Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Cell ; 167(5): 1281-1295.e18, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863244

ABSTRACT

Glioblastoma stem cells (GSCs) are implicated in tumor neovascularization, invasiveness, and therapeutic resistance. To illuminate mechanisms governing these hallmark features, we developed a de novo glioblastoma multiforme (GBM) model derived from immortalized human neural stem/progenitor cells (hNSCs) to enable precise system-level comparisons of pre-malignant and oncogene-induced malignant states of NSCs. Integrated transcriptomic and epigenomic analyses uncovered a PAX6/DLX5 transcriptional program driving WNT5A-mediated GSC differentiation into endothelial-like cells (GdECs). GdECs recruit existing endothelial cells to promote peritumoral satellite lesions, which serve as a niche supporting the growth of invasive glioma cells away from the primary tumor. Clinical data reveal higher WNT5A and GdECs expression in peritumoral and recurrent GBMs relative to matched intratumoral and primary GBMs, respectively, supporting WNT5A-mediated GSC differentiation and invasive growth in disease recurrence. Thus, the PAX6/DLX5-WNT5A axis governs the diffuse spread of glioma cells throughout the brain parenchyma, contributing to the lethality of GBM.


Subject(s)
Glioblastoma/genetics , Glioblastoma/pathology , Neoplasm Invasiveness/genetics , Wnt-5a Protein/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epigenomics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Neural Stem Cells/metabolism , PAX6 Transcription Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism
3.
Cell ; 158(1): 185-197, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24954535

ABSTRACT

Activating mutations in KRAS are among the most frequent events in diverse human carcinomas and are particularly prominent in human pancreatic ductal adenocarcinoma (PDAC). An inducible Kras(G12D)-driven mouse model of PDAC has established a critical role for sustained Kras(G12D) expression in tumor maintenance, providing a model to determine the potential for and the underlying mechanisms of Kras(G12D)-independent PDAC recurrence. Here, we show that some tumors undergo spontaneous relapse and are devoid of Kras(G12D) expression and downstream canonical MAPK signaling and instead acquire amplification and overexpression of the transcriptional coactivator Yap1. Functional studies established the role of Yap1 and the transcriptional factor Tead2 in driving Kras(G12D)-independent tumor maintenance. The Yap1/Tead2 complex acts cooperatively with E2F transcription factors to activate a cell cycle and DNA replication program. Our studies, along with corroborating evidence from human PDAC models, portend a novel mechanism of escape from oncogenic Kras addiction in PDAC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle , Cell Cycle Proteins , Cell Line, Tumor , DNA Replication , DNA-Binding Proteins/metabolism , Disease Models, Animal , E2F Transcription Factors/metabolism , Humans , Mice , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , TEA Domain Transcription Factors , Transcription Factors/metabolism , YAP-Signaling Proteins , ras Proteins/metabolism
4.
Nature ; 619(7970): 632-639, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37344599

ABSTRACT

Sex exerts a profound impact on cancer incidence, spectrum and outcomes, yet the molecular and genetic bases of such sex differences are ill-defined and presumptively ascribed to X-chromosome genes and sex hormones1. Such sex differences are particularly prominent in colorectal cancer (CRC) in which men experience higher metastases and mortality. A murine CRC model, engineered with an inducible transgene encoding oncogenic mutant KRASG12D and conditional null alleles of Apc and Trp53 tumour suppressors (designated iKAP)2, revealed higher metastases and worse outcomes specifically in males with oncogenic mutant KRAS (KRAS*) CRC. Integrated cross-species molecular and transcriptomic analyses identified Y-chromosome gene histone demethylase KDM5D as a transcriptionally upregulated gene driven by KRAS*-mediated activation of the STAT4 transcription factor. KDM5D-dependent chromatin mark and transcriptome changes showed repression of regulators of the epithelial cell tight junction and major histocompatibility complex class I complex components. Deletion of Kdm5d in iKAP cancer cells increased tight junction integrity, decreased cell invasiveness and enhanced cancer cell killing by CD8+ T cells. Conversely, iAP mice engineered with a Kdm5d transgene to provide constitutive Kdm5d expression specifically in iAP cancer cells showed an increased propensity for more invasive tumours in vivo. Thus, KRAS*-STAT4-mediated upregulation of Y chromosome KDM5D contributes substantially to the sex differences in KRAS* CRC by means of its disruption of cancer cell adhesion properties and tumour immunity, providing an actionable therapeutic strategy for metastasis risk reduction for men afflicted with KRAS* CRC.


Subject(s)
Colorectal Neoplasms , Histone Demethylases , Minor Histocompatibility Antigens , Sex Characteristics , Animals , Female , Humans , Male , Mice , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Histone Demethylases/genetics , Histone Demethylases/metabolism , Mice, Transgenic , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Up-Regulation
5.
Genes Dev ; 35(19-20): 1327-1332, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34531315

ABSTRACT

Activating mutations in KRAS (KRAS*) are present in nearly all pancreatic ductal adenocarcinoma (PDAC) cases and critical for tumor maintenance. By using an inducible KRAS* PDAC mouse model, we identified a deubiquitinase USP21-driven resistance mechanism to anti-KRAS* therapy. USP21 promotes KRAS*-independent tumor growth via its regulation of MARK3-induced macropinocytosis, which serves to maintain intracellular amino acid levels for anabolic growth. The USP21-mediated KRAS* bypass, coupled with the frequent amplification of USP21 in human PDAC tumors, encourages the assessment of USP21 as a novel drug target as well as a potential parameter that may affect responsiveness to emergent anti-KRAS* therapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deubiquitinating Enzymes/metabolism , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Ubiquitin Thiolesterase
6.
Cell ; 148(5): 896-907, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22341455

ABSTRACT

To determine the role of telomere dysfunction and telomerase reactivation in generating pro-oncogenic genomic events and in carcinoma progression, an inducible telomerase reverse transcriptase (mTert) allele was crossed onto a prostate cancer-prone mouse model null for Pten and p53 tumor suppressors. Constitutive telomerase deficiency and associated telomere dysfunction constrained cancer progression. In contrast, telomerase reactivation in the setting of telomere dysfunction alleviated intratumoral DNA-damage signaling and generated aggressive cancers with rearranged genomes and new tumor biological properties (bone metastases). Comparative oncogenomic analysis revealed numerous recurrent amplifications and deletions of relevance to human prostate cancer. Murine tumors show enrichment of the TGF-ß/SMAD4 network, and genetic validation studies confirmed the cooperative roles of Pten, p53, and Smad4 deficiencies in prostate cancer progression, including skeletal metastases. Thus, telomerase reactivation in tumor cells experiencing telomere dysfunction enables full malignant progression and provides a mechanism for acquisition of cancer-relevant genomic events endowing new tumor biological capabilities.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Telomerase/metabolism , Telomere/metabolism , Animals , Bone Neoplasms/secondary , Cell Line, Tumor , Crosses, Genetic , DNA Copy Number Variations , Disease Models, Animal , Female , Genomic Instability , Humans , Male , Mice , Tumor Suppressor Protein p53/metabolism
7.
Cell ; 149(3): 656-70, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541435

ABSTRACT

Tumor maintenance relies on continued activity of driver oncogenes, although their rate-limiting role is highly context dependent. Oncogenic Kras mutation is the signature event in pancreatic ductal adenocarcinoma (PDAC), serving a critical role in tumor initiation. Here, an inducible Kras(G12D)-driven PDAC mouse model establishes that advanced PDAC remains strictly dependent on Kras(G12D) expression. Transcriptome and metabolomic analyses indicate that Kras(G12D) serves a vital role in controlling tumor metabolism through stimulation of glucose uptake and channeling of glucose intermediates into the hexosamine biosynthesis and pentose phosphate pathways (PPP). These studies also reveal that oncogenic Kras promotes ribose biogenesis. Unlike canonical models, we demonstrate that Kras(G12D) drives glycolysis intermediates into the nonoxidative PPP, thereby decoupling ribose biogenesis from NADP/NADPH-mediated redox control. Together, this work provides in vivo mechanistic insights into how oncogenic Kras promotes metabolic reprogramming in native tumors and illuminates potential metabolic targets that can be exploited for therapeutic benefit in PDAC.


Subject(s)
Adenocarcinoma/metabolism , Disease Models, Animal , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Humans , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Transcription, Genetic
8.
Genes Dev ; 33(19-20): 1361-1366, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31488580

ABSTRACT

The ubiquitin-specific protease (USP) family is the largest group of cysteine proteases. Cancer genomic analysis identified frequent amplification of USP21 (22%) in human pancreatic ductal adenocarcinoma (PDAC). USP21 overexpression correlates with human PDAC progression, and enforced expression of USP21 accelerates murine PDAC tumor growth and drives PanIN to PDAC progression in immortalized human pancreatic ductal cells. Conversely, depletion of USP21 impairs PDAC tumor growth. Mechanistically, USP21 deubiquitinates and stabilizes the TCF/LEF transcription factor TCF7, which promotes cancer cell stemness. Our work identifies and validates USP21 as a PDAC oncogene, providing a potential druggable target for this intractable disease.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Pancreatic Neoplasms/enzymology , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Line, Tumor , Humans , Mice , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology , T Cell Transcription Factor 1 , Ubiquitination , Pancreatic Neoplasms
9.
Nature ; 568(7752): 410-414, 2019 04.
Article in English | MEDLINE | ID: mdl-30918400

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) remains recalcitrant to all forms of cancer treatment and carries a five-year survival rate of only 8%1. Inhibition of oncogenic KRAS (hereafter KRAS*), the earliest lesion in disease development that is present in more than 90% of PDACs, and its signalling surrogates has yielded encouraging preclinical results with experimental agents2-4. However, KRAS*-independent disease recurrence following genetic extinction of Kras* in mouse models anticipates the need for co-extinction strategies5,6. Multiple oncogenic processes are initiated at the cell surface, where KRAS* physically and functionally interacts to direct signalling that is essential for malignant transformation and tumour maintenance. Insights into the complexity of the functional cell-surface-protein repertoire (surfaceome) have been technologically limited until recently and-in the case of PDAC-the genetic control of the function and composition of the PDAC surfaceome in the context of KRAS* signalling remains largely unknown. Here we develop an unbiased, functional target-discovery platform to query KRAS*-dependent changes of the PDAC surfaceome, which reveals syndecan 1 (SDC1, also known as CD138) as a protein that is upregulated at the cell surface by KRAS*. Localization of SDC1 at the cell surface-where it regulates macropinocytosis, an essential metabolic pathway that fuels PDAC cell growth-is essential for disease maintenance and progression. Thus, our study forges a mechanistic link between KRAS* signalling and a targetable molecule driving nutrient salvage pathways in PDAC and validates oncogene-driven surfaceome annotation as a strategy to identify cancer-specific vulnerabilities.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Pinocytosis , Syndecan-1/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Disease Progression , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction
10.
Genes Dev ; 31(23-24): 2337-2342, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29352019

ABSTRACT

SMAD4 constrains progression of Pten-null prostate cancer and serves as a common downstream node of transforming growth factor ß (TGFß) and bone morphogenetic protein (BMP) pathways. Here, we dissected the roles of TGFß receptor II (TGFBR2) and BMP receptor II (BMPR2) using a Pten-null prostate cancer model. These studies demonstrated that the molecular actions of TGFBR2 result in both SMAD4-dependent constraint of proliferation and SMAD4-independent activation of apoptosis. In contrast, BMPR2 deletion extended survival relative to Pten deletion alone, establishing its promoting role in BMP6-driven prostate cancer progression. These analyses reveal the complexity of TGFß-BMP signaling and illuminate potential therapeutic targets for prostate cancer.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Prostatic Neoplasms/physiopathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Animals , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Disease Models, Animal , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genotype , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Receptor, Transforming Growth Factor-beta Type II , Smad4 Protein/genetics , Smad4 Protein/metabolism
11.
Genes Dev ; 31(4): 370-382, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28289141

ABSTRACT

Human colorectal cancer (CRC) is a major cause of cancer mortality and frequently harbors activating mutations in the KRAS gene. To understand the role of oncogenic KRAS in CRC, we engineered a mouse model of metastatic CRC that harbors an inducible oncogenic Kras allele (Krasmut ) and conditional null alleles of Apc and Trp53 (iKAP). The iKAP model recapitulates tumor progression from adenoma through metastases. Whole-exome sequencing revealed that the Krasmut allele was heterogenous in primary tumors yet homogenous in metastases, a pattern consistent with activated Krasmut signaling being a driver of progression to metastasis. System-level and functional analyses revealed the TGF-ß pathway as a key mediator of Krasmut -driven invasiveness. Genetic extinction of Krasmut resulted in specific elimination of the Krasmut subpopulation in primary and metastatic tumors, leading to apoptotic elimination of advanced invasive and metastatic disease. This faithful CRC model provides genetic evidence that Krasmut drives CRC invasion and maintenance of metastases.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/physiopathology , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Genotype , Humans , Mice , Mice, Inbred C57BL , Mutation , Neoplasm Metastasis , Proto-Oncogene Proteins p21(ras)/genetics , Transcriptome , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34475205

ABSTRACT

Prostate cancer is a leading cause of cancer-related mortality in men. The widespread use of androgen receptor (AR) inhibitors has generated an increased incidence of AR-negative prostate cancer, triggering the need for effective therapies for such patients. Here, analysis of public genome-wide CRISPR screens in human prostate cancer cell lines identified histone demethylase JMJD1C (KDM3C) as an AR-negative context-specific vulnerability. Secondary validation studies in multiple cell lines and organoids, including isogenic models, confirmed that small hairpin RNA (shRNA)-mediated depletion of JMJD1C potently inhibited growth specifically in AR-negative prostate cancer cells. To explore the cooperative interactions of AR and JMJD1C, we performed comparative transcriptomics of 1) isogenic AR-positive versus AR-negative prostate cancer cells, 2) AR-positive versus AR-negative prostate cancer tumors, and 3) isogenic JMJD1C-expressing versus JMJD1C-depleted AR-negative prostate cancer cells. Loss of AR or JMJD1C generates a modest tumor necrosis factor alpha (TNFα) signature, whereas combined loss of AR and JMJD1C strongly up-regulates the TNFα signature in human prostate cancer, suggesting TNFα signaling as a point of convergence for the combined actions of AR and JMJD1C. Correspondingly, AR-negative prostate cancer cells showed exquisite sensitivity to TNFα treatment and, conversely, TNFα pathway inhibition via inhibition of its downstream effector MAP4K4 partially reversed the growth defect of JMJD1C-depleted AR-negative prostate cancer cells. Given the deleterious systemic side effects of TNFα therapy in humans and the viability of JMJD1C-knockout mice, the identification of JMJD1C inhibition as a specific vulnerability in AR-negative prostate cancer may provide an alternative drug target for prostate cancer patients progressing on AR inhibitor therapy.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/genetics , Oxidoreductases, N-Demethylating/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Databases, Genetic , Histone Demethylases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic/drug effects , Prostate/pathology , Protein Serine-Threonine Kinases/genetics , Receptors, Androgen/genetics , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/metabolism
13.
Nature ; 543(7647): 728-732, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28321130

ABSTRACT

A significant fraction of patients with advanced prostate cancer treated with androgen deprivation therapy experience relapse with relentless progression to lethal metastatic castration-resistant prostate cancer (mCRPC). Immune checkpoint blockade using antibodies against cytotoxic-T-lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1/programmed cell death 1 ligand 1 (PD1/PD-L1) generates durable therapeutic responses in a significant subset of patients across a variety of cancer types. However, mCRPC showed overwhelming de novo resistance to immune checkpoint blockade, motivating a search for targeted therapies that overcome this resistance. Myeloid-derived suppressor cells (MDSCs) are known to play important roles in tumour immune evasion. The abundance of circulating MDSCs correlates with prostate-specific antigen levels and metastasis in patients with prostate cancer. Mouse models of prostate cancer show that MDSCs (CD11b+Gr1+) promote tumour initiation and progression. These observations prompted us to hypothesize that robust immunotherapy responses in mCRPC may be elicited by the combined actions of immune checkpoint blockade agents together with targeted agents that neutralize MDSCs yet preserve T-cell function. Here we develop a novel chimaeric mouse model of mCRPC to efficiently test combination therapies in an autochthonous setting. Combination of anti-CTLA4 and anti-PD1 engendered only modest efficacy. Targeted therapy against mCRPC-infiltrating MDSCs, using multikinase inhibitors such as cabozantinib and BEZ235, also showed minimal anti-tumour activities. Strikingly, primary and metastatic CRPC showed robust synergistic responses when immune checkpoint blockade was combined with MDSC-targeted therapy. Mechanistically, combination therapy efficacy stemmed from the upregulation of interleukin-1 receptor antagonist and suppression of MDSC-promoting cytokines secreted by prostate cancer cells. These observations illuminate a clinical path hypothesis for combining immune checkpoint blockade with MDSC-targeted therapies in the treatment of mCRPC.


Subject(s)
Immunotherapy/methods , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/therapy , Anilides/pharmacology , Anilides/therapeutic use , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chimera , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Female , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Molecular Targeted Therapy , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms, Castration-Resistant/pathology , Pyridines/pharmacology , Pyridines/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
Nature ; 542(7642): 484-488, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28166537

ABSTRACT

Synthetic lethality and collateral lethality are two well-validated conceptual strategies for identifying therapeutic targets in cancers with tumour-suppressor gene deletions. Here, we explore an approach to identify potential synthetic-lethal interactions by screening mutually exclusive deletion patterns in cancer genomes. We sought to identify 'synthetic-essential' genes: those that are occasionally deleted in some cancers but are almost always retained in the context of a specific tumour-suppressor deficiency. We also posited that such synthetic-essential genes would be therapeutic targets in cancers that harbour specific tumour-suppressor deficiencies. In addition to known synthetic-lethal interactions, this approach uncovered the chromatin helicase DNA-binding factor CHD1 as a putative synthetic-essential gene in PTEN-deficient cancers. In PTEN-deficient prostate and breast cancers, CHD1 depletion profoundly and specifically suppressed cell proliferation, cell survival and tumorigenic potential. Mechanistically, functional PTEN stimulates the GSK3ß-mediated phosphorylation of CHD1 degron domains, which promotes CHD1 degradation via the ß-TrCP-mediated ubiquitination-proteasome pathway. Conversely, PTEN deficiency results in stabilization of CHD1, which in turn engages the trimethyl lysine-4 histone H3 modification to activate transcription of the pro-tumorigenic TNF-NF-κB gene network. This study identifies a novel PTEN pathway in cancer and provides a framework for the discovery of 'trackable' targets in cancers that harbour specific tumour-suppressor deficiencies.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Genes, Essential/genetics , Neoplasms/metabolism , Neoplasms/pathology , PTEN Phosphohydrolase/deficiency , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , DNA Helicases/chemistry , DNA Helicases/deficiency , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Male , Methylation , Molecular Targeted Therapy , NF-kappa B/metabolism , Neoplasms/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination , beta-Transducin Repeat-Containing Proteins/metabolism
15.
Nature ; 542(7639): 119-123, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28099419

ABSTRACT

The genome of pancreatic ductal adenocarcinoma (PDAC) frequently contains deletions of tumour suppressor gene loci, most notably SMAD4, which is homozygously deleted in nearly one-third of cases. As loss of neighbouring housekeeping genes can confer collateral lethality, we sought to determine whether loss of the metabolic gene malic enzyme 2 (ME2) in the SMAD4 locus would create cancer-specific metabolic vulnerability upon targeting of its paralogous isoform ME3. The mitochondrial malic enzymes (ME2 and ME3) are oxidative decarboxylases that catalyse the conversion of malate to pyruvate and are essential for NADPH regeneration and reactive oxygen species homeostasis. Here we show that ME3 depletion selectively kills ME2-null PDAC cells in a manner consistent with an essential function for ME3 in ME2-null cancer cells. Mechanistically, integrated metabolomic and molecular investigation of cells deficient in mitochondrial malic enzymes revealed diminished NADPH production and consequent high levels of reactive oxygen species. These changes activate AMP activated protein kinase (AMPK), which in turn directly suppresses sterol regulatory element-binding protein 1 (SREBP1)-directed transcription of its direct targets including the BCAT2 branched-chain amino acid transaminase 2) gene. BCAT2 catalyses the transfer of the amino group from branched-chain amino acids to α-ketoglutarate (α-KG) thereby regenerating glutamate, which functions in part to support de novo nucleotide synthesis. Thus, mitochondrial malic enzyme deficiency, which results in impaired NADPH production, provides a prime 'collateral lethality' therapeutic strategy for the treatment of a substantial fraction of patients diagnosed with this intractable disease.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Deletion , Malate Dehydrogenase/deficiency , Pancreatic Neoplasms/genetics , AMP-Activated Protein Kinases/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Biocatalysis , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/psychology , Carcinoma, Pancreatic Ductal/therapy , Humans , Ketoglutaric Acids/metabolism , Malate Dehydrogenase/genetics , Male , Mice , Minor Histocompatibility Antigens/biosynthesis , Minor Histocompatibility Antigens/genetics , Mitochondria/enzymology , Mitochondria/pathology , NADP/biosynthesis , NADP/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Pregnancy Proteins/biosynthesis , Pregnancy Proteins/genetics , Reactive Oxygen Species/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transaminases/biosynthesis , Transaminases/genetics
16.
Nature ; 514(7524): 628-32, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25119024

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in western countries, with a median survival of 6 months and an extremely low percentage of long-term surviving patients. KRAS mutations are known to be a driver event of PDAC, but targeting mutant KRAS has proved challenging. Targeting oncogene-driven signalling pathways is a clinically validated approach for several devastating diseases. Still, despite marked tumour shrinkage, the frequency of relapse indicates that a fraction of tumour cells survives shut down of oncogenic signalling. Here we explore the role of mutant KRAS in PDAC maintenance using a recently developed inducible mouse model of mutated Kras (Kras(G12D), herein KRas) in a p53(LoxP/WT) background. We demonstrate that a subpopulation of dormant tumour cells surviving oncogene ablation (surviving cells) and responsible for tumour relapse has features of cancer stem cells and relies on oxidative phosphorylation for survival. Transcriptomic and metabolic analyses of surviving cells reveal prominent expression of genes governing mitochondrial function, autophagy and lysosome activity, as well as a strong reliance on mitochondrial respiration and a decreased dependence on glycolysis for cellular energetics. Accordingly, surviving cells show high sensitivity to oxidative phosphorylation inhibitors, which can inhibit tumour recurrence. Our integrated analyses illuminate a therapeutic strategy of combined targeting of the KRAS pathway and mitochondrial respiration to manage pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Mitochondria/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Autophagy , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cell Respiration/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Glycolysis , Lysosomes/metabolism , Mice , Mitochondria/drug effects , Mutation/genetics , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxidative Phosphorylation/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Recurrence , Signal Transduction , Pancreatic Neoplasms
17.
Proc Natl Acad Sci U S A ; 114(43): E9086-E9095, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073105

ABSTRACT

An integrated genomic and functional analysis to elucidate DNA damage signaling factors promoting self-renewal of glioma stem cells (GSCs) identified proliferating cell nuclear antigen (PCNA)-associated factor (PAF) up-regulation in glioblastoma. PAF is preferentially overexpressed in GSCs. Its depletion impairs maintenance of self-renewal without promoting differentiation and reduces tumor-initiating cell frequency. Combined transcriptomic and metabolomic analyses revealed that PAF supports GSC maintenance, in part, by influencing DNA replication and pyrimidine metabolism pathways. PAF interacts with PCNA and regulates PCNA-associated DNA translesion synthesis (TLS); consequently, PAF depletion in combination with radiation generated fewer tumorspheres compared with radiation alone. Correspondingly, pharmacological impairment of DNA replication and TLS phenocopied the effect of PAF depletion in compromising GSC self-renewal and radioresistance, providing preclinical proof of principle that combined TLS inhibition and radiation therapy may be a viable therapeutic option in the treatment of glioblastoma multiforme (GBM).


Subject(s)
Brain Neoplasms/radiotherapy , Carrier Proteins/genetics , Glioblastoma/radiotherapy , Neoplastic Stem Cells/radiation effects , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Carrier Proteins/metabolism , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , DNA Replication/drug effects , DNA-Binding Proteins , Female , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Green Fluorescent Proteins/genetics , Humans , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Pyrimidines/biosynthesis , Radiation Tolerance , Xenograft Model Antitumor Assays
18.
Genes Dev ; 26(13): 1459-72, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22751500

ABSTRACT

Multidimensional cancer genome analysis and validation has defined Quaking (QKI), a member of the signal transduction and activation of RNA (STAR) family of RNA-binding proteins, as a novel glioblastoma multiforme (GBM) tumor suppressor. Here, we establish that p53 directly regulates QKI gene expression, and QKI protein associates with and leads to the stabilization of miR-20a; miR-20a, in turn, regulates TGFßR2 and the TGFß signaling network. This pathway circuitry is substantiated by in silico epistasis analysis of its components in the human GBM TCGA (The Cancer Genome Atlas Project) collection and by their gain- and loss-of-function interactions in in vitro and in vivo complementation studies. This p53-QKI-miR-20a-TGFß pathway expands our understanding of the p53 tumor suppression network in cancer and reveals a novel tumor suppression mechanism involving regulation of specific cancer-relevant microRNAs.


Subject(s)
Cell Line , Glioblastoma/metabolism , MicroRNAs/metabolism , RNA Stability , RNA-Binding Proteins/metabolism , Animals , Glioblastoma/genetics , Humans , Mice , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
Nat Chem Biol ; 12(12): 1053-1058, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27723749

ABSTRACT

Despite being crucial for energy generation in most forms of life, few if any microbial antibiotics specifically inhibit glycolysis. To develop a specific inhibitor of the glycolytic enzyme enolase 2 (ENO2) for the treatment of cancers with deletion of ENO1 (encoding enolase 1), we modeled the synthetic tool compound inhibitor phosphonoacetohydroxamate (PhAH) into the active site of human ENO2. A ring-stabilized analog of PhAH, in which the hydroxamic nitrogen is linked to Cα by an ethylene bridge, was predicted to increase binding affinity by stabilizing the inhibitor in a bound conformation. Unexpectedly, a structure-based search revealed that our hypothesized backbone-stabilized PhAH bears strong similarity to SF2312, a phosphonate antibiotic of unknown mode of action produced by the actinomycete Micromonospora, which is active under anaerobic conditions. Here, we present multiple lines of evidence, including a novel X-ray structure, that SF2312 is a highly potent, low-nanomolar inhibitor of enolase.


Subject(s)
Enzyme Inhibitors/pharmacology , Organophosphonates/pharmacology , Phosphopyruvate Hydratase/antagonists & inhibitors , Pyrrolidinones/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Organophosphonates/chemistry , Phosphopyruvate Hydratase/metabolism , Pyrrolidinones/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL