Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 23(1): 58-67, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617673

ABSTRACT

A hallmark of advanced prostate cancer (PC) is the concomitant loss of PTEN and p53 function. To selectively eliminate such cells, we screened cytotoxic compounds on Pten-/-;Trp53-/- fibroblasts and their Pten-WT reference. Highly selective killing of Pten-null cells can be achieved by deguelin, a natural insecticide. Deguelin eliminates Pten-deficient cells through inhibition of mitochondrial complex I (CI). Five hundred-fold higher drug doses are needed to obtain the same killing of Pten-WT cells, even though deguelin blocks their electron transport chain equally well. Selectivity arises because mitochondria of Pten-null cells consume ATP through complex V, instead of producing it. The resulting glucose dependency can be exploited to selectively kill Pten-null cells with clinically relevant CI inhibitors, especially if they are lipophilic. In vivo, deguelin suppressed disease in our genetically engineered mouse model for metastatic PC. Our data thus introduce a vulnerability for highly selective targeting of incurable PC with inhibitors of CI.


Subject(s)
Antineoplastic Agents/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Prostatic Neoplasms/drug therapy , Rotenone/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cells, Cultured , Electron Transport Complex I/metabolism , Enzyme Inhibitors/therapeutic use , Fibroblasts/metabolism , Glucose/metabolism , Male , Mice , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Rotenone/pharmacology , Rotenone/therapeutic use , Tumor Suppressor Protein p53/genetics
2.
J Cell Biol ; 216(3): 641-656, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28193700

ABSTRACT

Phosphatase and tensin homologue (PTEN) protein levels are critical for tumor suppression. However, the search for a recurrent cancer-associated gene alteration that causes PTEN degradation has remained futile. In this study, we show that Importin-11 (Ipo11) is a transport receptor for PTEN that is required to physically separate PTEN from elements of the PTEN degradation machinery. Mechanistically, we find that the E2 ubiquitin-conjugating enzyme and IPO11 cargo, UBE2E1, is a limiting factor for PTEN degradation. Using in vitro and in vivo gene-targeting methods, we show that Ipo11 loss results in degradation of Pten, lung adenocarcinoma, and neoplasia in mouse prostate with aberrantly high levels of Ube2e1 in the cytoplasm. These findings explain the correlation between loss of IPO11 and PTEN protein in human lung tumors. Furthermore, we find that IPO11 status predicts disease recurrence and progression to metastasis in patients choosing radical prostatectomy. Thus, our data introduce the IPO11 gene as a tumor-suppressor locus, which is of special importance in cancers that still retain at least one intact PTEN allele.


Subject(s)
PTEN Phosphohydrolase/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Proteins/metabolism , beta Karyopherins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , HeLa Cells , Humans , Lung Neoplasms/metabolism , Mice , Ubiquitin-Conjugating Enzymes/metabolism
3.
Cancer Discov ; 5(6): 636-51, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25829425

ABSTRACT

UNLABELLED: We have recently recapitulated metastasis of human PTEN/TP53-mutant prostate cancer in the mouse using the RapidCaP system. Surprisingly, we found that this metastasis is driven by MYC, and not AKT, activation. Here, we show that cell-cell communication by IL6 drives the AKT-MYC switch through activation of the AKT-suppressing phosphatase PHLPP2, when PTEN and p53 are lost together, but not separately. IL6 then communicates a downstream program of STAT3-mediated MYC activation, which drives cell proliferation. Similarly, in tissues, peak proliferation in Pten/Trp53-mutant primary and metastatic prostate cancer does not correlate with activated AKT, but with STAT3/MYC activation instead. Mechanistically, MYC strongly activates the AKT phosphatase PHLPP2 in primary cells and prostate cancer metastasis. We show genetically that Phlpp2 is essential for dictating the proliferation of MYC-mediated AKT suppression. Collectively, our data reveal competition between two proto-oncogenes, MYC and AKT, which ensnarls the Phlpp2 gene to facilitate MYC-driven prostate cancer metastasis after loss of Pten and Trp53. SIGNIFICANCE: Our data identify IL6 detection as a potential causal biomarker for MYC-driven metastasis after loss of PTEN and p53. Second, our finding that MYC then must supersede AKT to drive cell proliferation points to MYC inhibition as a critical part of PI3K pathway therapy in lethal prostate cancer.


Subject(s)
Genes, myc , Interleukin-6/metabolism , Neoplasms/genetics , Neoplasms/metabolism , PTEN Phosphohydrolase/deficiency , Phosphoprotein Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/deficiency , Animals , Cell Communication/genetics , Cell Proliferation , Epithelium/metabolism , Epithelium/pathology , Gene Deletion , Genotype , Humans , Lung Neoplasms/secondary , Male , Mice , Mutation , Neoplasm Metastasis , Neoplasms/pathology , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , STAT3 Transcription Factor/metabolism , Signal Transduction , Stromal Cells/metabolism
4.
Methods ; 77-78: 197-204, 2015 May.
Article in English | MEDLINE | ID: mdl-25592467

ABSTRACT

Human genome analyses have revealed that increasing gene copy number alteration is a driving force of incurable cancer of the prostate (CaP). Since most of the affected genes are hidden within large amplifications or deletions, there is a need for fast and faithful validation of drivers. However, classic genetic CaP engineering in mouse makes this a daunting task because generation, breeding based combination of alterations and non-invasive monitoring of disease are too time consuming and costly. To address the unmet need, we recently developed RapidCaP mice, which endogenously recreate human PTEN-mutant metastatic CaP based on Cre/Luciferase expressing viral infection, that is guided to Pten(loxP)/Trp53(loxP) prostate. Here we use a sensitized, non-metastatic Pten/Trp53-mutant RapidCaP system for functional validation of human metastasis drivers in a much accelerated time frame of only 3-4months. We used in vivo RNAi to target three candidate tumor suppressor genes FOXP1, RYBP and SHQ1, which reside in a frequent deletion on chromosome 3p and show that Shq1 cooperates with Pten and p53 to suppress metastasis. Our results thus demonstrate that the RapidCaP system forms a much needed platform for in vivo screening and validation of genes that drive endogenous lethal CaP.


Subject(s)
Genetic Association Studies/methods , Genome/genetics , Mutation/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Humans , Male , Mice , Mice, Knockout , PTEN Phosphohydrolase/biosynthesis , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Time Factors , Tumor Suppressor Proteins/biosynthesis
5.
Cell Rep ; 10(1): 8-19, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25543136

ABSTRACT

Phosphatidylinositol phosphate (PIP) second messengers relay extracellular growth cues through the phosphorylation status of the inositol sugar, a signal transduction system that is deregulated in cancer. In stark contrast to PIP inositol head-group phosphorylation, changes in phosphatidylinositol (PI) lipid acyl chains in cancer have remained ill-defined. Here, we apply a mass-spectrometry-based method capable of unbiased high-throughput identification and quantification of cellular PI acyl chain composition. Using this approach, we find that PI lipid chains represent a cell-specific fingerprint and are unperturbed by serum-mediated signaling in contrast to the inositol head group. We find that mutation of Trp53 results in PIs containing reduced-length fatty acid moieties. Our results suggest that the anchoring tails of lipid second messengers form an additional layer of PIP signaling in cancer that operates independently of PTEN/PI3-kinase activity but is instead linked to p53.


Subject(s)
Fatty Acids/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol Phosphates/metabolism , Tumor Suppressor Protein p53/genetics , Cell Line , High-Throughput Screening Assays , Humans , Lipid Metabolism/genetics , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/isolation & purification , Phosphorylation , Second Messenger Systems/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism
6.
Cancer Discov ; 4(3): 318-33, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24444712

ABSTRACT

Genetically engineered mouse (GEM) models are a pillar of functional cancer research. Here, we developed RapidCaP, a GEM modeling system that uses surgical injection for viral gene delivery to the prostate. We show that in Pten deficiency, loss of p53 suffices to trigger metastasis to distant sites at greater than 50% penetrance by four months, consistent with results from human prostate cancer genome analysis. Live bioluminescence tracking showed that endogenous primary and metastatic disease responds to castration before developing lethal castration resistance. To our surprise, the resulting lesions showed no activation of Akt but activation of the Myc oncogene. Using RapidCaP, we find that Myc drives local prostate metastasis and is critical for maintenance of metastasis, as shown by using the Brd4 inhibitor JQ1. Taken together, our data suggest that a "MYC-switch" away from AKT forms a critical and druggable event in PTEN-mutant prostate cancer metastasis and castration resistance.


Subject(s)
Neoplasm Metastasis/pathology , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Retroviridae/genetics , Tumor Suppressor Protein p53/genetics , Animals , Azepines/pharmacology , Gene Expression Regulation, Neoplastic , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-myc/genetics , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...