Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Cell ; 144(5): 703-18, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376233

ABSTRACT

Among breast cancers, triple-negative breast cancer (TNBC) is the most poorly understood and is refractory to current targeted therapies. Using a genetic screen, we identify the PTPN12 tyrosine phosphatase as a tumor suppressor in TNBC. PTPN12 potently suppresses mammary epithelial cell proliferation and transformation. PTPN12 is frequently compromised in human TNBCs, and we identify an upstream tumor-suppressor network that posttranscriptionally controls PTPN12. PTPN12 suppresses transformation by interacting with and inhibiting multiple oncogenic tyrosine kinases, including HER2 and EGFR. The tumorigenic and metastatic potential of PTPN12-deficient TNBC cells is severely impaired upon restoration of PTPN12 function or combined inhibition of PTPN12-regulated tyrosine kinases, suggesting that TNBCs are dependent on the proto-oncogenic tyrosine kinases constrained by PTPN12. Collectively, these data identify PTPN12 as a commonly inactivated tumor suppressor and provide a rationale for combinatorially targeting proto-oncogenic tyrosine kinases in TNBC and other cancers based on their profile of tyrosine-phosphatase activity.


Subject(s)
Breast Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Transformation, Neoplastic , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , MicroRNAs/metabolism , Mutation , Neoplasm Metastasis , Protein Processing, Post-Translational
2.
Proc Natl Acad Sci U S A ; 108(9): 3665-70, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21307310

ABSTRACT

The discovery of RNAi has revolutionized loss-of-function genetic studies in mammalian systems. However, significant challenges still remain to fully exploit RNAi for mammalian genetics. For instance, genetic screens and in vivo studies could be broadly improved by methods that allow inducible and uniform gene expression control. To achieve this, we built the lentiviral pINDUCER series of expression vehicles for inducible RNAi in vivo. Using a multicistronic design, pINDUCER vehicles enable tracking of viral transduction and shRNA or cDNA induction in a broad spectrum of mammalian cell types in vivo. They achieve this uniform temporal, dose-dependent, and reversible control of gene expression across heterogenous cell populations via fluorescence-based quantification of reverse tet-transactivator expression. This feature allows isolation of cell populations that exhibit a potent, inducible target knockdown in vitro and in vivo that can be used in human xenotransplantation models to examine cancer drug targets.


Subject(s)
Genetic Techniques , Genetic Vectors/genetics , Lentivirus/genetics , RNA Interference , Animals , Breast Neoplasms/pathology , Cell Line , DNA, Complementary/genetics , Diagnostic Imaging , Female , Gene Expression , Humans , Luminescence , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , RNA, Small Interfering/metabolism , Reproducibility of Results , Xenograft Model Antitumor Assays
3.
Cancer Discov ; 14(9): 1699-1716, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39193992

ABSTRACT

Upregulation of MYC is a hallmark of cancer, wherein MYC drives oncogenic gene expression and elevates total RNA synthesis across cancer cell transcriptomes. Although this transcriptional anabolism fuels cancer growth and survival, the consequences and metabolic stresses induced by excess cellular RNA are poorly understood. Herein, we discover that RNA degradation and downstream ribonucleotide catabolism is a novel mechanism of MYC-induced cancer cell death. Combining genetics and metabolomics, we find that MYC increases RNA decay through the cytoplasmic exosome, resulting in the accumulation of cytotoxic RNA catabolites and reactive oxygen species. Notably, tumor-derived exosome mutations abrogate MYC-induced cell death, suggesting excess RNA decay may be toxic to human cancers. In agreement, purine salvage acts as a compensatory pathway that mitigates MYC-induced ribonucleotide catabolism, and inhibitors of purine salvage impair MYC+ tumor progression. Together, these data suggest that MYC-induced RNA decay is an oncogenic stress that can be exploited therapeutically. Significance: MYC is the most common oncogenic driver of poor-prognosis cancers but has been recalcitrant to therapeutic inhibition. We discovered a new vulnerability in MYC+ cancer where MYC induces cell death through excess RNA decay. Therapeutics that exacerbate downstream ribonucleotide catabolism provide a therapeutically tractable approach to TNBC (Triple-negative Breast Cancer) and other MYC-driven cancers.


Subject(s)
Breast Neoplasms , Proto-Oncogene Proteins c-myc , RNA Stability , Ribonucleotides , Humans , Female , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Ribonucleotides/pharmacology , Cell Line, Tumor , Mice , Gene Expression Regulation, Neoplastic , Animals
4.
Int J Oncol ; 35(2): 375-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578752

ABSTRACT

The development of ovarian cancer, unlike that of most human tumors, is rarely dependent upon the mutually exclusive loss of RB and p16 cell cycle proteins. RB+/p16+ ovarian cancer cell lines are, however, insensitive to the growth-suppressive effects of ectopically expressed p16 protein, which suggests that they harbor as yet unidentified defects that compromise cell cycle regulation in late G1/S. In the current study, we used Western blotting to analyze cyclin E protein expression in a panel of normal and tumor ovarian tissues and ovarian cancer cell lines (including the p16-insensitive RB+/p16+ ovarian cancer cell line, NIH-OVCAR-3). Both the NIH-OVCAR-3 cell line and 70% of RB+/p16+ ovarian tumors showed abnormally elevated levels of the full-length cyclin E protein (EL1) in addition to several low molecular weight (LMW) isoforms of cyclin E. Using small interference RNA (siRNA), we have inhibited the synthesis of cyclin EL1 protein by approximately 80% and eliminated the LMW isoforms in NIH-OVCAR-3 ovarian cancer cells. Associated with the down-regulation of cyclin E expression, we observed both a marked shift in RB protein expression to the active, hypophosphorylated state and barely detectable expression of cyclin A (which is usually expressed upon entry into S-phase). Consistent with the protein expression data, cell cycle distribution analysis indicated that the NIH-OVCAR-3 cells had undergone a marked accumulation in G1 phase of the cell cycle. These data indicate the therapeutic potential of targeted RNA interference in the treatment of ovarian cancer patients whose tumors overexpress cyclin E protein.


Subject(s)
Cyclin E/physiology , G1 Phase , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , S Phase , Cell Line, Tumor , Cyclin E/antagonists & inhibitors , Cyclin E/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27 , Female , Humans , Intracellular Signaling Peptides and Proteins/physiology
5.
Science ; 335(6066): 348-53, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22157079

ABSTRACT

Myc is an oncogenic transcription factor frequently dysregulated in human cancer. To identify pathways supporting the Myc oncogenic program, we used a genome-wide RNA interference screen to search for Myc-synthetic lethal genes and uncovered a role for the SUMO-activating enzyme (SAE1/2). Loss of SAE1/2 enzymatic activity drives synthetic lethality with Myc. Inactivation of SAE2 leads to mitotic catastrophe and cell death upon Myc hyperactivation. Mechanistically, SAE2 inhibition switches a transcriptional subprogram of Myc from activated to repressed. A subset of these SUMOylation-dependent Myc switchers (SMS genes) is required for mitotic spindle function and to support the Myc oncogenic program. SAE2 is required for growth of Myc-dependent tumors in mice, and gene expression analyses of Myc-high human breast cancers suggest that low SAE1 and SAE2 abundance in the tumors correlates with longer metastasis-free survival of the patients. Thus, inhibition of SUMOylation may merit investigation as a possible therapy for Myc-driven human cancers.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Genes, myc , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Ubiquitin-Activating Enzymes/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/mortality , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Mitosis , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering , Spindle Apparatus/physiology , Sumoylation , Transplantation, Heterologous , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL