Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 52
Filtrer
1.
Sci Rep ; 14(1): 13227, 2024 06 09.
Article de Anglais | MEDLINE | ID: mdl-38851782

RÉSUMÉ

There are hundreds of genes typically overexpressed in breast cancer cells and it's often assumed that their overexpression contributes to cancer progression. However, the precise proportion of these overexpressed genes contributing to tumorigenicity remains unclear. To address this gap, we undertook a comprehensive screening of a diverse set of seventy-two genes overexpressed in breast cancer. This systematic screening evaluated their potential for inducing malignant transformation and, concurrently, assessed their impact on breast cancer cell proliferation and viability. Select genes including ALDH3B1, CEACAM5, IL8, PYGO2, and WWTR1, exhibited pronounced activity in promoting tumor formation and establishing gene dependencies critical for tumorigenicity. Subsequent investigations revealed that CEACAM5 overexpression triggered the activation of signaling pathways involving ß-catenin, Cdk4, and mTOR. Additionally, it conferred a growth advantage independent of exogenous insulin in defined medium and facilitated spheroid expansion by inducing multiple layers of epithelial cells while preserving a hollow lumen. Furthermore, the silencing of CEACAM5 expression synergized with tamoxifen-induced growth inhibition in breast cancer cells. These findings underscore the potential of screening overexpressed genes for both oncogenic drivers and tumor dependencies to expand the repertoire of therapeutic targets for breast cancer treatment.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Transduction du signal , Oncogènes , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Tamoxifène/pharmacologie , Animaux , Molécules d'adhérence cellulaire/génétique , Molécules d'adhérence cellulaire/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Transformation cellulaire néoplasique/génétique
2.
Neuro Oncol ; 25(3): 471-481, 2023 03 14.
Article de Anglais | MEDLINE | ID: mdl-36044040

RÉSUMÉ

BACKGROUND: Glioblastoma is the most common and aggressive primary brain tumor. Large-scale sequencing initiatives have cataloged its mutational landscape in hopes of elucidating mechanisms driving this deadly disease. However, a major bottleneck in harnessing this data for new therapies is deciphering "driver" and "passenger" events amongst the vast volume of information. METHODS: We utilized an autochthonous, in vivo screening approach to identify driver, EGFR variants. RNA-Seq identified unique molecular signatures of mouse gliomas across these variants, which only differ by a single amino acid change. In particular, we identified alterations to lipid metabolism, which we further validated through an unbiased lipidomics screen. RESULTS: Our screen identified A289I as the most potent EGFR variant, which has previously not been characterized. One of the mechanisms through which A289I promotes gliomagenesis is to alter cellular triacylglycerides through MTTP. Knockout of Mttp in mouse gliomas, reduces gliomagenesis in multiple models. CONCLUSIONS: EGFR variants that differ by a single amino acid residue differentially promote gliomagenesis. Among the identified mechanism that drives glioma growth include lipid metabolism through MTTP. Understanding triacylglyceride accumulation may present a prospective therapeutic pathway for this deadly disease.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Gliome , Souris , Animaux , Glioblastome/anatomopathologie , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Souris knockout , Gliome/traitement médicamenteux , Mutation , Tumeurs du cerveau/traitement médicamenteux
3.
Sci Adv ; 8(6): eabm2382, 2022 02 11.
Article de Anglais | MEDLINE | ID: mdl-35138907

RÉSUMÉ

Fusion genes represent a class of attractive therapeutic targets. Thousands of fusion genes have been identified in patients with cancer, but the functional consequences and therapeutic implications of most of these remain largely unknown. Here, we develop a functional genomic approach that consists of efficient fusion reconstruction and sensitive cell viability and drug response assays. Applying this approach, we characterize ~100 fusion genes detected in patient samples of The Cancer Genome Atlas, revealing a notable fraction of low-frequency fusions with activating effects on tumor growth. Focusing on those in the RTK-RAS pathway, we identify a number of activating fusions that can markedly affect sensitivity to relevant drugs. Last, we propose an integrated, level-of-evidence classification system to prioritize gene fusions systematically. Our study reiterates the urgent clinical need to incorporate similar functional genomic approaches to characterize gene fusions, thereby maximizing the utility of gene fusions for precision oncology.


Sujet(s)
Tumeurs , Fusion de gènes , Génome , Génomique , Humains , Tumeurs/génétique , Médecine de précision
4.
Cancer Res ; 81(10): 2703-2713, 2021 05 15.
Article de Anglais | MEDLINE | ID: mdl-33782098

RÉSUMÉ

Germline POT1 mutations are found in a spectrum of cancers and confer increased risk. Recently, we identified a series of novel germline POT1 mutations that predispose carrier families to the development of glioma. Despite these strong associations, how these glioma-associated POT1 mutations contribute to glioma tumorigenesis remains undefined. Here we show that POT1-G95C increases proliferation in glioma-initiating cells in vitro and in progenitor populations in the developing brain. In a native mouse model of glioma, loss of Pot1a/b resulted in decreased survival in females compared with males. These findings were corroborated in human glioma, where low POT1 expression correlated with decreased survival in females. Transcriptomic and IHC profiling of Pot1a/b-deficient glioma revealed that tumors in females exhibited decreased expression of immune markers and increased expression of cell-cycle signatures. Similar sex-dependent trends were observed in human gliomas that had low expression of POT1. Together, our studies demonstrate context-dependent functions for POT1 mutation or loss in driving progenitor proliferation in the developing brain and sexual dimorphism in glioma. SIGNIFICANCE: This study shows that manipulation of POT1 expression in glioma has sex-specific effects on tumorigenesis and associated immune signatures.


Sujet(s)
Carcinogenèse/anatomopathologie , Prolifération cellulaire , Gliome/anatomopathologie , Mutation , Caractères sexuels , Protéines télomériques/métabolisme , Transcriptome , Animaux , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Cycle cellulaire , Femelle , Gliome/génétique , Gliome/immunologie , Gliome/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Complexe shelterine , Protéines télomériques/génétique
5.
Oncogene ; 39(37): 5979-5994, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32753652

RÉSUMÉ

Non-small cell lung cancer (NSCLC) is the deadliest form of cancer worldwide, due in part to its proclivity to metastasize. Identifying novel drivers of invasion and metastasis holds therapeutic potential for the disease. We conducted a gain-of-function invasion screen, which identified two separate hits, IMPAD1 and KDELR2, as robust, independent drivers of lung cancer invasion and metastasis. Given that IMPAD1 and KDELR2 are known to be localized to the ER-Golgi pathway, we studied their common mechanism of driving in vitro invasion and in vivo metastasis and demonstrated that they enhance Golgi-mediated function and secretion. Therapeutically inhibiting matrix metalloproteases (MMPs) suppressed both IMPAD1- and KDELR2-mediated invasion. The hits from this unbiased screen and the mechanistic validation highlight Golgi function as one of the key cellular features altered during invasion and metastasis.


Sujet(s)
Appareil de Golgi/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Phosphoric monoester hydrolases/génétique , Protéines du transport vésiculaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Évolution de la maladie , Technique d'immunofluorescence , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du poumon/anatomopathologie , Matrix metalloproteinases/métabolisme , Invasion tumorale , Phosphoric monoester hydrolases/métabolisme , Protéines du transport vésiculaire/métabolisme
6.
Elife ; 92020 04 09.
Article de Anglais | MEDLINE | ID: mdl-32270762

RÉSUMÉ

The melanoma-associated antigen family A (MAGEA) antigens are expressed in a wide variety of malignant tumors but not in adult somatic cells, rendering them attractive targets for cancer immunotherapy. Here we show that a number of cancer-associated MAGEA mutants that undergo proteasome-dependent degradation in vitro could negatively impact their utility as immunotherapeutic targets. Importantly, in pancreatic ductal adenocarcinoma cell models, MAGEA6 suppresses macroautophagy (autophagy). The inhibition of autophagy is released upon MAGEA6 degradation, which can be induced by nutrient deficiency or by acquisition of cancer-associated mutations. Using xenograft mouse models, we demonstrated that inhibition of autophagy is critical for tumor initiation whereas reinstitution of autophagy as a consequence of MAGEA6 degradation contributes to tumor progression. These findings could inform cancer immunotherapeutic strategies for targeting MAGEA antigens and provide mechanistic insight into the divergent roles of MAGEA6 during pancreatic cancer initiation and progression.


Sujet(s)
Antigènes néoplasiques/physiologie , Autophagie/physiologie , Carcinome du canal pancréatique/étiologie , Protéines tumorales/physiologie , Tumeurs du pancréas/étiologie , Animaux , Antigènes néoplasiques/génétique , Autophagie/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Évolution de la maladie , Femelle , Humains , Souris , Mutation , Protéines tumorales/génétique , Tumeurs du pancréas/anatomopathologie , Proteasome endopeptidase complex/physiologie
7.
Nature ; 578(7793): 166-171, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31996845

RÉSUMÉ

Glioblastoma is a universally lethal form of brain cancer that exhibits an array of pathophysiological phenotypes, many of which are mediated by interactions with the neuronal microenvironment1,2. Recent studies have shown that increases in neuronal activity have an important role in the proliferation and progression of glioblastoma3,4. Whether there is reciprocal crosstalk between glioblastoma and neurons remains poorly defined, as the mechanisms that underlie how these tumours remodel the neuronal milieu towards increased activity are unknown. Here, using a native mouse model of glioblastoma, we develop a high-throughput in vivo screening platform and discover several driver variants of PIK3CA. We show that tumours driven by these variants have divergent molecular properties that manifest in selective initiation of brain hyperexcitability and remodelling of the synaptic constituency. Furthermore, secreted members of the glypican (GPC) family are selectively expressed in these tumours, and GPC3 drives gliomagenesis and hyperexcitability. Together, our studies illustrate the importance of functionally interrogating diverse tumour phenotypes driven by individual, yet related, variants and reveal how glioblastoma alters the neuronal microenvironment.


Sujet(s)
Tumeurs du cerveau/enzymologie , Phosphatidylinositol 3-kinases de classe I/métabolisme , Glioblastome/enzymologie , Animaux , Tumeurs du cerveau/anatomopathologie , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Phosphatidylinositol 3-kinases de classe I/composition chimique , Phosphatidylinositol 3-kinases de classe I/génétique , Modèles animaux de maladie humaine , Glioblastome/anatomopathologie , Glypicanes/métabolisme , Souris
8.
Sci Transl Med ; 12(527)2020 01 22.
Article de Anglais | MEDLINE | ID: mdl-31969487

RÉSUMÉ

Heightened secretion of protumorigenic effector proteins is a feature of malignant cells. Yet, the molecular underpinnings and therapeutic implications of this feature remain unclear. Here, we identify a chromosome 1q region that is frequently amplified in diverse cancer types and encodes multiple regulators of secretory vesicle biogenesis and trafficking, including the Golgi-dedicated enzyme phosphatidylinositol (PI)-4-kinase IIIß (PI4KIIIß). Molecular, biochemical, and cell biological studies show that PI4KIIIß-derived PI-4-phosphate (PI4P) synthesis enhances secretion and accelerates lung adenocarcinoma progression by activating Golgi phosphoprotein 3 (GOLPH3)-dependent vesicular release from the Golgi. PI4KIIIß-dependent secreted factors maintain 1q-amplified cancer cell survival and influence prometastatic processes in the tumor microenvironment. Disruption of this functional circuitry in 1q-amplified cancer cells with selective PI4KIIIß antagonists induces apoptosis and suppresses tumor growth and metastasis. These results support a model in which chromosome 1q amplifications create a dependency on PI4KIIIß-dependent secretion for cancer cell survival and tumor progression.


Sujet(s)
Adénocarcinome pulmonaire/métabolisme , Chromosomes humains de la paire 1/métabolisme , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Adénocarcinome pulmonaire/génétique , Animaux , Chromosomes humains de la paire 1/génétique , Test ELISA , Appareil de Golgi/métabolisme , Humains , Techniques in vitro , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris , Phosphotransferases (Alcohol Group Acceptor)/antagonistes et inhibiteurs , Microtomographie aux rayons X
9.
Nat Commun ; 11(1): 486, 2020 01 24.
Article de Anglais | MEDLINE | ID: mdl-31980632

RÉSUMÉ

Alternative splicing has been shown to causally contribute to the epithelial-mesenchymal transition (EMT) and tumor metastasis. However, the scope of splicing factors that govern alternative splicing in these processes remains largely unexplored. Here we report the identification of A-Kinase Anchor Protein (AKAP8) as a splicing regulatory factor that impedes EMT and breast cancer metastasis. AKAP8 not only is capable of inhibiting splicing activity of the EMT-promoting splicing regulator hnRNPM through protein-protein interaction, it also directly binds to RNA and alters splicing outcomes. Genome-wide analysis shows that AKAP8 promotes an epithelial cell state splicing program. Experimental manipulation of an AKAP8 splicing target CLSTN1 revealed that splice isoform switching of CLSTN1 is crucial for EMT. Moreover, AKAP8 expression and the alternative splicing of CLSTN1 predict breast cancer patient survival. Together, our work demonstrates the essentiality of RNA metabolism that impinges on metastatic breast cancer.


Sujet(s)
Protéines d'ancrage aux protéines kinases A/métabolisme , Épissage alternatif/génétique , Transition épithélio-mésenchymateuse/génétique , Protéines d'ancrage aux protéines kinases A/antagonistes et inhibiteurs , Protéines d'ancrage aux protéines kinases A/génétique , Animaux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Protéines de liaison au calcium/génétique , Protéines de liaison au calcium/métabolisme , Lignée cellulaire tumorale , Femelle , Techniques de knock-down de gènes , Cellules HCT116 , Cellules HEK293 , Ribonucléoprotéine nucléaire hétérogène du groupe M/génétique , Ribonucléoprotéine nucléaire hétérogène du groupe M/métabolisme , Hétérogreffes , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/prévention et contrôle , Tumeurs du poumon/secondaire , Souris , Souris nude , Motifs et domaines d'intéraction protéique , ARN/génétique , ARN/métabolisme , Protéines de liaison à l'ARN/antagonistes et inhibiteurs , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme
10.
Cell ; 176(1-2): 127-143.e24, 2019 01 10.
Article de Anglais | MEDLINE | ID: mdl-30633903

RÉSUMÉ

DNA damage provokes mutations and cancer and results from external carcinogens or endogenous cellular processes. However, the intrinsic instigators of endogenous DNA damage are poorly understood. Here, we identify proteins that promote endogenous DNA damage when overproduced: the DNA "damage-up" proteins (DDPs). We discover a large network of DDPs in Escherichia coli and deconvolute them into six function clusters, demonstrating DDP mechanisms in three: reactive oxygen increase by transmembrane transporters, chromosome loss by replisome binding, and replication stalling by transcription factors. Their 284 human homologs are over-represented among known cancer drivers, and their RNAs in tumors predict heavy mutagenesis and a poor prognosis. Half of the tested human homologs promote DNA damage and mutation when overproduced in human cells, with DNA damage-elevating mechanisms like those in E. coli. Our work identifies networks of DDPs that provoke endogenous DNA damage and may reveal DNA damage-associated functions of many human known and newly implicated cancer-promoting proteins.


Sujet(s)
Altération de l'ADN/génétique , Altération de l'ADN/physiologie , Réparation de l'ADN/physiologie , Protéines bactériennes/métabolisme , Instabilité des chromosomes/physiologie , Réplication de l'ADN/physiologie , Protéines de liaison à l'ADN/métabolisme , Escherichia coli/métabolisme , Instabilité du génome , Humains , Protéines de transport membranaire/physiologie , Mutagenèse , Mutation , Facteurs de transcription/métabolisme
11.
Nat Commun ; 9(1): 4583, 2018 11 02.
Article de Anglais | MEDLINE | ID: mdl-30389923

RÉSUMÉ

Activation of platelet-derived growth factor receptor alpha (PDGFRA) by genomic aberrations contributes to tumor progression in several tumor types. In this study, we characterize 16 novel PDGFRA mutations identified from different tumor types and identify three previously uncharacterized activating mutations that promote cell survival and proliferation. PDGFRA Y288C, an extracellular domain mutation, is primarily high mannose glycosylated consistent with trapping in the endoplasmic reticulum (ER). Strikingly, PDGFRA Y288C is constitutively dimerized and phosphorylated in the absence of ligand suggesting that trapping in the ER or aberrant glycosylation is sufficient for receptor activation. Importantly, PDGFRA Y288C induces constitutive phosphorylation of Akt, ERK1/2, and STAT3. PDGFRA Y288C is resistant to PDGFR inhibitors but sensitive to PI3K/mTOR and MEK inhibitors consistent with pathway activation results. Our findings further highlight the importance of characterizing functional consequences of individual mutations for precision medicine.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Espace extracellulaire/composition chimique , Thérapie moléculaire ciblée , Mutation/génétique , Récepteur au PDGF alpha/composition chimique , Récepteur au PDGF alpha/génétique , Animaux , Lignée cellulaire , Prolifération cellulaire , Réticulum endoplasmique/métabolisme , Glycosylation , Appareil de Golgi/métabolisme , Humains , Souris , Phénotype , Domaines protéiques , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal
12.
Proc Natl Acad Sci U S A ; 115(42): 10666-10671, 2018 10 16.
Article de Anglais | MEDLINE | ID: mdl-30266789

RÉSUMÉ

Scientific progress depends on formulating testable hypotheses informed by the literature. In many domains, however, this model is strained because the number of research papers exceeds human readability. Here, we developed computational assistance to analyze the biomedical literature by reading PubMed abstracts to suggest new hypotheses. The approach was tested experimentally on the tumor suppressor p53 by ranking its most likely kinases, based on all available abstracts. Many of the best-ranked kinases were found to bind and phosphorylate p53 (P value = 0.005), suggesting six likely p53 kinases so far. One of these, NEK2, was studied in detail. A known mitosis promoter, NEK2 was shown to phosphorylate p53 at Ser315 in vitro and in vivo and to functionally inhibit p53. These bona fide validations of text-based predictions of p53 phosphorylation, and the discovery of an inhibitory p53 kinase of pharmaceutical interest, suggest that automated reasoning using a large body of literature can generate valuable molecular hypotheses and has the potential to accelerate scientific discovery.


Sujet(s)
, Kinases apparentées à NIMA/métabolisme , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs , Protéine p53 suppresseur de tumeur/métabolisme , Cellules HCT116 , Cellules HEK293 , Humains , Kinases apparentées à NIMA/génétique , Traitement du langage naturel , Phosphorylation , PubMed , Protéine p53 suppresseur de tumeur/génétique
13.
Nat Commun ; 9(1): 3919, 2018 09 25.
Article de Anglais | MEDLINE | ID: mdl-30254248

RÉSUMÉ

In addition to genomic mutations, RNA editing is another major mechanism creating sequence variations in proteins by introducing nucleotide changes in mRNA sequences. Deregulated RNA editing contributes to different types of human diseases, including cancers. Here we report that peptides generated as a consequence of RNA editing are indeed naturally presented by human leukocyte antigen (HLA) molecules. We provide evidence that effector CD8+ T cells specific for edited peptides derived from cyclin I are present in human tumours and attack tumour cells that are presenting these epitopes. We show that subpopulations of cancer patients have increased peptide levels and that levels of edited RNA correlate with peptide copy numbers. These findings demonstrate that RNA editing extends the classes of HLA presented self-antigens and that these antigens can be recognised by the immune system.


Sujet(s)
Antigènes néoplasiques/immunologie , Épitopes/immunologie , Système immunitaire/immunologie , Tumeurs/immunologie , Édition des ARN/immunologie , Présentation d'antigène/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Cellules cultivées , Cycline I/génétique , Cycline I/immunologie , Cycline I/métabolisme , Cytotoxicité immunologique/immunologie , Antigènes HLA/immunologie , Humains , Tumeurs/génétique , Tumeurs/métabolisme , Peptides/génétique , Peptides/immunologie , Peptides/métabolisme , Protéogénomique/méthodes
15.
Nat Commun ; 9(1): 2732, 2018 07 16.
Article de Anglais | MEDLINE | ID: mdl-30013058

RÉSUMÉ

Genetic aberrations driving pro-oncogenic and pro-metastatic activity remain an elusive target in the quest of precision oncology. To identify such drivers, we use an animal model of KRAS-mutant lung adenocarcinoma to perform an in vivo functional screen of 217 genetic aberrations selected from lung cancer genomics datasets. We identify 28 genes whose expression promoted tumor metastasis to the lung in mice. We employ two tools for examining the KRAS-dependence of genes identified from our screen: 1) a human lung cell model containing a regulatable mutant KRAS allele and 2) a lentiviral system permitting co-expression of DNA-barcoded cDNAs with Cre recombinase to activate a mutant KRAS allele in the lungs of mice. Mechanistic evaluation of one gene, GATAD2B, illuminates its role as a dual activity gene, promoting both pro-tumorigenic and pro-metastatic activities in KRAS-mutant lung cancer through interaction with c-MYC and hyperactivation of the c-MYC pathway.


Sujet(s)
Adénocarcinome pulmonaire/génétique , Facteurs de transcription GATA/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon/génétique , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes p21(ras)/génétique , Adénocarcinome pulmonaire/mortalité , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/thérapie , Animaux , Lignée cellulaire tumorale , Femelle , Facteurs de transcription GATA/antagonistes et inhibiteurs , Facteurs de transcription GATA/métabolisme , Vecteurs génétiques/composition chimique , Vecteurs génétiques/métabolisme , Tests de criblage à haut débit , Humains , Integrases/génétique , Integrases/métabolisme , Lentivirus/génétique , Lentivirus/métabolisme , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/thérapie , Souris , Souris nude , Métastase tumorale , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes p21(ras)/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Protéines de répression , Transduction du signal , Analyse de survie , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Nat Commun ; 9(1): 2731, 2018 07 16.
Article de Anglais | MEDLINE | ID: mdl-30013069

RÉSUMÉ

Metastatic lung cancer is the leading cause of cancer-associated mortality worldwide, therefore necessitating novel approaches to identify specific genetic drivers for lung cancer progression and metastasis. We recently performed an in vivo gain-of-function genetic screen to identify driver genes of lung cancer metastasis. In the study reported here, we identify TMEM106B as a primary robust driver of lung cancer metastasis. Ectopic expression of TMEM106B could significantly promote the synthesis of enlarged vesicular lysosomes that are laden with elevated levels of active cathepsins. In a TFEB-dependent manner, TMEM106B could modulate the expression of lysosomal genes of the coordinated lysosomal expression and regulation (CLEAR) pathway in lung cancer cells and patient samples. We also demonstrate that TMEM106B-induced lysosomes undergo calcium-dependent exocytosis, thereby releasing active lysosomal cathepsins necessary for TMEM106B-mediated cancer cell invasion and metastasis in vivo, which could be therapeutically prevented by pharmacological inhibition of cathepsins. Further, in TCGA LUAD data sets, 19% of patients show elevated expression of TMEM106B, which predicts for poor disease-free and overall-survival.


Sujet(s)
Adénocarcinome pulmonaire/génétique , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Cathepsines/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon/génétique , Protéines membranaires/génétique , Protéines de tissu nerveux/génétique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/mortalité , Adénocarcinome pulmonaire/anatomopathologie , Animaux , Antinéoplasiques/pharmacologie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Calcium/métabolisme , Cathepsines/antagonistes et inhibiteurs , Cathepsines/métabolisme , Lignée cellulaire tumorale , Inhibiteurs de la cystéine protéinase/pharmacologie , Exocytose , Humains , Leucine/analogues et dérivés , Leucine/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Lysosomes/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Protéines membranaires/métabolisme , Souris , Métastase tumorale , Protéines de tissu nerveux/métabolisme , Pronostic , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Protéolyse , Transduction du signal , Analyse de survie , Tests d'activité antitumorale sur modèle de xénogreffe
17.
PLoS One ; 13(5): e0196939, 2018.
Article de Anglais | MEDLINE | ID: mdl-29738578

RÉSUMÉ

Identification of cancer driver mutations is critical for advancing cancer research and personalized medicine. Due to inter-tumor genetic heterogeneity, many driver mutations occur at low frequencies, which make it challenging to distinguish them from passenger mutations. Here, we show that a novel Bayesian hierarchical modeling approach, named rDriver can achieve enhanced prediction accuracy by identifying mutations that not only have high functional impact scores but also are associated with systemic variation in gene expression levels. In examining 3,080 tumor samples from 8 cancer types in The Cancer Genome Atlas, rDriver predicted 1,389 driver mutations. Compared with existing tools, rDriver identified more low frequency mutations associated with lineage specific functional properties, timing of occurrence and patient survival. Evaluation of rDriver predictions using engineered cell-line models resulted in a positive predictive value of 0.94 in PIK3CA genes. Our study highlights the importance of integrating multi-omic data in predicting cancer driver mutations and provides a statistically rigorous solution for cancer target discovery and development.


Sujet(s)
Biologie informatique , Mutation/génétique , Tumeurs/génétique , Algorithmes , Théorème de Bayes , Bases de données génétiques , Humains , Médecine de précision
18.
NPJ Breast Cancer ; 4: 9, 2018.
Article de Anglais | MEDLINE | ID: mdl-29736411

RÉSUMÉ

Tumor cells disseminate early in tumor development making metastasis-prevention strategies difficult. Identifying proteins that promote the outgrowth of disseminated tumor cells may provide opportunities for novel therapeutic strategies. Despite multiple studies demonstrating that the mesenchymal-to-epithelial transition (MET) is critical for metastatic colonization, key regulators that initiate this transition remain unknown. We serially passaged lung metastases from a primary triple negative breast cancer xenograft to the mammary fat pads of recipient mice to enrich for gene expression changes that drive metastasis. An unbiased transcriptomic signature of potential metastatic drivers was generated, and a high throughput gain-of-function screen was performed in vivo to validate candidates. Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) was identified as a metastatic driver. CEACAM5 overproduction enriched for an epithelial gene expression pattern and facilitated tumor outgrowth at metastatic sites. Tissues from patients with metastatic breast cancer confirmed elevated levels of CEACAM5 in lung metastases relative to breast tumors, and an inverse correlation between CEACAM5 and the mesenchymal marker vimentin was demonstrated. Thus, CEACAM5 facilitates tumor outgrowth at metastatic sites by promoting MET, warranting its investigation as a therapeutic target and biomarker of aggressiveness in breast cancer.

19.
Cancer Cell ; 33(5): 817-828.e7, 2018 05 14.
Article de Anglais | MEDLINE | ID: mdl-29706454

RÉSUMÉ

Adenosine (A) to inosine (I) RNA editing introduces many nucleotide changes in cancer transcriptomes. However, due to the complexity of post-transcriptional regulation, the contribution of RNA editing to proteomic diversity in human cancers remains unclear. Here, we performed an integrated analysis of TCGA genomic data and CPTAC proteomic data. Despite limited site diversity, we demonstrate that A-to-I RNA editing contributes to proteomic diversity in breast cancer through changes in amino acid sequences. We validate the presence of editing events at both RNA and protein levels. The edited COPA protein increases proliferation, migration, and invasion of cancer cells in vitro. Our study suggests an important contribution of A-to-I RNA editing to protein diversity in cancer and highlights its translational potential.


Sujet(s)
Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Protéomique/méthodes , Édition des ARN , Adénosine/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Bases de données génétiques , Régulation de l'expression des gènes tumoraux , Humains , Inosine/génétique , Analyse de séquence d'ARN , Spectrométrie de masse en tandem
20.
Cancer Cell ; 33(3): 450-462.e10, 2018 03 12.
Article de Anglais | MEDLINE | ID: mdl-29533785

RÉSUMÉ

The functional impact of the vast majority of cancer somatic mutations remains unknown, representing a critical knowledge gap for implementing precision oncology. Here, we report the development of a moderate-throughput functional genomic platform consisting of efficient mutant generation, sensitive viability assays using two growth factor-dependent cell models, and functional proteomic profiling of signaling effects for select aberrations. We apply the platform to annotate >1,000 genomic aberrations, including gene amplifications, point mutations, indels, and gene fusions, potentially doubling the number of driver mutations characterized in clinically actionable genes. Further, the platform is sufficiently sensitive to identify weak drivers. Our data are accessible through a user-friendly, public data portal. Our study will facilitate biomarker discovery, prediction algorithm improvement, and drug development.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Mutation/génétique , Tumeurs/diagnostic , Tumeurs/génétique , Algorithmes , Génomique , Séquençage nucléotidique à haut débit/méthodes , Humains , Médecine de précision , Protéomique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE