Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 25
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cancer Res ; 82(20): 3637-3649, 2022 10 17.
Article de Anglais | MEDLINE | ID: mdl-35904353

RÉSUMÉ

Immune-checkpoint blockade therapy has been successfully applied to many cancers, particularly tumors that harbor a high mutational burden and consequently express a high abundance of neoantigens. However, novel approaches are needed to improve the efficacy of immunotherapy for treating tumors that lack a high load of classic genetically derived neoantigens. Recent discoveries of broad classes of nongenetically encoded and inducible neoepitopes open up new avenues for therapeutic development to enhance sensitivity to immunotherapies. In this review, we discuss recent work on neoantigen discovery, with an emphasis on novel classes of noncanonical neoepitopes.


Sujet(s)
Antigènes néoplasiques , Tumeurs , Antigènes néoplasiques/génétique , Humains , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Mutation , Tumeurs/génétique , Tumeurs/thérapie
3.
Trends Genet ; 38(11): 1123-1133, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35641342

RÉSUMÉ

Programmed ribosomal frameshifting (PRF) is a key mechanism that viruses use to generate essential proteins for replication, and as a means of regulating gene expression. PRF generally involves recoding signals or frameshift stimulators to elevate the occurrence of frameshifting at shift-prone 'slippery' sequences. Given its essential role in viral replication, targeting PRF was envisioned as an attractive tool to block viral infection. However, in contrast to controlled-PRF mechanisms, recent studies have shown that ribosomes of many human cancer cell types are prone to frameshifting upon amino acid shortage; thus, these cells are deemed to be sloppy. The resulting products of a sloppy frameshift at the 'hungry' codons are aberrant proteins the degradation and display of which at the cell surface can trigger T cell activation. In this review, we address recent discoveries in ribosomal frameshifting and their functional consequences for the proteome in human cancer cells.


Sujet(s)
Décalage ribosomique , Protéome , Acides aminés/génétique , Codon/génétique , Décalage ribosomique/génétique , Humains , Protéome/génétique , Ribosomes/génétique , Ribosomes/métabolisme
4.
Nature ; 603(7902): 721-727, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35264796

RÉSUMÉ

Activated T cells secrete interferon-γ, which triggers intracellular tryptophan shortage by upregulating the indoleamine 2,3-dioxygenase 1 (IDO1) enzyme1-4. Here we show that despite tryptophan depletion, in-frame protein synthesis continues across tryptophan codons. We identified tryptophan-to-phenylalanine codon reassignment (W>F) as the major event facilitating this process, and pinpointed tryptophanyl-tRNA synthetase (WARS1) as its source. We call these W>F peptides 'substitutants' to distinguish them from genetically encoded mutants. Using large-scale proteomics analyses, we demonstrate W>F substitutants to be highly abundant in multiple cancer types. W>F substitutants were enriched in tumours relative to matching adjacent normal tissues, and were associated with increased IDO1 expression, oncogenic signalling and the tumour-immune microenvironment. Functionally, W>F substitutants can impair protein activity, but also expand the landscape of antigens presented at the cell surface to activate T cell responses. Thus, substitutants are generated by an alternative decoding mechanism with potential effects on gene function and tumour immunoreactivity.


Sujet(s)
Tryptophane-tRNA ligase , Tryptophane , Codon/métabolisme , Indoleamine-pyrrole 2,3,-dioxygenase/génétique , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Interféron gamma , Tumeurs/immunologie , Phénylalanine , Lymphocytes T , Tryptophane/métabolisme , Tryptophane 2,3-dioxygenase/génétique , Tryptophane 2,3-dioxygenase/métabolisme , Tryptophane-tRNA ligase/génétique , Tryptophane-tRNA ligase/métabolisme
5.
Mol Cell ; 81(22): 4709-4721.e9, 2021 11 18.
Article de Anglais | MEDLINE | ID: mdl-34562372

RÉSUMÉ

mRNA translation is a highly conserved and tightly controlled mechanism for protein synthesis. Despite protein quality control mechanisms, amino acid shortage in melanoma induces aberrant proteins by ribosomal frameshifting. The extent and the underlying mechanisms related to this phenomenon are yet unknown. Here, we show that tryptophan depletion-induced ribosomal frameshifting is a widespread phenomenon in cancer. We termed this event sloppiness and strikingly observed its association with MAPK pathway hyperactivation. Sloppiness is stimulated by RAS activation in primary cells, suppressed by pharmacological inhibition of the oncogenic MAPK pathway in sloppy cells, and restored in cells with acquired resistance to MAPK pathway inhibition. Interestingly, sloppiness causes aberrant peptide presentation at the cell surface, allowing recognition and specific killing of drug-resistant cancer cells by T lymphocytes. Thus, while oncogenes empower cancer progression and aggressiveness, they also expose a vulnerability by provoking the production of aberrant peptides through sloppiness.


Sujet(s)
Tumeurs/génétique , Oncogènes , Biosynthèse des protéines , ARN messager/métabolisme , Lymphocytes T/cytologie , Animaux , Carcinogenèse , Membrane cellulaire/métabolisme , Évolution de la maladie , Résistance aux médicaments antinéoplasiques , Mutation avec décalage du cadre de lecture , Décalage ribosomique , Humains , Immunothérapie/méthodes , Système de signalisation des MAP kinases , Mélanome/métabolisme , Souris , Tumeurs/métabolisme , Peptides/composition chimique , Inhibiteurs de protéines kinases , Ribosomes/métabolisme , Lymphocytes T/métabolisme , Tryptophane/composition chimique , Tryptophane/métabolisme
6.
Genome Biol ; 22(1): 54, 2021 01 29.
Article de Anglais | MEDLINE | ID: mdl-33514403

RÉSUMÉ

BACKGROUND: Frequent activation of the co-transcriptional factor YAP is observed in a large number of solid tumors. Activated YAP associates with enhancer loci via TEAD4-DNA-binding protein and stimulates cancer aggressiveness. Although thousands of YAP/TEAD4 binding-sites are annotated, their functional importance is unknown. Here, we aim at further identification of enhancer elements that are required for YAP functions. RESULTS: We first apply genome-wide ChIP profiling of YAP to systematically identify enhancers that are bound by YAP/TEAD4. Next, we implement a genetic approach to uncover functions of YAP/TEAD4-associated enhancers, demonstrate its robustness, and use it to reveal a network of enhancers required for YAP-mediated proliferation. We focus on EnhancerTRAM2, as its target gene TRAM2 shows the strongest expression-correlation with YAP activity in nearly all tumor types. Interestingly, TRAM2 phenocopies the YAP-induced cell proliferation, migration, and invasion phenotypes and correlates with poor patient survival. Mechanistically, we identify FSTL-1 as a major direct client of TRAM2 that is involved in these phenotypes. Thus, TRAM2 is a key novel mediator of YAP-induced oncogenic proliferation and cellular invasiveness. CONCLUSIONS: YAP is a transcription co-factor that binds to thousands of enhancer loci and stimulates tumor aggressiveness. Using unbiased functional approaches, we dissect YAP enhancer network and characterize TRAM2 as a novel mediator of cellular proliferation, migration, and invasion. Our findings elucidate how YAP induces cancer aggressiveness and may assist diagnosis of cancer metastasis.


Sujet(s)
Carcinogenèse/génétique , Éléments activateurs (génétique) , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Animaux , Sites de fixation , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Protéines de liaison à l'ADN/métabolisme , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Glycoprotéines membranaires/composition chimique , Souris , Souris de lignée NOD , Souris SCID , Facteurs de transcription à domaine TEA/génétique , Facteurs de transcription à domaine TEA/métabolisme , Facteurs de transcription/métabolisme , Transcriptome
7.
Nature ; 590(7845): 332-337, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33328638

RÉSUMÉ

Extensive tumour inflammation, which is reflected by high levels of infiltrating T cells and interferon-γ (IFNγ) signalling, improves the response of patients with melanoma to checkpoint immunotherapy1,2. Many tumours, however, escape by activating cellular pathways that lead to immunosuppression. One such mechanism is the production of tryptophan metabolites along the kynurenine pathway by the enzyme indoleamine 2,3-dioxygenase 1 (IDO1), which is induced by IFNγ3-5. However, clinical trials using inhibition of IDO1 in combination with blockade of the PD1 pathway in patients with melanoma did not improve the efficacy of treatment compared to PD1 pathway blockade alone6,7, pointing to an incomplete understanding of the role of IDO1 and the consequent degradation of tryptophan in mRNA translation and cancer progression. Here we used ribosome profiling in melanoma cells to investigate the effects of prolonged IFNγ treatment on mRNA translation. Notably, we observed accumulations of ribosomes downstream of tryptophan codons, along with their expected stalling at the tryptophan codon. This suggested that ribosomes bypass tryptophan codons in the absence of tryptophan. A detailed examination of these tryptophan-associated accumulations of ribosomes-which we term 'W-bumps'-showed that they were characterized by ribosomal frameshifting events. Consistently, reporter assays combined with proteomic and immunopeptidomic analyses demonstrated the induction of ribosomal frameshifting, and the generation and presentation of aberrant trans-frame peptides at the cell surface after treatment with IFNγ. Priming of naive T cells from healthy donors with aberrant peptides induced peptide-specific T cells. Together, our results suggest that IDO1-mediated depletion of tryptophan, which is induced by IFNγ, has a role in the immune recognition of melanoma cells by contributing to diversification of the peptidome landscape.


Sujet(s)
Présentation d'antigène , Mutation avec décalage du cadre de lecture , Mélanome/immunologie , Peptides/génétique , Peptides/immunologie , Biosynthèse des protéines/immunologie , Lymphocytes T/immunologie , Lignée cellulaire , Codon/génétique , Décalage ribosomique/effets des médicaments et des substances chimiques , Décalage ribosomique/génétique , Décalage ribosomique/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Humains , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Interféron gamma/immunologie , Interféron gamma/pharmacologie , Mélanome/anatomopathologie , Peptides/composition chimique , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Biosynthèse des protéines/génétique , Protéome , Ribosomes/effets des médicaments et des substances chimiques , Ribosomes/métabolisme , Tryptophane/déficit , Tryptophane/génétique , Tryptophane/métabolisme
8.
Mol Syst Biol ; 16(9): e9443, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32960509

RÉSUMÉ

Tumor relapse as a consequence of chemotherapy resistance is a major clinical challenge in advanced stage breast tumors. To identify processes associated with poor clinical outcome, we took a mass spectrometry-based proteomic approach and analyzed a breast cancer cohort of 113 formalin-fixed paraffin-embedded samples. Proteomic profiling of matched tumors before and after chemotherapy, and tumor-adjacent normal tissue, all from the same patients, allowed us to define eight patterns of protein level changes, two of which correlate to better chemotherapy response. Supervised analysis identified two proteins of proline biosynthesis pathway, PYCR1 and ALDH18A1, that were significantly associated with resistance to treatment based on pattern dominance. Weighted gene correlation network analysis of post-treatment samples revealed that these proteins are associated with tumor relapse and affect patient survival. Functional analysis showed that knockdown of PYCR1 reduced invasion and migration capabilities of breast cancer cell lines. PYCR1 knockout significantly reduced tumor burden and increased drug sensitivity of orthotopically injected ER-positive tumor in vivo, thus emphasizing the role of PYCR1 in resistance to chemotherapy.


Sujet(s)
Tumeurs du sein/métabolisme , Tumeurs du sein/thérapie , Traitement néoadjuvant , Protéomique , Tumeurs du sein/anatomopathologie , Systèmes CRISPR-Cas/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Cycle citrique , Femelle , Réseaux de régulation génique , Humains , Invasion tumorale , Protéines tumorales/métabolisme , Pronostic , Cartes d'interactions protéiques , Pyrroline carboxylate reductases/métabolisme , Récidive , Analyse de survie ,
9.
EMBO J ; 38(21): e102147, 2019 10 04.
Article de Anglais | MEDLINE | ID: mdl-31523835

RÉSUMÉ

L-asparaginase (ASNase) serves as an effective drug for adolescent acute lymphoblastic leukemia. However, many clinical trials indicated severe ASNase toxicity in patients with solid tumors, with resistant mechanisms not well understood. Here, we took a functional genetic approach and identified SLC1A3 as a novel contributor to ASNase resistance in cancer cells. In combination with ASNase, SLC1A3 inhibition caused cell cycle arrest or apoptosis, and myriads of metabolic vulnerabilities in tricarboxylic acid (TCA) cycle, urea cycle, nucleotides biosynthesis, energy production, redox homeostasis, and lipid biosynthesis. SLC1A3 is an aspartate and glutamate transporter, mainly expressed in brain tissues, but high expression levels were also observed in some tumor types. Here, we demonstrate that ASNase stimulates aspartate and glutamate consumptions, and their refilling through SLC1A3 promotes cancer cell proliferation. Lastly, in vivo experiments indicated that SLC1A3 expression promoted tumor development and metastasis while negating the suppressive effects of ASNase by fueling aspartate, glutamate, and glutamine metabolisms despite of asparagine shortage. Altogether, our findings identify a novel role for SLC1A3 in ASNase resistance and suggest that restrictive aspartate and glutamate uptake might improve ASNase efficacy with solid tumors.


Sujet(s)
Asparaginase/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Transporteur-1 d'acides aminés excitateurs/métabolisme , Tumeurs/traitement médicamenteux , Animaux , Antinéoplasiques/pharmacologie , Apoptose , Systèmes CRISPR-Cas , Prolifération cellulaire , Transporteur-1 d'acides aminés excitateurs/antagonistes et inhibiteurs , Transporteur-1 d'acides aminés excitateurs/génétique , Humains , Souris , Souris de lignée NOD , Souris SCID , Tumeurs/enzymologie , Tumeurs/anatomopathologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Mol Cancer Ther ; 18(4): 762-770, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30872379

RÉSUMÉ

Small cell lung cancer (SCLC) is generally regarded as very difficult to treat, mostly due to the development of metastases early in the disease and a quick relapse with resistant disease. SCLC patients initially show a good response to treatment with the DNA damaging agents cisplatin and etoposide. This is, however, quickly followed by the development of resistant disease, which urges the development of novel therapies for this type of cancer. In this study, we set out to compile a comprehensive overview of the vulnerabilities of SCLC. A functional genome-wide screen where all individual genes were knocked out was performed to identify novel vulnerabilities of SCLC. By analysis of the knockouts that were lethal to these cancer cells, we identified several processes to be synthetic vulnerabilities in SCLC. We were able to validate the vulnerability to inhibition of the replication stress response machinery by use of Chk1 and ATR inhibitors. Strikingly, SCLC cells were more sensitive to these inhibitors than nontransformed cells. In addition, these inhibitors work synergistically with either etoposide and cisplatin, where the interaction is largest with the latter. ATR inhibition by VE-822 treatment in combination with cisplatin also outperforms the combination of cisplatin with etoposide in vivo Altogether, our study uncovered a critical dependence of SCLC on the replication stress response and urges the validation of ATR inhibitors in combination with cisplatin in a clinical setting.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Cisplatine/usage thérapeutique , Isoxazoles/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Pyrazines/usage thérapeutique , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Cellules A549 , Animaux , Antinéoplasiques/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Protéine-9 associée à CRISPR/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Checkpoint kinase 1/antagonistes et inhibiteurs , Cisplatine/administration et posologie , Altération de l'ADN/effets des médicaments et des substances chimiques , Synergie des médicaments , Étoposide/administration et posologie , Étoposide/usage thérapeutique , Humains , Isoxazoles/administration et posologie , Isoxazoles/pharmacologie , Souris , Souris de lignée BALB C , Souris nude , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/pharmacologie , Pyrazines/administration et posologie , Pyrazines/pharmacologie , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
11.
J Proteomics ; 196: 106-119, 2019 03 30.
Article de Anglais | MEDLINE | ID: mdl-30710758

RÉSUMÉ

Molecular markers are urgently needed to select non-small cell lung cancer (NSCLC) patients most likely to benefit from platinum-based chemotherapies. Of particular interest are proteins that can be found in biofluids like sputum for non-invasive detection. Therefore, we profiled the secretomes of 6 NSCLC cell lines with varying IC50-values for cisplatin, using label-free GeLC-MS/MS-based proteomics. Out of a total dataset of 2610 proteins, 304 proteins showed significant differences in expression levels between cisplatin sensitive and insensitive cell lines. Functional data mining revealed that the secretion of typically extracellular factors was associated with a higher sensitivity towards cisplatin, while cisplatin insensitivity correlated with increased secretion of theoretically intra-cellular proteins. Stringent statistical analysis and quantitative filtering yielded 58 biomarker candidates, 34 of which could be detected in clinical biofluids of lung cancer patients such as sputum using label-free LC-MS/MS-based proteomics. To assess performance of these biofluid biomarker candidates, we correlated protein expression with patient survival using a publically available clinical gene expression data set (GSE14814). We thus identified 3 top candidates with potential predictive value in determining cisplatin response (UGGT1, COL6A1 and MAP4) for future development as non-invasive biomarkers to guide treatment decisions. SIGNIFICANCE: Platinum-based chemotherapies are still the standard of care for NSCLC and other lung cancer types in the clinic today. However, due to chemoresistance, many patients suffer from the toxic side effects of these treatments without gaining any benefit in terms of survival. To date, no molecular biomarkers are available to predict clinical outcome of platinum-based chemotherapy. Because proteins present the functional read-out of genetic, epigenetic and translational events in the cell, a protein test is likely to be particularly suitable for response prediction. Of high relevance are proteins that are shed or secreted from cells, for example at primary tumor sites, and can be found in easily accessible biofluids like sputum for non-invasive detection. Here, we report the proteome profiling of the conditioned media (secretomes) of a panel of NSCLC cell lines in relation to cisplatin IC50 values, as a pre-clinical model, and of patient sputum as a clinical, lung cancer relevant biofluid. Using this approach in conjunction with exploration of the predictive potential in a transcriptome lung cancer patient dataset, we reveal biofluid biomarker candidates that, with further validation, may be used for non-invasive cisplatin response prediction in the future.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Carcinome pulmonaire non à petites cellules , Cisplatine/métabolisme , Tumeurs du poumon , Protéines tumorales/métabolisme , Protéomique , Expectoration/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Lignée cellulaire tumorale , Femelle , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Mâle , Valeur prédictive des tests
12.
Cancer Lett ; 436: 87-95, 2018 11 01.
Article de Anglais | MEDLINE | ID: mdl-30145202

RÉSUMÉ

Breast cancer is the most prevalent type of malignancy in women with ∼1.7 million new cases diagnosed annually, of which the majority express ERα (ESR1), a ligand-dependent transcription factor. Genome-wide chromatin binding maps suggest that ERα may control the expression of thousands of genes, posing a great challenge in identifying functional targets. Recently, we developed a CRISPR-Cas9 functional genetic screening approach to identify enhancers required for ERα-positive breast cancer cell proliferation. We validated several candidates, including CUTE, a putative ERα-responsive enhancer located in the first intron of CUEDC1 (CUE-domain containing protein). Here, we show that CUTE controls CUEDC1 expression, and that this interaction is essential for ERα-mediated cell proliferation. Moreover, ectopic expression of CUEDC1, but not a CUE-domain mutant, rescues the defects in CUTE activity. Finally, CUEDC1 expression correlates positively with ERα in breast cancer. Thus, CUEDC1 is a functional target gene of ERα and is required for breast cancer cell proliferation.


Sujet(s)
Tumeurs du sein/génétique , Prolifération cellulaire/génétique , Récepteur alpha des oestrogènes/génétique , Régulation de l'expression des gènes tumoraux , Protéines et peptides de signalisation intracellulaire/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Systèmes CRISPR-Cas , Lignée cellulaire tumorale , Éléments activateurs (génétique)/génétique , Récepteur alpha des oestrogènes/métabolisme , Femelle , Cellules HEK293 , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Cellules MCF-7
13.
EMBO Rep ; 17(11): 1516-1531, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27702988

RÉSUMÉ

Historically, cancers have been treated with chemotherapeutics aimed to have profound effects on tumor cells with only limited effects on normal tissue. This approach was followed by the development of small-molecule inhibitors that can target oncogenic pathways critical for the survival of tumor cells. The clinical targeting of these so-called oncogene addictions, however, is in many instances hampered by the outgrowth of resistant clones. More recently, the proper functioning of non-mutated genes has been shown to enhance the survival of many cancers, a phenomenon called non-oncogene addiction. In the current review, we will focus on the distinct non-oncogenic addictions found in cancer cells, including synthetic lethal interactions, the underlying stress phenotypes, and arising therapeutic opportunities.


Sujet(s)
Mutation , Tumeurs/thérapie , Animaux , Apoptose , Lignée cellulaire tumorale , Humains , Souris , Thérapie moléculaire ciblée , Tumeurs/génétique , Tumeurs/métabolisme , Oncogènes , Stress physiologique , Mutations synthétiques létales
14.
Genes Dev ; 29(14): 1447-62, 2015 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-26220992

RÉSUMÉ

Lung cancer is the leading cause of cancer deaths, with small cell lung cancer (SCLC) representing the most aggressive subtype. Standard treatments have not changed in decades, and the 5-year survival rate has remained <7%. Genomic analyses have identified key driver mutations of SCLC that were subsequently validated in animal models of SCLC. To provide better treatment options, a deeper understanding of the cellular and molecular mechanisms underlying SCLC initiation, progression, metastasis, and acquisition of resistance is required. In this review, we describe the genetic landscape of SCLC, features of the cell of origin, and targeted therapeutic approaches.


Sujet(s)
Tumeurs du poumon/génétique , Tumeurs du poumon/thérapie , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/thérapie , Animaux , Antinéoplasiques/usage thérapeutique , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques , Humains , Tumeurs du poumon/anatomopathologie , Mutation , Carcinome pulmonaire à petites cellules/mortalité , Carcinome pulmonaire à petites cellules/anatomopathologie
15.
Mol Cancer Ther ; 14(6): 1434-44, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25808837

RÉSUMÉ

Lung cancer is the most common cancer worldwide and on top of that has a very poor prognosis, which is reflected by a 5-year survival rate of 5% to 15%. Radiotherapy is an integral part of most treatment regimens for this type of tumor, often combined with radiosensitizing cytotoxic drugs. In this study, we identified many genes that could potentially be exploited for targeted radiosensitization using a genome-wide siRNA screen in non-small cell lung cancer (NSCLC) cells. The screen identified 433 siRNAs that potentially sensitize lung cancer cells to radiation. Validation experiments showed that knockdown of expression of Forkhead box M1 (FOXM1) or microtubule-associated serine/threonine kinase-like (MASTL) indeed causes radiosensitization in a panel of NSCLC cells. Strikingly, this effect was not observed in primary human fibroblasts, suggesting that the observed radiosensitization is specific for cancer cells. Phosphoproteomics analyses with and without irradiation showed that a number of cell-cycle-related proteins were significantly less phosphorylated after MASTL knockdown in comparison to the control, while there were no changes in the levels of phosphorylation of DNA damage response proteins. Subsequent analyses showed that MASTL knockdown cells respond differently to radiation, with a significantly shortened G2-M phase arrest and defects in cytokinesis, which are followed by a cell-cycle arrest. In summary, we have identified many potential therapeutic targets that could be used for radiosensitization of NSCLC cells, with MASTL being a very promising and druggable target to combine with radiotherapy.


Sujet(s)
Carcinome pulmonaire non à petites cellules/radiothérapie , Facteurs de transcription Forkhead/génétique , Génome humain/génétique , Tumeurs du poumon/radiothérapie , Protéines associées aux microtubules/génétique , Protein-Serine-Threonine Kinases/génétique , Interférence par ARN , Animaux , Technique de Western , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Cycle cellulaire/génétique , Cycle cellulaire/effets des radiations , Lignée cellulaire tumorale , Régulation négative/génétique , Régulation négative/effets des radiations , Protéine M1 à motif en tête de fourche , Facteurs de transcription Forkhead/métabolisme , Rayons gamma , Régulation de l'expression des gènes tumoraux/génétique , Régulation de l'expression des gènes tumoraux/effets des radiations , Cellules HEK293 , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Souris nude , Microscopie confocale , Protéines associées aux microtubules/métabolisme , Phosphorylation/génétique , Phosphorylation/effets des radiations , Protein-Serine-Threonine Kinases/métabolisme , Petit ARN interférent/génétique , RT-PCR , Analyse de survie , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Oral Oncol ; 49(6): 560-6, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23578372

RÉSUMÉ

OBJECTIVES: Infection with the human papillomavirus (HPV) is an important risk factor for development of head and neck squamous cell carcinoma (HNSCC). Strikingly, HPV-positive HNSCCs have a more favorable prognosis than their HPV-negative counterparts. The current study was designed to explain this favorable prognosis of HPV-positive HNSCC. MATERIALS AND METHODS: This was performed by investigating the response of four HPV-positive and fourteen HPV-negative HNSCC cell lines to cisplatin, cetuximab and radiation. RESULTS: Analysis of the responses of this cell line panel indicated that HPV-positive cells are more resistant to cisplatin treatment than the HPV-negative HNSCCs, whereas the response to radiation and cetuximab did not differ. CONCLUSIONS: The current study suggests that the favorable prognosis for patients with HPV-positive HNSCC does not seem to be related to an intrinsic sensitivity of these tumor cells to chemotherapy or radiation in vitro.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Carcinome épidermoïde/thérapie , Tumeurs de la tête et du cou/thérapie , Papillomaviridae/isolement et purification , Radiothérapie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/virologie , Lignée cellulaire tumorale , Association thérapeutique , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/virologie , Humains , Radiotolérance
17.
Clin Cancer Res ; 19(8): 1994-2003, 2013 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-23444224

RÉSUMÉ

PURPOSE: Despite continuous improvement of treatment regimes, the mortality rates for non-small cell lung cancer (NSCLC) and head and neck squamous cell carcinoma (HNSCC) remain disappointingly high and novel anticancer agents are urgently awaited. EXPERIMENTAL DESIGN: We combined the data from genome-wide siRNA screens on tumor cell lethality in a lung and a head and neck cancer cell line. RESULTS: We identified 71 target genes that seem essential for the survival of both cancer types. We identified a cluster of 20 genes that play an important role during G2-M phase transition, underlining the importance of this cell-cycle checkpoint for tumor cell survival. Five genes from this cluster (CKAP5, KPNB1, RAN, TPX2, and KIF11) were evaluated in more detail and have been shown to be essential for tumor cell survival in both tumor types, but most particularly in HNSCC. Phenotypes that were observed following siRNA-mediated knockdown of KIF11 (kinesin family member 11) were reproduced by inhibition of KIF11 using the small-molecule inhibitor ispinesib (SB-715992). We showed that ispinesib induces a G2 arrest, causes aberrant chromosome segregation, and induces cell death in HNSCC in vitro, whereas primary keratinocytes are less sensitive. Furthermore, growth of HNSCC cells engrafted in immunodeficient mice was significantly inhibited after ispinesib treatment. CONCLUSION: This study identified a wide array of druggable genes for both lung and head and neck cancer. In particular, multiple genes involved in the G2-M checkpoint were shown to be essential for tumor cell survival, indicating their potential as anticancer targets.


Sujet(s)
Carcinome pulmonaire non à petites cellules/génétique , Carcinome épidermoïde/génétique , Tumeurs de la tête et du cou/génétique , Tumeurs du poumon/génétique , Interférence par ARN , Animaux , Benzamides/pharmacologie , Technique de Western , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/prévention et contrôle , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Cellules cultivées , Relation dose-effet des médicaments , Points de contrôle de la phase G2 du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle de la phase G2 du cycle cellulaire/génétique , Gènes essentiels/génétique , Étude d'association pangénomique/méthodes , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/prévention et contrôle , Humains , Kinésine/génétique , Kinésine/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Quinazolines/pharmacologie , RT-PCR , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Caryophérines bêta/génétique , Caryophérines bêta/métabolisme , Protéine G ran/génétique , Protéine G ran/métabolisme
18.
FEBS J ; 276(19): 5447-55, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19682069

RÉSUMÉ

Several biological functions in mammals are regulated in a circadian fashion. The molecular mechanisms orchestrating these circadian rhythms have been unravelled. The biological clock, with its core transcriptional unit Bmal1/CLOCK, is composed of several self-sustaining feedback loops. In this study, we describe another mechanism impinging on the core components of the circadian clock. Using a forward genetic screen, we identified the miR-192/194 cluster as a potent inhibitor of the entire Period gene family. In accordance, the exogenous expression of miR-192/194 leads to an altered circadian rhythm. Thus, our results have uncovered a new mechanism for the control of the circadian clock at the post-transcriptional level.


Sujet(s)
Rythme circadien/génétique , Protéines et peptides de signalisation intracellulaire/génétique , microARN/génétique , Famille multigénique , Régions 3' non traduites , Animaux , Séquence nucléotidique , Lignée cellulaire , Amorces ADN/génétique , Gènes rapporteurs , Protéines à fluorescence verte/génétique , Humains , Souris , Mutation , Protéines circadiennes Period
19.
Cancer Res ; 68(14): 5795-802, 2008 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-18632633

RÉSUMÉ

Inactivation of the adenomatous polyposis coli (APC) gene is a major initiating event in colorectal tumorigenesis. Most of the mutations in APC generate premature stop codons leading to truncated proteins that have lost beta-catenin binding sites. APC-free beta-catenin stimulates the Wnt signaling pathway, leading to active transcription of target genes. In the current study, we describe a novel mechanism for APC regulation. We show that miR-135a&b target the 3' untranslated region of APC, suppress its expression, and induce downstream Wnt pathway activity. Interestingly, we find a considerable up-regulation of miR-135a&b in colorectal adenomas and carcinomas, which significantly correlated with low APC mRNA levels. This genetic interaction is also preserved in full-blown cancer cell lines expressing miR-135a&b, regardless of the mutational status of APC. Thus, our results uncover a miRNA-mediated mechanism for the control of APC expression and Wnt pathway activity, and suggest its contribution to colorectal cancer pathogenesis.


Sujet(s)
Protéine de la polypose adénomateuse colique/génétique , Protéine de la polypose adénomateuse colique/physiologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Régulation de l'expression des gènes tumoraux , microARN/génétique , Régions 3' non traduites , Lignée cellulaire , Lignée cellulaire tumorale , Tumeurs colorectales/anatomopathologie , Analyse de mutations d'ADN , Analyse de profil d'expression de gènes , Cellules HeLa , Humains , Modèles biologiques , Régions promotrices (génétique) , Transduction du signal , Protéines de type Wingless/métabolisme
20.
Nat Cell Biol ; 10(2): 202-10, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18193036

RÉSUMÉ

MicroRNAs (miRNAs) are single-stranded, noncoding RNAs that are important in many biological processes. Although the oncogenic and tumour-suppressive functions of several miRNAs have been characterized, the role of miRNAs in mediating tumour metastasis was addressed only recently and still remains largely unexplored. To identify potential metastasis-promoting miRNAs, we set up a genetic screen using a non-metastatic, human breast tumour cell line that was transduced with a miRNA-expression library and subjected to a trans-well migration assay. We found that human miR-373 and miR-520c stimulated cancer cell migration and invasion in vitro and in vivo, and that certain cancer cell lines depend on endogenous miR-373 activity to migrate efficiently. Mechanistically, the migration phenotype of miR-373 and miR-520c can be explained by suppression of CD44. We found significant upregulation of miR-373 in clinical breast cancer metastasis samples that correlated inversely with CD44 expression. Taken together, our findings indicate that miRNAs are involved in tumour migration and invasion, and implicate miR-373 and miR-520c as metastasis-promoting miRNAs.


Sujet(s)
Mouvement cellulaire/physiologie , microARN/physiologie , Invasion tumorale/génétique , Métastase tumorale/génétique , Animaux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Tests de migration cellulaire , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Femelle , Humains , Antigènes CD44/génétique , Antigènes CD44/métabolisme , Métastase lymphatique , Mâle , Souris , Souris SCID , microARN/biosynthèse , Invasion tumorale/anatomopathologie , Métastase tumorale/anatomopathologie , Transplantation tumorale , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Transplantation hétérologue
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...