Tumor Suppressor , MicroRNAs/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Animals , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Child , Child, Preschool , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Infant , Luciferases/metabolism , Male , Mice , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Young Adult
11.
Dis Model Mech ; 3(11-12): 701-4, 2010.
Article in English | MEDLINE | ID: mdl-20876355

ABSTRACT

Genetically and pathologically accurate mouse models of leukemia and lymphoma have been developed in recent years. Adoptive transfer of genetically modified hematopoietic progenitor cells enables rapid and highly controlled gain- and loss-of-function studies for these types of cancer. In this Commentary, we discuss how these highly versatile experimental approaches can be used as biological filters to pinpoint transformation-relevant activities from complex cancer genome data. We anticipate that the functional identification of genetic 'drivers' using mouse models of leukemia and lymphoma will facilitate the development of molecular diagnostics and mechanism-based therapies for patients that suffer from these diseases.


Subject(s)
Databases, Genetic , Disease Models, Animal , Genome/genetics , Hematologic Neoplasms/genetics , Translational Research, Biomedical , Animals , Humans , Mice
12.
Cell Cycle ; 9(11): 2080-4, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20505335

ABSTRACT

We recently identified miR-19 as the critical activity for leukemogenesis within the oncogenic 17~92 cluster of microRNAs. ( 1) This finding prompted us to test an unbiased method for pinpointing those miR-19 targets may be key to its oncogenic action. Specifically, we used a large-scale short hairpin RNA screen to identify those miR-19 target genes, whose knockdown could reproduce miR-19's effects on lymphocyte transformation. In this way, we found that miR-19 produces a coordinate clampdown on multiple negative regulators of PI3K-related survival signals. These findings have implications for the therapy of miR-19 expressing tumors. They also validate a new strategy for the unbiased identification of functionally important microRNA target genes. Using the example of miR-19 in leukemia, we will discuss some possibilities and limitations of this new approach.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia/genetics , MicroRNAs/metabolism , Gene Knockdown Techniques , Humans , Leukemia/therapy , MicroRNAs/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , RNA Interference
13.
Nat Cell Biol ; 12(4): 372-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20190740

ABSTRACT

MicroRNAs (miRNAs) have emerged as novel cancer genes. In particular, the miR-17-92 cluster, containing six individual miRNAs, is highly expressed in haematopoietic cancers and promotes lymphomagenesis in vivo. Clinical use of these findings hinges on isolating the oncogenic activity within the 17-92 cluster and defining its relevant target genes. Here we show that miR-19 is sufficient to promote leukaemogenesis in Notch1-induced T-cell acute lymphoblastic leukaemia (T-ALL) in vivo. In concord with the pathogenic importance of this interaction in T-ALL, we report a novel translocation that targets the 17-92 cluster and coincides with a second rearrangement that activates Notch1. To identify the miR-19 targets responsible for its oncogenic action, we conducted a large-scale short hairpin RNA screen for genes whose knockdown can phenocopy miR-19. Strikingly, the results of this screen were enriched for miR-19 target genes, and include Bim (Bcl2L11), AMP-activated kinase (Prkaa1) and the phosphatases Pten and PP2A (Ppp2r5e). Hence, an unbiased, functional genomics approach reveals a coordinate clampdown on several regulators of phosphatidylinositol-3-OH kinase-related survival signals by the leukaemogenic miR-19.


Subject(s)
Gene Expression Regulation, Leukemic , MicroRNAs/metabolism , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA Interference , Receptor, Notch1/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Knockdown Techniques , Gene Rearrangement, T-Lymphocyte , Genome-Wide Association Study , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/metabolism , Signal Transduction/genetics , Time Factors , Transduction, Genetic , Translocation, Genetic
14.
PLoS One ; 4(1): e4268, 2009.
Article in English | MEDLINE | ID: mdl-19172185

ABSTRACT

The Transforming Growth Factor (TGF) beta signalling family includes morphogens, such as Nodal and Activin, with important functions in vertebrate development. The concentration of the morphogen is critical for fate decisions in the responding cells. Smad2 and Smad3 are effectors of the Nodal/Activin branch of TGFbeta signalling: they are activated by receptors, enter the nucleus and directly transcribe target genes. However, there have been no studies correlating levels of Smad2/3 activation with expression patterns of endogenous target genes in a developmental context over time. We used mouse Embryonic Stem (ES) cells to create a system whereby levels of activated Smad2/3 can be manipulated by an inducible constitutively active receptor (Alk4*) and an inhibitor (SB-431542) that blocks specifically Smad2/3 activation. The transcriptional responses were analysed by microarrays at different time points during activation and repression. We identified several genes that follow faithfully and reproducibly the Smad2/3 activation profile. Twenty-seven of these were novel and expressed in the early embryo downstream of Smad2/3 signalling. As they responded to Smad2/3 activation in the absence of protein synthesis, they were considered direct. These immediate responsive genes included negative intracellular feedback factors, like SnoN and I-Smad7, which inhibit the transcriptional activity of Smad2/3. However, their activation did not lead to subsequent repression of target genes over time, suggesting that this type of feedback is inefficient in ES cells or it is counteracted by mechanisms such as ubiquitin-mediated degradation by Arkadia. Here we present an ES cell system along with a database containing the expression profile of thousands of genes downstream of Smad2/3 activation patterns, in the presence or absence of protein synthesis. Furthermore, we identify primary target genes that follow proportionately and with high sensitivity changes in Smad2/3 levels over 15-30 hours. The above system and resource provide tools to study morphogen function in development.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Activin Receptors, Type I/metabolism , Animals , Benzamides/pharmacology , Dioxoles/pharmacology , Flow Cytometry , Mice , Models, Biological , Nodal Protein/metabolism , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases
15.
Cell Cycle ; 7(18): 2791-4, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18787409

ABSTRACT

Our recent findings on Rheb and eIF4E address key questions of translational control in cancer and have implications for tumor therapy.(1) Briefly, we find that Rheb a proximal activator of mTORC1 and protein translation can cooperate with c-Myc in tumorigenesis in vivo in a manner resembling Akt or the oncogenic eIF4E translation initiation factor. Rheb is highly expressed in some human lymphomas as well as other cancers and likely contributes to malignancies in different tissues.(2) The cancer-relevant activities emanating from increased Rheb depend on activation of mTORC1 and are sensitive to rapamycin. Moreover, farnesyltransferase inhibitors (FTIs) can directly block Rheb activity and this is responsible for the therapeutic effect of these drugs in certain tumors. We will discuss here how translational control mechanisms contribute to oncogenesis and speculate on the potential and limitations of targeting these co-operating oncogenic events for therapy.


Subject(s)
Neoplasms/therapy , Protein Biosynthesis , Animals , Chickens , Eukaryotic Initiation Factor-4E/metabolism , Humans , Monomeric GTP-Binding Proteins/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases
16.
Genes Dev ; 22(16): 2178-88, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18708578

ABSTRACT

The AKT-mTOR pathway harbors several known and putative oncogenes and tumor suppressors. In a phenotypic screen for lymphomagenesis, we tested candidate genes acting upstream of and downstream from mTOR in vivo. We find that Rheb, a proximal activator of mTORC1, can produce rapid development of aggressive and drug-resistant lymphomas. Rheb causes mTORC1-dependent effects on apoptosis, senescence, and treatment responses that resemble those of Akt. Moreover, Rheb activity toward mTORC1 requires farnesylation and is readily blocked by a pharmacological inhibitor of farnesyltransferase (FTI). In Pten-deficient tumor cells, inhibition of Rheb by FTI is responsible for the drug's anti-tumor effects, such that a farnesylation-independent mutant of Rheb renders these tumors resistant to FTI therapy. Notably, RHEB is highly expressed in some human lymphomas, resulting in mTORC1 activation and increased sensitivity to rapamycin and FTI. Downstream from mTOR, we examined translation initiation factors that have been implicated in transformation in vitro. Of these, only eIF4E was able to enhance lymphomagenesis in vivo. In summary, the Rheb GTPase is an oncogenic activity upstream of mTORC1 and eIF4E and a direct therapeutic target of farnesyltransferase inhibitors in cancer.


Subject(s)
Cell Transformation, Neoplastic/pathology , Eukaryotic Initiation Factor-4E/metabolism , Farnesyltranstransferase/antagonists & inhibitors , Lymphoma/pathology , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Cells, Cultured , Cellular Senescence , Doxorubicin/pharmacology , Farnesyltranstransferase/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Dosage , Humans , Immunophenotyping , Immunosuppressive Agents/pharmacology , Lymphoma/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes , Neuropeptides/metabolism , PTEN Phosphohydrolase/physiology , Phosphorylation , Piperidines/pharmacology , Proteins , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/physiology , Pyridines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ras Homolog Enriched in Brain Protein , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tumor Suppressor Protein p53/physiology
17.
J Biol Chem ; 282(28): 20492-501, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17510063

ABSTRACT

Transforming growth factor-beta (TGF-beta) signaling is controlled by a variety of regulators that target either signaling receptors or activated Smad complexes. Among the negative regulators, Smad7 antagonizes TGF-beta signaling mainly through targeting the signaling receptors, whereas SnoN and c-Ski repress signaling at the transcriptional level through inactivation of Smad complexes. We previously found that Arkadia is a positive regulator of TGF-beta signaling that induces ubiquitin-dependent degradation of Smad7 through its C-terminal RING domain. We report here that Arkadia induces degradation of SnoN and c-Ski in addition to Smad7. Arkadia interacts with SnoN and c-Ski in their free forms as well as in the forms bound to Smad proteins, and constitutively down-regulates levels of their expression. Arkadia thus appears to effectively enhance TGF-beta signaling through simultaneous down-regulation of two distinct types of negative regulators, Smad7 and SnoN/c-Ski, and may play an important role in determining the intensity of TGF-beta family signaling in target cells.


Subject(s)
DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Ubiquitin/metabolism , Animals , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Down-Regulation/physiology , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Protein Binding/drug effects , Protein Binding/physiology , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Transforming Growth Factor beta/pharmacology , Ubiquitin/genetics , Ubiquitin-Protein Ligases
18.
PLoS Biol ; 5(3): e67, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17341133

ABSTRACT

Regulation of transforming growth factor-beta (TGF-beta) signaling is critical in vertebrate development, as several members of the TGF-beta family have been shown to act as morphogens, controlling a variety of cell fate decisions depending on concentration. Little is known about the role of intracellular regulation of the TGF-beta pathway in development. E3 ubiquitin ligases target specific protein substrates for proteasome-mediated degradation, and several are implicated in signaling. We have shown that Arkadia, a nuclear RING-domain E3 ubiquitin ligase, is essential for a subset of Nodal functions in the embryo, but the molecular mechanism of its action in embryonic cells had not been addressed. Here, we find that Arkadia facilitates Nodal signaling broadly in the embryo, and that it is indispensable for cell fates that depend on maximum signaling. Loss of Arkadia in embryonic cells causes nuclear accumulation of phospho-Smad2/3 (P-Smad2/3), the effectors of Nodal signaling; however, these must be repressed or hypoactive as the expression of their direct target genes is reduced or lost. Molecular and functional analysis shows that Arkadia interacts with and ubiquitinates P-Smad2/3 causing their degradation, and that this is via the same domains required for enhancing their activity. Consistent with this dual function, introduction of Arkadia in homozygous null (-/-) embryonic stem cells activates the accumulated and hypoactive P-Smad2/3 at the expense of their abundance. Arkadia-/- cells, like Smad2-/- cells, cannot form foregut and prechordal plate in chimeras, confirming this functional interaction in vivo. As Arkadia overexpression never represses, and in some cells enhances signaling, the degradation of P-Smad2/3 by Arkadia cannot occur prior to their activation in the nucleus. Therefore, Arkadia provides a mechanism for signaling termination at the end of the cascade by coupling degradation of P-Smad2/3 with the activation of target gene transcription. This mechanism can account for achieving efficient and maximum Nodal signaling during embryogenesis and for rapid resetting of target gene promoters allowing cells to respond to dynamic changes in extracellular signals.


Subject(s)
Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin/physiology , Animals , Base Sequence , Chimera , DNA Primers , Mice , Ubiquitin-Protein Ligases
19.
Exp Cell Res ; 294(2): 335-44, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15023524

ABSTRACT

In this paper, we describe the characterization of DEF6, a novel PH-DH-like protein related to SWAP-70 that functions as an upstream activator of Rho GTPases. In NIH 3T3 cells, stimulation of the PI 3-kinase signaling pathway with either H2O2 or platelet-derived growth factor (PDGF) resulted in the translocation of an overexpressed DEF6-GFP fusion protein to the cell membrane and induced the formation of filopodia and lamellipodia. In contrast to full-length DEF6, expression of the DH-like (DHL) domain as a GFP fusion protein potently induced actin polymerization, including stress fiber formation in COS-7 cells, in the absence of PI 3-kinase signaling, indicating that it was constitutively active. The GTP-loading of Cdc42 was strongly enhanced in NIH 3T3 cells expressing the DH domain while filopodia formation, membrane ruffling, and stress fiber formation could be inhibited by the co-expression of the DH domain with dominant negative mutants of either N17Rac1, N17Cdc42, or N19RhoA, respectively. This indicated that DEF6 acts upstream of the Rho GTPases resulting in the activation of the Cdc42, Rac1, and RhoA signaling pathways. In vitro, DEF6 specifically interacted with Rac1, Rac2, Cdc42, and RhoA, suggesting a direct role for DEF6 in the activation of Rho GTPases. The ability of DEF6 to both stimulate actin polymerization and bind to filamentous actin suggests a role for DEF6 in regulating cell shape, polarity, and movement.


Subject(s)
Actin Cytoskeleton/metabolism , DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , Nuclear Proteins/metabolism , Pseudopodia/metabolism , Signal Transduction/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , COS Cells , Cell Movement/drug effects , Cell Movement/genetics , Cell Polarity/drug effects , Cell Polarity/genetics , Cell Size/drug effects , Cell Size/genetics , DNA, Complementary/analysis , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Enzyme Activators/isolation & purification , Enzyme Activators/metabolism , HeLa Cells , Humans , Mice , Minor Histocompatibility Antigens , Molecular Sequence Data , Mutation/genetics , NIH 3T3 Cells , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Pseudopodia/drug effects , Pseudopodia/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/genetics , Stress Fibers/metabolism , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...