Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Metab ; 5(12): 2131-2147, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37957387

ABSTRACT

Glutamine is a critical metabolite for rapidly proliferating cells as it is used for the synthesis of key metabolites necessary for cell growth and proliferation. Glutamine metabolism has been proposed as a therapeutic target in cancer and several chemical inhibitors are in development or in clinical trials. How cells subsist when glutamine is limiting is poorly understood. Here, using an unbiased screen, we identify ALDH18A1, which encodes P5CS, the rate-limiting enzyme in the proline biosynthetic pathway, as a gene that cells can downregulate in response to glutamine starvation. Notably, P5CS downregulation promotes de novo glutamine synthesis, highlighting a previously unrecognized metabolic plasticity of cancer cells. The glutamate conserved from reducing proline synthesis allows cells to produce the key metabolites necessary for cell survival and proliferation under glutamine-restricted conditions. Our findings reveal an adaptive pathway that cancer cells acquire under nutrient stress, identifying proline biosynthesis as a previously unrecognized major consumer of glutamate, a pathway that could be exploited for developing effective metabolism-driven anticancer therapies.


Subject(s)
Glutamine , Neoplasms , Humans , Glutamine/metabolism , Cell Proliferation , Proline , Glutamates
2.
Nat Cell Biol ; 25(2): 285-297, 2023 02.
Article in English | MEDLINE | ID: mdl-36658220

ABSTRACT

Transcription factors (TFs) are frequently mutated in cancer. Paediatric cancers exhibit few mutations genome-wide but frequently harbour sentinel mutations that affect TFs, which provides a context to precisely study the transcriptional circuits that support mutant TF-driven oncogenesis. A broadly relevant mechanism that has garnered intense focus involves the ability of mutant TFs to hijack wild-type lineage-specific TFs in self-reinforcing transcriptional circuits. However, it is not known whether this specific type of circuitry is equally crucial in all mutant TF-driven cancers. Here we describe an alternative yet central transcriptional mechanism that promotes Ewing sarcoma, wherein constraint, rather than reinforcement, of the activity of the fusion TF EWS-FLI supports cancer growth. We discover that ETV6 is a crucial TF dependency that is specific to this disease because it, counter-intuitively, represses the transcriptional output of EWS-FLI. This work discovers a previously undescribed transcriptional mechanism that promotes cancer.


Subject(s)
Sarcoma, Ewing , Child , Humans , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins c-ets/genetics , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/genetics
3.
Nat Cancer ; 3(8): 976-993, 2022 08.
Article in English | MEDLINE | ID: mdl-35817829

ABSTRACT

Immunotherapy with anti-GD2 antibodies has advanced the treatment of children with high-risk neuroblastoma, but nearly half of patients relapse, and little is known about mechanisms of resistance to anti-GD2 therapy. Here, we show that reduced GD2 expression was significantly correlated with the mesenchymal cell state in neuroblastoma and that a forced adrenergic-to-mesenchymal transition (AMT) conferred downregulation of GD2 and resistance to anti-GD2 antibody. Mechanistically, low-GD2-expressing cell lines demonstrated significantly reduced expression of the ganglioside synthesis enzyme ST8SIA1 (GD3 synthase), resulting in a bottlenecking of GD2 synthesis. Pharmacologic inhibition of EZH2 resulted in epigenetic rewiring of mesenchymal neuroblastoma cells and re-expression of ST8SIA1, restoring surface expression of GD2 and sensitivity to anti-GD2 antibody. These data identify developmental lineage as a key determinant of sensitivity to anti-GD2 based immunotherapies and credential EZH2 inhibitors for clinical testing in combination with anti-GD2 antibody to enhance outcomes for children with neuroblastoma.


Subject(s)
Gangliosides , Neuroblastoma , Antibodies, Monoclonal , Child , Humans , Immunotherapy , Neoplasm Recurrence, Local/chemically induced , Neuroblastoma/drug therapy
4.
Nat Metab ; 3(2): 182-195, 2021 02.
Article in English | MEDLINE | ID: mdl-33619381

ABSTRACT

Head and neck squamous cell carcinoma (SCC) remains among the most aggressive human cancers. Tumour progression and aggressiveness in SCC are largely driven by tumour-propagating cells (TPCs). Aerobic glycolysis, also known as the Warburg effect, is a characteristic of many cancers; however, whether this adaptation is functionally important in SCC, and at which stage, remains poorly understood. Here, we show that the NAD+-dependent histone deacetylase sirtuin 6 is a robust tumour suppressor in SCC, acting as a modulator of glycolysis in these tumours. Remarkably, rather than a late adaptation, we find enhanced glycolysis specifically in TPCs. More importantly, using single-cell RNA sequencing of TPCs, we identify a subset of TPCs with higher glycolysis and enhanced pentose phosphate pathway and glutathione metabolism, characteristics that are strongly associated with a better antioxidant response. Together, our studies uncover enhanced glycolysis as a main driver in SCC, and, more importantly, identify a subset of TPCs as the cell of origin for the Warburg effect, defining metabolism as a key feature of intra-tumour heterogeneity.


Subject(s)
Glycolysis , Head and Neck Neoplasms/pathology , Neoplastic Stem Cells/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Antioxidants/metabolism , Disease Progression , Glutathione/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Pentose Phosphate Pathway , RNA, Neoplasm/genetics , Single-Cell Analysis , Sirtuins/genetics , Sirtuins/metabolism , Xenograft Model Antitumor Assays
5.
Dis Model Mech ; 13(8)2020 08 27.
Article in English | MEDLINE | ID: mdl-32651197

ABSTRACT

Polycomb repressive complex 2 (PRC2) is an epigenetic regulator of gene expression that possesses histone methyltransferase activity. PRC2 trimethylates lysine 27 of histone H3 proteins (H3K27me3) as a chromatin modification associated with repressed transcription of genes frequently involved in cell proliferation or self-renewal. Loss-of-function mutations in the PRC2 core subunit SUZ12 have been identified in a variety of tumors, including malignant peripheral nerve sheath tumors (MPNSTs). To determine the consequences of SUZ12 loss in the pathogenesis of MPNST and other cancers, we used CRISPR-Cas9 to disrupt the open reading frame of each of two orthologous suz12 genes in zebrafish: suz12a and suz12b We generated these knockout alleles in the germline of our previously described p53 (also known as tp53)- and nf1-deficient zebrafish model of MPNSTs. Loss of suz12 significantly accelerated the onset and increased the penetrance of MPNSTs compared to that in control zebrafish. Moreover, in suz12-deficient zebrafish, we detected additional types of tumors besides MPNSTs, including leukemia with histological characteristics of lymphoid malignancies, soft tissue sarcoma and pancreatic adenocarcinoma, which were not detected in p53/nf1-deficient control fish, and are also contained in the human spectrum of SUZ12-deficient malignancies identified in the AACR Genie database. The suz12-knockout tumors displayed reduced or abolished H3K27me3 epigenetic marks and upregulation of gene sets reported to be targeted by PRC2. Thus, these zebrafish lines with inactivation of suz12 in combination with loss of p53/nf1 provide a model of human MPNSTs and multiple other tumor types, which will be useful for mechanistic studies of molecular pathogenesis and targeted therapy with small molecule inhibitors.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Silencing , Neurofibromin 1/genetics , Neurofibrosarcoma/genetics , Tumor Suppressor Protein p53/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Neurofibromin 1/deficiency , Neurofibrosarcoma/drug therapy , Neurofibrosarcoma/metabolism , Neurofibrosarcoma/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Sarcoma/genetics , Sarcoma/metabolism , Sarcoma/pathology , Signal Transduction , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology , Tumor Suppressor Protein p53/deficiency , Zebrafish/metabolism , Zebrafish Proteins/deficiency
6.
Proc Natl Acad Sci U S A ; 117(28): 16516-16526, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601179

ABSTRACT

LIN28B is highly expressed in neuroblastoma and promotes tumorigenesis, at least, in part, through inhibition of let-7 microRNA biogenesis. Here, we report that overexpression of either wild-type (WT) LIN28B or a LIN28B mutant that is unable to inhibit let-7 processing increases the penetrance of MYCN-induced neuroblastoma, potentiates the invasion and migration of transformed sympathetic neuroblasts, and drives distant metastases in vivo. Genome-wide chromatin immunoprecipitation coupled with massively parallel DNA sequencing (ChIP-seq) and coimmunoprecipitation experiments show that LIN28B binds active gene promoters in neuroblastoma cells through protein-protein interaction with the sequence-specific zinc-finger transcription factor ZNF143 and activates the expression of downstream targets, including transcription factors forming the adrenergic core regulatory circuitry that controls the malignant cell state in neuroblastoma as well as GSK3B and L1CAM that are involved in neuronal cell adhesion and migration. These findings reveal an unexpected let-7-independent function of LIN28B in transcriptional regulation during neuroblastoma pathogenesis.


Subject(s)
N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , Animals, Genetically Modified , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/physiopathology , Protein Binding , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Zebrafish
7.
Sci Adv ; 6(5): eaay2611, 2020 01.
Article in English | MEDLINE | ID: mdl-32064343

ABSTRACT

Women harboring heterozygous germline mutations of BRCA2 have a 50 to 80% risk of developing breast cancer, yet the pathogenesis of these cancers is poorly understood. To reveal early steps in BRCA2-associated carcinogenesis, we analyzed sorted cell populations from freshly-isolated, non-cancerous breast tissues of BRCA2 mutation carriers and matched controls. Single-cell whole-genome sequencing demonstrates that >25% of BRCA2 carrier (BRCA2mut/+ ) luminal progenitor (LP) cells exhibit sub-chromosomal copy number variations, which are rarely observed in non-carriers. Correspondingly, primary BRCA2mut/+ breast epithelia exhibit DNA damage together with attenuated replication checkpoint and apoptotic responses, and an age-associated expansion of the LP compartment. We provide evidence that these phenotypes do not require loss of the wild-type BRCA2 allele. Collectively, our findings suggest that BRCA2 haploinsufficiency and associated DNA damage precede histologic abnormalities in vivo. Using these hallmarks of cancer predisposition will yield unanticipated opportunities for improved risk assessment and prevention strategies in high-risk patients.


Subject(s)
BRCA2 Protein/genetics , Breast Neoplasms/genetics , Genetic Predisposition to Disease , Haploinsufficiency/genetics , Adult , Aneuploidy , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Copy Number Variations/genetics , DNA Damage/genetics , Female , Germ-Line Mutation/genetics , Heterozygote , Humans , Middle Aged , Single-Cell Analysis
8.
Cancer Cell ; 36(6): 660-673.e11, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31821784

ABSTRACT

Inhibition of the Menin (MEN1) and MLL (MLL1, KMT2A) interaction is a potential therapeutic strategy for MLL-rearranged (MLL-r) leukemia. Structure-based design yielded the potent, highly selective, and orally bioavailable small-molecule inhibitor VTP50469. Cell lines carrying MLL rearrangements were selectively responsive to VTP50469. VTP50469 displaced Menin from protein complexes and inhibited chromatin occupancy of MLL at select genes. Loss of MLL binding led to changes in gene expression, differentiation, and apoptosis. Patient-derived xenograft (PDX) models derived from patients with either MLL-r acute myeloid leukemia or MLL-r acute lymphoblastic leukemia (ALL) showed dramatic reductions of leukemia burden when treated with VTP50469. Multiple mice engrafted with MLL-r ALL remained disease free for more than 1 year after treatment. These data support rapid translation of this approach to clinical trials.


Subject(s)
Chromatin/drug effects , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin/genetics , Gene Expression Regulation, Leukemic/genetics , Gene Rearrangement/drug effects , Gene Rearrangement/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Proto-Oncogene Proteins/genetics , Transcription Factors/drug effects , Transcription Factors/genetics
9.
Mol Cell ; 75(4): 683-699.e7, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31399344

ABSTRACT

Transcriptional regulation in eukaryotes occurs at promoter-proximal regions wherein transcriptionally engaged RNA polymerase II (Pol II) pauses before proceeding toward productive elongation. The role of chromatin in pausing remains poorly understood. Here, we demonstrate that the histone deacetylase SIRT6 binds to Pol II and prevents the release of the negative elongation factor (NELF), thus stabilizing Pol II promoter-proximal pausing. Genetic depletion of SIRT6 or its chromatin deficiency upon glucose deprivation causes intragenic enrichment of acetylated histone H3 at lysines 9 (H3K9ac) and 56 (H3K56ac), activation of cyclin-dependent kinase 9 (CDK9)-that phosphorylates NELF and the carboxyl terminal domain of Pol II-and enrichment of the positive transcription elongation factors MYC, BRD4, PAF1, and the super elongation factors AFF4 and ELL2. These events lead to increased expression of genes involved in metabolism, protein synthesis, and embryonic development. Our results identified SIRT6 as a Pol II promoter-proximal pausing-dedicated histone deacetylase.


Subject(s)
Promoter Regions, Genetic , RNA Polymerase II/metabolism , Sirtuins/metabolism , Transcription Elongation, Genetic , Acetylation , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Gene Deletion , Histones/genetics , Histones/metabolism , Humans , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/genetics , Sirtuins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
10.
PLoS Genet ; 15(4): e1008039, 2019 04.
Article in English | MEDLINE | ID: mdl-30970016

ABSTRACT

The SWI/SNF-family chromatin remodeling protein ATRX is a tumor suppressor in sarcomas, gliomas and other malignancies. Its loss of function facilitates the alternative lengthening of telomeres (ALT) pathway in tumor cells, while it also affects Polycomb repressive complex 2 (PRC2) silencing of its target genes. To further define the role of inactivating ATRX mutations in carcinogenesis, we knocked out atrx in our previously reported p53/nf1-deficient zebrafish line that develops malignant peripheral nerve sheath tumors and gliomas. Complete inactivation of atrx using CRISPR/Cas9 was lethal in developing fish and resulted in an alpha-thalassemia-like phenotype including reduced alpha-globin expression. In p53/nf1-deficient zebrafish neither peripheral nerve sheath tumors nor gliomas showed accelerated onset in atrx+/- fish, but these fish developed various tumors that were not observed in their atrx+/+ siblings, including epithelioid sarcoma, angiosarcoma, undifferentiated pleomorphic sarcoma and rare types of carcinoma. These cancer types are included in the AACR Genie database of human tumors associated with mutant ATRX, indicating that our zebrafish model reliably mimics a role for ATRX-loss in the early pathogenesis of these human cancer types. RNA-seq of p53/nf1- and p53/nf1/atrx-deficient tumors revealed that down-regulation of telomerase accompanied ALT-mediated lengthening of the telomeres in atrx-mutant samples. Moreover, inactivating mutations in atrx disturbed PRC2-target gene silencing, indicating a connection between ATRX loss and PRC2 dysfunction in cancer development.


Subject(s)
Sarcoma, Experimental/etiology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , X-linked Nuclear Protein/deficiency , X-linked Nuclear Protein/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Carcinogenesis/genetics , Carcinogenesis/metabolism , Disease Models, Animal , Erythropoiesis , Female , Gene Knockout Techniques , Globins/genetics , Humans , Loss of Function Mutation , Male , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Telomere Homeostasis/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
11.
Mol Cancer Ther ; 17(1): 254-263, 2018 01.
Article in English | MEDLINE | ID: mdl-29054988

ABSTRACT

Human tumor growth depends on rapidly dividing cancer cells driving population expansion. Even advanced tumors, however, contain slowly proliferating cancer cells for reasons that remain unclear. Here, we selectively disrupt the ability of rapidly proliferating cancer cells to spawn AKT1low daughter cells that are rare, slowly proliferating, tumor-initiating, and chemotherapy-resistant, using ß1-integrin activation and the AKT1-E17K-mutant oncoprotein as experimental tools in vivo Surprisingly, we find that selective depletion of AKT1low slow proliferators actually reduces the growth of a molecularly diverse panel of human cancer cell xenograft models without globally altering cell proliferation or survival in vivo Moreover, we find that unusual cancer patients with AKT1-E17K-mutant solid tumors also fail to produce AKT1low quiescent cancer cells and that this correlates with significantly prolonged survival after adjuvant treatment compared with other patients. These findings support a model whereby human solid tumor growth depends on not only rapidly proliferating cancer cells but also on the continuous production of AKT1low slow proliferators. Mol Cancer Ther; 17(1); 254-63. ©2017 AACR.


Subject(s)
Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Cell Transformation, Neoplastic , Female , HCT116 Cells , Heterografts , Humans , MCF-7 Cells , Mice , Neoplasms/pathology
12.
PLoS One ; 12(4): e0175300, 2017.
Article in English | MEDLINE | ID: mdl-28448494

ABSTRACT

TGF-ß is a cytokine thought to function as a tumor promoter in advanced malignancies. In this setting, TGF-ß increases cancer cell proliferation, survival, and migration, and orchestrates complex, pro-tumorigenic changes in the tumor microenvironment. Here, we find that in melanoma, integrin ß1-mediated TGF-ß activation may also produce tumor suppression via an altered host response. In the A375 human melanoma cell nu/nu xenograft model, we demonstrate that cell surface integrin ß1-activation increases TGF-ß activity, resulting in stromal activation, neo-angiogenesis and, unexpectedly for this nude mouse model, increase in the number of intra-tumoral CD8+ T lymphocytes within the tumor microenvironment. This is associated with attenuation of tumor growth and long-term survival benefit. Correspondingly, in human melanomas, TGF-ß1 correlates with integrin ß1/TGF-ß1 activation and the expression of markers for vasculature and stromal activation. Surprisingly, this integrin ß1/TGF-ß1 transcriptional footprint also correlates with the expression of markers for tumor-infiltrating lymphocytes, multiple immune checkpoints and regulatory pathways, and, importantly, better long-term survival of patients. These correlations are unique to melanoma, in that we do not observe similar associations between ß1 integrin/TGF-ß1 activation and better long-term survival in other human tumor types. These results suggest that activation of TGF-ß1 in melanoma may be associated with the generation of an anti-tumor host response that warrants further study.


Subject(s)
Antibodies/immunology , Integrin beta1/immunology , Integrin beta1/metabolism , Melanoma/immunology , Melanoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Extracellular Space/metabolism , Female , Melanoma/blood supply , Melanoma/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/immunology , Signal Transduction/immunology , Survival Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/immunology
13.
Oncotarget ; 8(13): 20572-20587, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28411283

ABSTRACT

Prognostic classifiers conceivably comprise biomarker genes that functionally contribute to the oncogenic and metastatic properties of cancer, but this has not been investigated systematically. The transcription factor Fra-1 not only has an essential role in breast cancer, but also drives the expression of a highly prognostic gene set. Here, we systematically perturbed the function of 31 individual Fra-1-dependent poor-prognosis genes and examined their impact on breast cancer growth in vivo. We find that stable shRNA depletion of each of nine individual signature genes strongly inhibits breast cancer growth and aggressiveness. Several factors within this nine-gene set regulate each other's expression, suggesting that together they form a network. The nine-gene set is regulated by estrogen, ERBB2 and EGF signaling, all established breast cancer factors. We also uncover three transcription factors, MYC, E2F1 and TP53, which act alongside Fra-1 at the core of this network. ChIP-Seq analysis reveals that a substantial number of genes are bound, and regulated, by all four transcription factors. The nine-gene set retains significant prognostic power and includes several potential therapeutic targets, including the bifunctional enzyme PAICS, which catalyzes purine biosynthesis. Depletion of PAICS largely cancelled breast cancer expansion, exemplifying a prognostic gene with breast cancer activity. Our data uncover a core genetic and prognostic network driving human breast cancer. We propose that pharmacological inhibition of components within this network, such as PAICS, may be used in conjunction with the Fra-1 prognostic classifier towards personalized management of poor prognosis breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Proto-Oncogene Proteins c-fos/genetics , Animals , Anoikis/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/enzymology , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Cell Line, Tumor , E2F1 Transcription Factor/genetics , Epidermal Growth Factor/genetics , Estrogens/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Peptide Synthases/genetics , Peptide Synthases/metabolism , Precision Medicine/methods , Prognosis , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/genetics , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
14.
Cell ; 165(6): 1401-1415, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27180906

ABSTRACT

Chromatin remodeling proteins are frequently dysregulated in human cancer, yet little is known about how they control tumorigenesis. Here, we uncover an epigenetic program mediated by the NAD(+)-dependent histone deacetylase Sirtuin 6 (SIRT6) that is critical for suppression of pancreatic ductal adenocarcinoma (PDAC), one of the most lethal malignancies. SIRT6 inactivation accelerates PDAC progression and metastasis via upregulation of Lin28b, a negative regulator of the let-7 microRNA. SIRT6 loss results in histone hyperacetylation at the Lin28b promoter, Myc recruitment, and pronounced induction of Lin28b and downstream let-7 target genes, HMGA2, IGF2BP1, and IGF2BP3. This epigenetic program defines a distinct subset with a poor prognosis, representing 30%-40% of human PDAC, characterized by reduced SIRT6 expression and an exquisite dependence on Lin28b for tumor growth. Thus, we identify SIRT6 as an important PDAC tumor suppressor and uncover the Lin28b pathway as a potential therapeutic target in a molecularly defined PDAC subset. PAPERCLIP.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , RNA-Binding Proteins/genetics , Sirtuins/genetics , Acetylation , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Female , Genes, ras , Histones/metabolism , Humans , Male , Mice , Mice, Knockout , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism
15.
Nat Biotechnol ; 34(4): 419-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26928769

ABSTRACT

Hundreds of genetically characterized cell lines are available for the discovery of genotype-specific cancer vulnerabilities. However, screening large numbers of compounds against large numbers of cell lines is currently impractical, and such experiments are often difficult to control. Here we report a method called PRISM that allows pooled screening of mixtures of cancer cell lines by labeling each cell line with 24-nucleotide barcodes. PRISM revealed the expected patterns of cell killing seen in conventional (unpooled) assays. In a screen of 102 cell lines across 8,400 compounds, PRISM led to the identification of BRD-7880 as a potent and highly specific inhibitor of aurora kinases B and C. Cell line pools also efficiently formed tumors as xenografts, and PRISM recapitulated the expected pattern of erlotinib sensitivity in vivo.


Subject(s)
DNA Barcoding, Taxonomic/methods , Drug Resistance, Neoplasm/genetics , Genotyping Techniques/methods , High-Throughput Nucleotide Sequencing/methods , Neoplasms/classification , Neoplasms/genetics , Animals , Cell Line, Tumor , Humans , Mice
16.
Nat Chem Biol ; 12(5): 317-23, 2016 May.
Article in English | MEDLINE | ID: mdl-26974814

ABSTRACT

Epigenetic gene regulation is a dynamic process orchestrated by chromatin-modifying enzymes. Many of these master regulators exert their function through covalent modification of DNA and histone proteins. Aberrant epigenetic processes have been implicated in the pathophysiology of multiple human diseases. Small-molecule inhibitors have been essential to advancing our understanding of the underlying molecular mechanisms of epigenetic processes. However, the resolution offered by small molecules is often insufficient to manipulate epigenetic processes with high spatiotemporal control. Here we present a generalizable approach, referred to as 'chemo-optical modulation of epigenetically regulated transcription' (COMET), enabling high-resolution, optical control of epigenetic mechanisms based on photochromic inhibitors of human histone deacetylases using visible light. COMET probes may be translated into new therapeutic strategies for diseases where conditional and selective epigenome modulation is required.


Subject(s)
Gene Expression Regulation/radiation effects , Light , Optogenetics/methods , Azo Compounds/chemistry , Epigenesis, Genetic , Humans , MCF-7 Cells , Models, Molecular , Molecular Structure
17.
Mol Cancer Ther ; 15(1): 142-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26637368

ABSTRACT

Small molecule inhibitors of AKT (v-akt murine thymoma viral oncogene homolog) signaling are being evaluated in patients with various cancer types, but have so far proven therapeutically disappointing for reasons that remain unclear. Here, we treat cancer cells with subtherapeutic doses of Akti-1/2, an allosteric small molecule AKT inhibitor, in order to experimentally model pharmacologic inhibition of AKT signaling in vitro. We then apply a combined RNA, protein, and metabolite profiling approach to develop an integrated, multiscale, molecular snapshot of this "AKT(low)" cancer cell state. We find that AKT-inhibited cancer cells suppress thousands of mRNA transcripts, and proteins related to the cell cycle, ribosome, and protein translation. Surprisingly, however, these AKT-inhibited cells simultaneously upregulate a host of other proteins and metabolites posttranscriptionally, reflecting activation of their endo-vesiculo-membrane system, secretion of inflammatory proteins, and elaboration of extracellular microvesicles. Importantly, these microvesicles enable rapidly proliferating cancer cells of various types to better withstand different stress conditions, including serum deprivation, hypoxia, or cytotoxic chemotherapy in vitro and xenografting in vivo. These findings suggest a model whereby cancer cells experiencing a partial inhibition of AKT signaling may actually promote the survival of neighbors through non-cell autonomous communication.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Metabolomics , Mice , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Nature ; 524(7565): 361-5, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26168401

ABSTRACT

Activation of cellular stress response pathways to maintain metabolic homeostasis is emerging as a critical growth and survival mechanism in many cancers. The pathogenesis of pancreatic ductal adenocarcinoma (PDA) requires high levels of autophagy, a conserved self-degradative process. However, the regulatory circuits that activate autophagy and reprogram PDA cell metabolism are unknown. Here we show that autophagy induction in PDA occurs as part of a broader transcriptional program that coordinates activation of lysosome biogenesis and function, and nutrient scavenging, mediated by the MiT/TFE family of transcription factors. In human PDA cells, the MiT/TFE proteins--MITF, TFE3 and TFEB--are decoupled from regulatory mechanisms that control their cytoplasmic retention. Increased nuclear import in turn drives the expression of a coherent network of genes that induce high levels of lysosomal catabolic function essential for PDA growth. Unbiased global metabolite profiling reveals that MiT/TFE-dependent autophagy-lysosome activation is specifically required to maintain intracellular amino acid pools. These results identify the MiT/TFE proteins as master regulators of metabolic reprogramming in pancreatic cancer and demonstrate that transcriptional activation of clearance pathways converging on the lysosome is a novel hallmark of aggressive malignancy.


Subject(s)
Autophagy/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic , Lysosomes/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Amino Acids/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Energy Metabolism , Female , Heterografts , Homeostasis , Humans , Lysosomes/genetics , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Transcription, Genetic
20.
Nat Cell Biol ; 17(5): 545-57, 2015 May.
Article in English | MEDLINE | ID: mdl-25915124

ABSTRACT

How embryonic stem cells (ESCs) commit to specific cell lineages and yield all cell types of a fully formed organism remains a major question. ESC differentiation is accompanied by large-scale histone and DNA modifications, but the relations between these epigenetic categories are not understood. Here we demonstrate the interplay between the histone deacetylase sirtuin 6 (SIRT6) and the ten-eleven translocation enzymes (TETs). SIRT6 targets acetylated histone H3 at Lys 9 and 56 (H3K9ac and H3K56ac), while TETs convert 5-methylcytosine into 5-hydroxymethylcytosine (5hmC). ESCs derived from Sirt6 knockout (S6KO) mice are skewed towards neuroectoderm development. This phenotype involves derepression of OCT4, SOX2 and NANOG, which causes an upregulation of TET-dependent production of 5hmC. Genome-wide analysis revealed neural genes marked with 5hmC in S6KO ESCs, thereby implicating TET enzymes in the neuroectoderm-skewed differentiation phenotype. We demonstrate that SIRT6 functions as a chromatin regulator safeguarding the balance between pluripotency and differentiation through Tet-mediated production of 5hmC.


Subject(s)
Cell Differentiation , Cell Lineage , Cytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/enzymology , Proto-Oncogene Proteins/metabolism , Sirtuins/metabolism , 5-Methylcytosine/analogs & derivatives , Acetylation , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Cytosine/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Embryonic Stem Cells/pathology , Embryonic Stem Cells/transplantation , Gene Expression Regulation, Developmental , Genotype , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/enzymology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Nanog Homeobox Protein , Neurogenesis , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , Proto-Oncogene Proteins/genetics , RNA Interference , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Sirtuins/deficiency , Sirtuins/genetics , Teratoma/enzymology , Teratoma/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...