Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
Add more filters

Country/Region as subject
Publication year range
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
2.
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.
Cell ; 153(4): 919-29, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23663786

ABSTRACT

Identification of somatic rearrangements in cancer genomes has accelerated through analysis of high-throughput sequencing data. However, characterization of complex structural alterations and their underlying mechanisms remains inadequate. Here, applying an algorithm to predict structural variations from short reads, we report a comprehensive catalog of somatic structural variations and the mechanisms generating them, using high-coverage whole-genome sequencing data from 140 patients across ten tumor types. We characterize the relative contributions of different types of rearrangements and their mutational mechanisms, find that ~20% of the somatic deletions are complex deletions formed by replication errors, and describe the differences between the mutational mechanisms in somatic and germline alterations. Importantly, we provide detailed reconstructions of the events responsible for loss of CDKN2A/B and gain of EGFR in glioblastoma, revealing that these alterations can result from multiple mechanisms even in a single genome and that both DNA double-strand breaks and replication errors drive somatic rearrangements.


Subject(s)
Algorithms , Genome, Human , Mutation , Neoplasms/genetics , Chromosome Aberrations , Genome-Wide Association Study , Glioblastoma/genetics , Humans , Neoplasms/pathology
5.
Cell ; 148(4): 651-63, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22341440

ABSTRACT

To assess telomerase as a cancer therapeutic target and determine adaptive mechanisms to telomerase inhibition, we modeled telomerase reactivation and subsequent extinction in T cell lymphomas arising in Atm(-/-) mice engineered with an inducible telomerase reverse transcriptase allele. Telomerase reactivation in the setting of telomere dysfunction enabled full malignant progression with alleviation of telomere dysfunction-induced checkpoints. These cancers possessed copy number alterations targeting key loci in human T cell lymphomagenesis. Upon telomerase extinction, tumor growth eventually slowed with reinstatement of telomere dysfunction-induced checkpoints, yet growth subsequently resumed as tumors acquired alternative lengthening of telomeres (ALT) and aberrant transcriptional networks centering on mitochondrial biology and oxidative defense. ALT+ tumors acquired amplification/overexpression of PGC-1ß, a master regulator of mitochondrial biogenesis and function, and they showed marked sensitivity to PGC-1ß or SOD2 knockdown. Genetic modeling of telomerase extinction reveals vulnerabilities that motivate coincidental inhibition of mitochondrial maintenance and oxidative defense mechanisms to enhance antitelomerase cancer therapy.


Subject(s)
Mitochondria , Telomerase/antagonists & inhibitors , Telomere Homeostasis , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Genes, cdc , Humans , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Mice , Mitochondria/metabolism , Neoplasm Invasiveness/pathology , Neoplasms/genetics , Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Telomerase/genetics , Telomerase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
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.
Cell ; 150(2): 251-63, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22817889

ABSTRACT

Despite recent insights into melanoma genetics, systematic surveys for driver mutations are challenged by an abundance of passenger mutations caused by carcinogenic UV light exposure. We developed a permutation-based framework to address this challenge, employing mutation data from intronic sequences to control for passenger mutational load on a per gene basis. Analysis of large-scale melanoma exome data by this approach discovered six novel melanoma genes (PPP6C, RAC1, SNX31, TACC1, STK19, and ARID2), three of which-RAC1, PPP6C, and STK19-harbored recurrent and potentially targetable mutations. Integration with chromosomal copy number data contextualized the landscape of driver mutations, providing oncogenic insights in BRAF- and NRAS-driven melanoma as well as those without known NRAS/BRAF mutations. The landscape also clarified a mutational basis for RB and p53 pathway deregulation in this malignancy. Finally, the spectrum of driver mutations provided unequivocal genomic evidence for a direct mutagenic role of UV light in melanoma pathogenesis.


Subject(s)
Genome-Wide Association Study , Melanoma/genetics , Mutagenesis , Ultraviolet Rays , Amino Acid Sequence , Cells, Cultured , Exome , Humans , Melanocytes/metabolism , Models, Molecular , Molecular Sequence Data , Proto-Oncogene Proteins B-raf/genetics , Sequence Alignment , rac1 GTP-Binding Protein/genetics
10.
Cell ; 145(6): 941-955, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663796

ABSTRACT

Melanocyte stem cells (McSCs) intimately interact with epithelial stem cells (EpSCs) in the hair follicle bulge and secondary hair germ (sHG). Together, they undergo activation and differentiation to regenerate pigmented hair. However, the mechanisms behind this coordinated stem cell behavior have not been elucidated. Here, we identified Wnt signaling as a key pathway that couples the behavior of the two stem cells. EpSCs and McSCs coordinately activate Wnt signaling at the onset of hair follicle regeneration within the sHG. Using genetic mouse models that specifically target either EpSCs or McSCs, we show that Wnt activation in McSCs drives their differentiation into pigment-producing melanocytes, while EpSC Wnt signaling not only dictates hair follicle formation but also regulates McSC proliferation during hair regeneration. Our data define a role for Wnt signaling in the regulation of McSCs and also illustrate a mechanism for regeneration of complex organs through collaboration between heterotypic stem cell populations.


Subject(s)
Epithelial Cells/cytology , Hair/physiology , Melanocytes/cytology , Pigmentation , Skin Physiological Phenomena , Stem Cells/cytology , Wnt Proteins/metabolism , Animals , Cell Differentiation , Hair/cytology , Hair Diseases/metabolism , Hair Diseases/pathology , Hair Follicle/cytology , Humans , Mice , Regeneration , Signal Transduction , Skin/cytology , beta Catenin/metabolism
11.
Cell ; 140(2): 180-2, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20141832

ABSTRACT

Targeted molecular therapies for cancer treatment have shown promise, but also have limitations. In this issue, Heidorn et al. (2010) find that a class of targeted molecular therapies with clinical effectiveness against one melanoma subtype may have adverse clinical effects in another.


Subject(s)
Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Antineoplastic Agents/adverse effects , Cell Line, Tumor , Humans , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism
12.
Cell ; 141(6): 994-1005, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20550935

ABSTRACT

DICER is a central regulator of microRNA maturation. However, little is known about mechanisms regulating its expression in development or disease. While profiling miRNA expression in differentiating melanocytes, two populations were observed: some upregulated at the pre-miRNA stage, and others upregulated as mature miRNAs (with stable pre-miRNA levels). Conversion of pre-miRNAs to fully processed miRNAs appeared to be dependent upon stimulation of DICER expression--an event found to occur via direct transcriptional targeting of DICER by the melanocyte master transcriptional regulator MITF. MITF binds and activates a conserved regulatory element upstream of DICER's transcriptional start site upon melanocyte differentiation. Targeted KO of DICER is lethal to melanocytes, at least partly via DICER-dependent processing of the pre-miRNA-17 approximately 92 cluster thus targeting BIM, a known proapoptotic regulator of melanocyte survival. These observations highlight a central mechanism underlying lineage-specific miRNA regulation which could exist for other cell types during development.


Subject(s)
Gene Expression Regulation , Melanocytes/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Ribonuclease III/metabolism , Transcription, Genetic , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Differentiation , Cell Survival , Cells, Cultured , Epidermal Cells , Gene Knockdown Techniques , Hair Follicle/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Up-Regulation
13.
Genes Dev ; 31(11): 1109-1121, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28698296

ABSTRACT

A key feature of high-grade serous ovarian carcinoma (HGSOC) is frequent amplification of the 3q26 locus harboring PRKC-ι (PRKCI). Here, we show that PRKCI is also expressed in early fallopian tube lesions, called serous tubal intraepithelial carcinoma. Transgenic mouse studies establish PRKCI as an ovarian cancer-specific oncogene. Mechanistically, we show that the oncogenic activity of PRKCI relates in part to the up-regulation of TNFα to promote an immune-suppressive tumor microenvironment characterized by an abundance of myeloid-derived suppressor cells and inhibition of cytotoxic T-cell infiltration. Furthermore, system-level and functional analyses identify YAP1 as a downstream effector in tumor progression. In human ovarian cancers, high PRKCI expression also correlates with high expression of TNFα and YAP1 and low infiltration of cytotoxic T cells. The PRKCI-YAP1 regulation of the tumor immunity provides a therapeutic strategy for highly lethal ovarian cancer.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Immune Tolerance/genetics , Isoenzymes/genetics , Isoenzymes/immunology , Ovarian Neoplasms/genetics , Protein Kinase C/genetics , Protein Kinase C/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Movement/genetics , Cytokines/genetics , Female , Humans , Isoenzymes/metabolism , Mice , Mice, Transgenic , Ovarian Neoplasms/immunology , Ovarian Neoplasms/physiopathology , Phosphoproteins/metabolism , Protein Kinase C/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/metabolism , YAP-Signaling Proteins
14.
Nature ; 542(7641): 362-366, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28178232

ABSTRACT

Malignant neoplasms evolve in response to changes in oncogenic signalling. Cancer cell plasticity in response to evolutionary pressures is fundamental to tumour progression and the development of therapeutic resistance. Here we determine the molecular and cellular mechanisms of cancer cell plasticity in a conditional oncogenic Kras mouse model of pancreatic ductal adenocarcinoma (PDAC), a malignancy that displays considerable phenotypic diversity and morphological heterogeneity. In this model, stochastic extinction of oncogenic Kras signalling and emergence of Kras-independent escaper populations (cells that acquire oncogenic properties) are associated with de-differentiation and aggressive biological behaviour. Transcriptomic and functional analyses of Kras-independent escapers reveal the presence of Smarcb1-Myc-network-driven mesenchymal reprogramming and independence from MAPK signalling. A somatic mosaic model of PDAC, which allows time-restricted perturbation of cell fate, shows that depletion of Smarcb1 activates the Myc network, driving an anabolic switch that increases protein metabolism and adaptive activation of endoplasmic-reticulum-stress-induced survival pathways. Increased protein turnover renders mesenchymal sub-populations highly susceptible to pharmacological and genetic perturbation of the cellular proteostatic machinery and the IRE1-α-MKK4 arm of the endoplasmic-reticulum-stress-response pathway. Specifically, combination regimens that impair the unfolded protein responses block the emergence of aggressive mesenchymal subpopulations in mouse and patient-derived PDAC models. These molecular and biological insights inform a potential therapeutic strategy for targeting aggressive mesenchymal features of PDAC.


Subject(s)
Mesoderm/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Endoplasmic Reticulum Stress/genetics , Female , Genes, myc , Genes, ras , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Male , Mesoderm/metabolism , Mice , Mosaicism , Oncogene Protein p55(v-myc)/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , SMARCB1 Protein/deficiency , SMARCB1 Protein/metabolism , Transcriptome/genetics , Gemcitabine
15.
Genes Dev ; 29(7): 732-45, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25838542

ABSTRACT

Glioblastoma multiforme (GBM) is a lethal, therapy-resistant brain cancer consisting of numerous tumor cell subpopulations, including stem-like glioma-initiating cells (GICs), which contribute to tumor recurrence following initial response to therapy. Here, we identified miR-182 as a regulator of apoptosis, growth, and differentiation programs whose expression level is correlated with GBM patient survival. Repression of Bcl2-like12 (Bcl2L12), c-Met, and hypoxia-inducible factor 2α (HIF2A) is of central importance to miR-182 anti-tumor activity, as it results in enhanced therapy susceptibility, decreased GIC sphere size, expansion, and stemness in vitro. To evaluate the tumor-suppressive function of miR-182 in vivo, we synthesized miR-182-based spherical nucleic acids (182-SNAs); i.e., gold nanoparticles covalently functionalized with mature miR-182 duplexes. Intravenously administered 182-SNAs penetrated the blood-brain/blood-tumor barriers (BBB/BTB) in orthotopic GBM xenografts and selectively disseminated throughout extravascular glioma parenchyma, causing reduced tumor burden and increased animal survival. Our results indicate that harnessing the anti-tumor activities of miR-182 via safe and robust delivery of 182-SNAs represents a novel strategy for therapeutic intervention in GBM.


Subject(s)
Apoptosis/genetics , Cell Differentiation/genetics , Glioblastoma/genetics , MicroRNAs/metabolism , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/physiopathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/physiopathology , Humans , Mice , Mice, SCID , MicroRNAs/administration & dosage , MicroRNAs/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Survival Analysis
16.
Genes Dev ; 27(13): 1462-72, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23796897

ABSTRACT

With the advent of high-throughput sequencing technologies, much progress has been made in the identification of somatic structural rearrangements in cancer genomes. However, characterization of the complex alterations and their associated mechanisms remains inadequate. Here, we report a comprehensive analysis of whole-genome sequencing and DNA copy number data sets from The Cancer Genome Atlas to relate chromosomal alterations to imbalances in DNA dosage and describe the landscape of intragenic breakpoints in glioblastoma multiforme (GBM). Gene length, guanine-cytosine (GC) content, and local presence of a copy number alteration were closely associated with breakpoint susceptibility. A dense pattern of repeated focal amplifications involving the murine double minute 2 (MDM2)/cyclin-dependent kinase 4 (CDK4) oncogenes and associated with poor survival was identified in 5% of GBMs. Gene fusions and rearrangements were detected concomitant within the breakpoint-enriched region. At the gene level, we noted recurrent breakpoints in genes such as apoptosis regulator FAF1. Structural alterations of the FAF1 gene disrupted expression and led to protein depletion. Restoration of the FAF1 protein in glioma cell lines significantly increased the FAS-mediated apoptosis response. Our study uncovered a previously underappreciated genomic mechanism of gene deregulation that can confer growth advantages on tumor cells and may generate cancer-specific vulnerabilities in subsets of GBM.


Subject(s)
Chromosome Breakage , Glioblastoma/genetics , Glioblastoma/mortality , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , DNA Copy Number Variations/genetics , Gene Fusion/genetics , Gene Rearrangement/genetics , Genomic Instability/genetics , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins , Mice , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Survival Analysis
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(11): 1131-55, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22661227

ABSTRACT

Melanoma is often considered one of the most aggressive and treatment-resistant human cancers. It is a disease that, due to the presence of melanin pigment, was accurately diagnosed earlier than most other malignancies and that has been subjected to countless therapeutic strategies. Aside from early surgical resection, no therapeutic modality has been found to afford a high likelihood of curative outcome. However, discoveries reported in recent years have revealed a near avalanche of breakthroughs in the melanoma field-breakthroughs that span fundamental understanding of the molecular basis of the disease all the way to new therapeutic strategies that produce unquestionable clinical benefit. These discoveries have been born from the successful fruits of numerous researchers working in many-sometimes-related, although also distinct-biomedical disciplines. Discoveries of frequent mutations involving BRAF(V600E), developmental and oncogenic roles for the microphthalmia-associated transcription factor (MITF) pathway, clinical efficacy of BRAF-targeted small molecules, and emerging mechanisms underlying resistance to targeted therapeutics represent just a sample of the findings that have created a striking inflection in the quest for clinically meaningful progress in the melanoma field.


Subject(s)
Melanoma/drug therapy , Melanoma/genetics , Animals , Cell Cycle , Humans , Melanoma/diagnosis , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction
19.
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
20.
Genes Dev ; 26(8): 756-84, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22508724

ABSTRACT

Glioblastoma is both the most common and lethal primary malignant brain tumor. Extensive multiplatform genomic characterization has provided a higher-resolution picture of the molecular alterations underlying this disease. These studies provide the emerging view that "glioblastoma" represents several histologically similar yet molecularly heterogeneous diseases, which influences taxonomic classification systems, prognosis, and therapeutic decisions.


Subject(s)
Brain Neoplasms/classification , Brain Neoplasms/genetics , Glioblastoma/classification , Glioblastoma/genetics , Brain Neoplasms/pathology , Gene Expression Profiling , Genes, Tumor Suppressor , Genomics , Glioblastoma/pathology , Humans , Neovascularization, Pathologic/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL