Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Cancer ; 3(7): 821-836, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35883003

RÉSUMÉ

Oncogenic alterations in human epidermal growth factor receptor 2 (HER2) occur in approximately 2% of patients with non-small cell lung cancer and predominantly affect the tyrosine kinase domain and cluster in exon 20 of the ERBB2 gene. Most clinical-grade tyrosine kinase inhibitors are limited by either insufficient selectivity against wild-type (WT) epidermal growth factor receptor (EGFR), which is a major cause of dose-limiting toxicity or by potency against HER2 exon 20 mutant variants. Here we report the discovery of covalent tyrosine kinase inhibitors that potently inhibit HER2 exon 20 mutants while sparing WT EGFR, which reduce tumor cell survival and proliferation in vitro and result in regressions in preclinical xenograft models of HER2 exon 20 mutant non-small cell lung cancer, concomitant with inhibition of downstream HER2 signaling. Our results suggest that HER2 exon 20 insertion-driven tumors can be effectively treated by a potent and highly selective HER2 inhibitor while sparing WT EGFR, paving the way for clinical translation.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Récepteurs ErbB/génétique , Exons/génétique , Gènes erbB-2 , Humains , Tumeurs du poumon/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Récepteur ErbB-2/génétique
2.
Cell Rep ; 39(2): 110636, 2022 04 12.
Article de Anglais | MEDLINE | ID: mdl-35417719

RÉSUMÉ

Genetic networks are characterized by extensive buffering. During tumor evolution, disruption of functional redundancies can create de novo vulnerabilities that are specific to cancer cells. Here, we systematically search for cancer-relevant paralog interactions using CRISPR screens and publicly available loss-of-function datasets. Our analysis reveals >2,000 candidate dependencies, several of which we validate experimentally, including CSTF2-CSTF2T, DNAJC15-DNAJC19, FAM50A-FAM50B, and RPP25-RPP25L. We provide evidence that RPP25L can physically and functionally compensate for the absence of RPP25 as a member of the RNase P/MRP complexes in tRNA processing. Our analysis also reveals unexpected redundancies between sex chromosome genes. We show that chrX- and chrY-encoded paralogs, such as ZFX-ZFY, DDX3X-DDX3Y, and EIF1AX-EIF1AY, are functionally linked. Tumor cell lines from male patients with loss of chromosome Y become dependent on the chrX-encoded gene. We propose targeting of chrX-encoded paralogs as a general therapeutic strategy for human tumors that have lost the Y chromosome.


Sujet(s)
Tumeurs , Oncogènes , DEAD-box RNA helicases/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Humains , Mâle , Antigènes mineurs d'histocompatibilité/métabolisme , Tumeurs/génétique , Protéines de liaison à l'ARN/génétique , Chromosomes sexuels/métabolisme , Chromosome X , Chromosome Y
3.
Sci Rep ; 9(1): 11661, 2019 08 12.
Article de Anglais | MEDLINE | ID: mdl-31406271

RÉSUMÉ

SMARCA4/BRG1 and SMARCA2/BRM, the two mutually exclusive catalytic subunits of the BAF complex, display a well-established synthetic lethal relationship in SMARCA4-deficient cancers. Using CRISPR-Cas9 screening, we identify SMARCA4 as a novel dependency in SMARCA2-deficient esophageal squamous cell carcinoma (ESCC) models, reciprocal to the known synthetic lethal interaction. Restoration of SMARCA2 expression alleviates the dependency on SMARCA4, while engineered loss of SMARCA2 renders ESCC models vulnerable to concomitant depletion of SMARCA4. Dependency on SMARCA4 is linked to its ATPase activity, but not to bromodomain function. We highlight the relevance of SMARCA4 as a drug target in esophageal cancer using an engineered ESCC cell model harboring a SMARCA4 allele amenable to targeted proteolysis and identify SMARCA4-dependent cell models with low or absent SMARCA2 expression from additional tumor types. These findings expand the concept of SMARCA2/SMARCA4 paralog dependency and suggest that pharmacological inhibition of SMARCA4 represents a novel therapeutic opportunity for SMARCA2-deficient cancers.


Sujet(s)
Helicase/antagonistes et inhibiteurs , Tumeurs de l'oesophage/traitement médicamenteux , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Protéines nucléaires/antagonistes et inhibiteurs , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Systèmes CRISPR-Cas/génétique , Lignée cellulaire tumorale , Survie cellulaire/génétique , Helicase/génétique , Épigenèse génétique , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Édition de gène , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Techniques de knock-down de gènes , Techniques de knock-out de gènes , Humains , Mutation perte de fonction , Thérapie moléculaire ciblée/méthodes , Protéines nucléaires/génétique , /génétique , Petit ARN interférent/métabolisme , Mutations synthétiques létales , Facteurs de transcription/déficit
4.
Elife ; 82019 03 25.
Article de Anglais | MEDLINE | ID: mdl-30910006

RÉSUMÉ

Targeted cancer therapy is based on exploiting selective dependencies of tumor cells. By leveraging recent functional screening data of cancer cell lines we identify Werner syndrome helicase (WRN) as a novel specific vulnerability of microsatellite instability-high (MSI-H) cancer cells. MSI, caused by defective mismatch repair (MMR), occurs frequently in colorectal, endometrial and gastric cancers. We demonstrate that WRN inactivation selectively impairs the viability of MSI-H but not microsatellite stable (MSS) colorectal and endometrial cancer cell lines. In MSI-H cells, WRN loss results in severe genome integrity defects. ATP-binding deficient variants of WRN fail to rescue the viability phenotype of WRN-depleted MSI-H cancer cells. Reconstitution and depletion studies indicate that WRN dependence is not attributable to acute loss of MMR gene function but might arise during sustained MMR-deficiency. Our study suggests that pharmacological inhibition of WRN helicase function represents an opportunity to develop a novel targeted therapy for MSI-H cancers.


Sujet(s)
Instabilité des microsatellites , Tumeurs/thérapie , Werner syndrome helicase/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Survie cellulaire , Réparation de mésappariement de l'ADN , Humains , Modèles théoriques , Werner syndrome helicase/génétique
5.
J Cell Sci ; 131(10)2018 05 16.
Article de Anglais | MEDLINE | ID: mdl-29685892

RÉSUMÉ

Protein ubiquitylation is a dynamic post-translational modification that can be reversed by deubiquitylating enzymes (DUBs). It is unclear how the small number (∼100) of DUBs present in mammalian cells regulate the thousands of different ubiquitylation events. Here, we analysed annotated transcripts of human DUBs and found ∼300 ribosome-associated transcripts annotated as protein coding, which thus increases the total number of DUBs. By using USP35, a poorly studied DUB, as a case study, we provide evidence that alternative isoforms contribute to the functional expansion of DUBs. We show that there are two different USP35 isoforms that localise to different intracellular compartments and have distinct functions. Our results reveal that isoform 1 is an anti-apoptotic factor that inhibits staurosporine- and TNF-related apoptosis-inducing ligand (TRAIL; also known as TNFSF10)-induced apoptosis. In contrast, USP35 isoform 2 is an integral membrane protein of the endoplasmic reticulum (ER) that is also present at lipid droplets. Manipulations of isoform 2 levels cause rapid ER stress, likely through deregulation of lipid homeostasis, and lead to cell death. Our work highlights how alternative isoforms provide functional expansion of DUBs and sets directions for future research.This article has an associated First Person interview with the first author of the paper.


Sujet(s)
Endopeptidases/métabolisme , Isoformes de protéines/métabolisme , Ubiquitin thiolesterase/métabolisme , Apoptose , Endopeptidases/génétique , Réticulum endoplasmique/génétique , Réticulum endoplasmique/métabolisme , Cellules HeLa , Humains , Isoformes de protéines/génétique , Transport des protéines , Ubiquitine/métabolisme , Ubiquitin thiolesterase/génétique , Ubiquitination
6.
Elife ; 62017 07 10.
Article de Anglais | MEDLINE | ID: mdl-28691904

RÉSUMÉ

Recent genome analyses have identified recurrent mutations in the cohesin complex in a wide range of human cancers. Here we demonstrate that the most frequently mutated subunit of the cohesin complex, STAG2, displays a strong synthetic lethal interaction with its paralog STAG1. Mechanistically, STAG1 loss abrogates sister chromatid cohesion in STAG2 mutated but not in wild-type cells leading to mitotic catastrophe, defective cell division and apoptosis. STAG1 inactivation inhibits the proliferation of STAG2 mutated but not wild-type bladder cancer and Ewing sarcoma cell lines. Restoration of STAG2 expression in a mutated bladder cancer model alleviates the dependency on STAG1. Thus, STAG1 and STAG2 support sister chromatid cohesion to redundantly ensure cell survival. STAG1 represents a vulnerability of cancer cells carrying mutations in the major emerging tumor suppressor STAG2 across different cancer contexts. Exploiting synthetic lethal interactions to target recurrent cohesin mutations in cancer, e.g. by inhibiting STAG1, holds the promise for the development of selective therapeutics.


Sujet(s)
Antigènes nucléaires/génétique , Antigènes nucléaires/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Mutations synthétiques létales , Protéines du cycle cellulaire , Division cellulaire , Lignée cellulaire tumorale , Survie cellulaire , Humains
7.
Oncoimmunology ; 5(9): e1186314, 2016.
Article de Anglais | MEDLINE | ID: mdl-27757297

RÉSUMÉ

STAT1 is an important regulator of NK cell maturation and cytotoxicity. Although the consequences of Stat1-deficiency have been described in detail the underlying molecular functions of STAT1 in NK cells are only partially understood. Here, we describe a novel non-canonical role of STAT1 that was unmasked in NK cells expressing a Stat1-Y701F mutant. This mutation prevents JAK-dependent phosphorylation, subsequent nuclear translocation and cytokine-induced transcriptional activity as verified by RNA-seq analysis. As expected Stat1-Y701F mice displayed impaired NK cell maturation comparable to Stat1-/- animals. In contrast Stat1-Y701F NK cells exerted a significantly enhanced cytotoxicity in vitro and in vivo compared to Stat1-/- NK cells in the absence of detectable transcriptional activity. We thus investigated the STAT1 interactome using primary NK cells derived from Stat1ind mice that inducibly express a FLAG-tagged STAT1. Mass spectrometry revealed that STAT1 directly binds proteins involved in cell junction formation and proteins associated to membrane or membrane-bound vesicles. In line, immunofluorescence studies uncovered the recruitment of STAT1 to the target-cell interphase during NK cell killing. This led us to propose a novel function for STAT1 at the immunological synapse in NK cells regulating tumor surveillance and cytotoxicity.

8.
Mol Cancer Ther ; 15(3): 354-65, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26916115

RÉSUMÉ

BI 882370 is a highly potent and selective RAF inhibitor that binds to the DFG-out (inactive) conformation of the BRAF kinase. The compound inhibited proliferation of human BRAF-mutant melanoma cells with 100× higher potency (1-10 nmol/L) than vemurafenib, whereas wild-type cells were not affected at 1,000 nmol/L. BI 882370 administered orally was efficacious in multiple mouse models of BRAF-mutant melanomas and colorectal carcinomas, and at 25 mg/kg twice daily showed superior efficacy compared with vemurafenib, dabrafenib, or trametinib (dosed to provide exposures reached in patients). To model drug resistance, A375 melanoma-bearing mice were initially treated with vemurafenib; all tumors responded with regression, but the majority subsequently resumed growth. Trametinib did not show any efficacy in this progressing population. BI 882370 induced tumor regression; however, resistance developed within 3 weeks. BI 882370 in combination with trametinib resulted in more pronounced regressions, and resistance was not observed during 5 weeks of second-line therapy. Importantly, mice treated with BI 882370 did not show any body weight loss or clinical signs of intolerability, and no pathologic changes were observed in several major organs investigated, including skin. Furthermore, a pilot study in rats (up to 60 mg/kg daily for 2 weeks) indicated lack of toxicity in terms of clinical chemistry, hematology, pathology, and toxicogenomics. Our results indicate the feasibility of developing novel compounds that provide an improved therapeutic window compared with first-generation BRAF inhibitors, resulting in more pronounced and long-lasting pathway suppression and thus improved efficacy.


Sujet(s)
Antinéoplasiques/pharmacologie , Mutation , Tumeurs/génétique , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/génétique , Animaux , Antinéoplasiques/composition chimique , Marqueurs biologiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques , Activation enzymatique/effets des médicaments et des substances chimiques , Femelle , Humains , Isoenzymes , Mâle , Souris , Modèles moléculaires , Conformation moléculaire , Structure moléculaire , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Inhibiteurs de protéines kinases/composition chimique , Multimérisation de protéines , Protéines proto-oncogènes B-raf/composition chimique , Rats , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Bipolar Disord ; 16(7): 764-8, 2014 Nov.
Article de Anglais | MEDLINE | ID: mdl-24754353

RÉSUMÉ

OBJECTIVES: Copy number variants (CNVs) have been shown to affect susceptibility for neuropsychiatric disorders. To date, studies implicating the serotonergic system in complex conditions have just focused on single nucleotide polymorphisms (SNPs). We therefore sought to identify novel common genetic copy number polymorphisms affecting genes of the serotonergic system, and to assess their putative role in bipolar affective disorder (BPAD) and major depressive disorder (MDD). METHODS: A selection of 41 genes of the serotonergic system encoding receptors, the serotonin transporter, metabolic enzymes and chaperones were investigated using a paired-end mapping (PEM) approach on next-generation sequencing data from the pilot project of the 1000 Genomes Project. For association testing, 593 patients with MDD, 1,145 patients with BPAD, and 1,738 healthy controls were included in the study. RESULTS: PEM led to the identification of a microdeletion in the gene encoding tryptophan hydroxylase 2 (TPH2), affecting an amygdala- and hippocampus-specific isoform. It was not associated with BPAD or MDD using a case-control association approach. CONCLUSIONS: We did not find evidence for a role of the TPH2 microdeletion in the pathoetiology of affective disorders. Further studies examining its putative role in behavioral traits regulated by the limbic system are warranted.


Sujet(s)
Amygdale (système limbique)/anatomopathologie , Prédisposition génétique à une maladie/génétique , Hippocampe/anatomopathologie , Troubles de l'humeur/génétique , Troubles de l'humeur/anatomopathologie , Polymorphisme de nucléotide simple/génétique , Tryptophane 5-monooxygenase/génétique , Femelle , Humains , Déséquilibre de liaison , Mâle
10.
Bioinformatics ; 28(18): i333-i339, 2012 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-22962449

RÉSUMÉ

MOTIVATION: The discovery of genomic structural variants (SVs) at high sensitivity and specificity is an essential requirement for characterizing naturally occurring variation and for understanding pathological somatic rearrangements in personal genome sequencing data. Of particular interest are integrated methods that accurately identify simple and complex rearrangements in heterogeneous sequencing datasets at single-nucleotide resolution, as an optimal basis for investigating the formation mechanisms and functional consequences of SVs. RESULTS: We have developed an SV discovery method, called DELLY, that integrates short insert paired-ends, long-range mate-pairs and split-read alignments to accurately delineate genomic rearrangements at single-nucleotide resolution. DELLY is suitable for detecting copy-number variable deletion and tandem duplication events as well as balanced rearrangements such as inversions or reciprocal translocations. DELLY, thus, enables to ascertain the full spectrum of genomic rearrangements, including complex events. On simulated data, DELLY compares favorably to other SV prediction methods across a wide range of sequencing parameters. On real data, DELLY reliably uncovers SVs from the 1000 Genomes Project and cancer genomes, and validation experiments of randomly selected deletion loci show a high specificity. AVAILABILITY: DELLY is available at www.korbel.embl.de/software.html CONTACT: tobias.rausch@embl.de.


Sujet(s)
Variation structurale du génome , Séquençage nucléotidique à haut débit/méthodes , Analyse de séquence d'ADN/méthodes , Logiciel , Cartographie chromosomique/méthodes , Génome humain , Génomique/méthodes , Humains , Délétion de séquence
11.
Genome Res ; 21(12): 2004-13, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21862627

RÉSUMÉ

Copy-number variants (CNVs) form an abundant class of genetic variation with a presumed widespread impact on individual traits. While recent advances, such as the population-scale sequencing of human genomes, facilitated the fine-scale mapping of CNVs, the phenotypic impact of most of these CNVs remains unclear. By relating copy-number genotypes to transcriptome sequencing data, we have evaluated the impact of CNVs, mapped at fine scale, on gene expression. Based on data from 129 individuals with ancestry from two populations, we identified CNVs associated with the expression of 110 genes, with 13% of the associations involving complex, multiallelic CNVs. Categorization of CNVs according to variant type, size, and gene overlap enabled us to examine the impact of different CNV classes on expression variation. While many small (<4 kb) CNVs were associated with expression variation, overall we observed an enrichment of large duplications and deletions, including large intergenic CNVs, relative to the entire set of expression-associated CNVs. Furthermore, the copy number of genes intersecting with CNVs typically correlated positively with the genes' expression, and also was more strongly correlated with expression than nearby single nucleotide polymorphisms, suggesting a frequent causal role of CNVs in expression quantitative trait loci (eQTLs). We also elucidated unexpected cases of negative correlations between copy number and expression by assessing the CNVs' effects on the structure and regulation of genes. Finally, we examined dosage compensation of transcript levels. Our results suggest that association studies can gain in resolution and power by including fine-scale CNV information, such as those obtained from population-scale sequencing.


Sujet(s)
Variations de nombre de copies de segment d'ADN/physiologie , Dosage génique/physiologie , Régulation de l'expression des gènes/physiologie , Génotype , Modèles génétiques , Transcriptome/physiologie , Femelle , Humains , Mâle , Polymorphisme de nucléotide simple
12.
PLoS Comput Biol ; 6(11): e1000988, 2010 Nov 11.
Article de Anglais | MEDLINE | ID: mdl-21085617

RÉSUMÉ

Copy-number variations (CNVs) are widespread in the human genome, but comprehensive assignments of integer locus copy-numbers (i.e., copy-number genotypes) that, for example, enable discrimination of homozygous from heterozygous CNVs, have remained challenging. Here we present CopySeq, a novel computational approach with an underlying statistical framework that analyzes the depth-of-coverage of high-throughput DNA sequencing reads, and can incorporate paired-end and breakpoint junction analysis based CNV-analysis approaches, to infer locus copy-number genotypes. We benchmarked CopySeq by genotyping 500 chromosome 1 CNV regions in 150 personal genomes sequenced at low-coverage. The assessed copy-number genotypes were highly concordant with our performed qPCR experiments (Pearson correlation coefficient 0.94), and with the published results of two microarray platforms (95-99% concordance). We further demonstrated the utility of CopySeq for analyzing gene regions enriched for segmental duplications by comprehensively inferring copy-number genotypes in the CNV-enriched >800 olfactory receptor (OR) human gene and pseudogene loci. CopySeq revealed that OR loci display an extensive range of locus copy-numbers across individuals, with zero to two copies in some OR loci, and two to nine copies in others. Among genetic variants affecting OR loci we identified deleterious variants including CNVs and SNPs affecting ~15% and ~20% of the human OR gene repertoire, respectively, implying that genetic variants with a possible impact on smell perception are widespread. Finally, we found that for several OR loci the reference genome appears to represent a minor-frequency variant, implying a necessary revision of the OR repertoire for future functional studies. CopySeq can ascertain genomic structural variation in specific gene families as well as at a genome-wide scale, where it may enable the quantitative evaluation of CNVs in genome-wide association studies involving high-throughput sequencing.


Sujet(s)
Variations de nombre de copies de segment d'ADN , Génomique/méthodes , Récepteurs olfactifs/génétique , Cartographie chromosomique , Bases de données génétiques , Variation génétique , Génétique des populations , Génome , Génotype , Humains , Séquençage par oligonucléotides en batterie , Réaction de polymérisation en chaîne , Polymorphisme de nucléotide simple , Analyse en composantes principales , Reproductibilité des résultats , Analyse de séquence d'ADN , Logiciel ,
13.
PLoS Genet ; 6(9): e1001109, 2010 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-20838597

RÉSUMÉ

Accurate estimates of mutation rates provide critical information to analyze genome evolution and organism fitness. We used whole-genome DNA sequencing, pulse-field gel electrophoresis, and comparative genome hybridization to determine mutation rates in diploid vegetative and meiotic mutation accumulation lines of Saccharomyces cerevisiae. The vegetative lines underwent only mitotic divisions while the meiotic lines underwent a meiotic cycle every ∼20 vegetative divisions. Similar base substitution rates were estimated for both lines. Given our experimental design, these measures indicated that the meiotic mutation rate is within the range of being equal to zero to being 55-fold higher than the vegetative rate. Mutations detected in vegetative lines were all heterozygous while those in meiotic lines were homozygous. A quantitative analysis of intra-tetrad mating events in the meiotic lines showed that inter-spore mating is primarily responsible for rapidly fixing mutations to homozygosity as well as for removing mutations. We did not observe 1-2 nt insertion/deletion (in-del) mutations in any of the sequenced lines and only one structural variant in a non-telomeric location was found. However, a large number of structural variations in subtelomeric sequences were seen in both vegetative and meiotic lines that did not affect viability. Our results indicate that the diploid yeast nuclear genome is remarkably stable during the vegetative and meiotic cell cycles and support the hypothesis that peripheral regions of chromosomes are more dynamic than gene-rich central sections where structural rearrangements could be deleterious. This work also provides an improved estimate for the mutational load carried by diploid organisms.


Sujet(s)
Diploïdie , Génome fongique/génétique , Méiose/génétique , Saccharomyces cerevisiae/croissance et développement , Saccharomyces cerevisiae/génétique , Substitution d'acide aminé/génétique , Chromosomes de champignon/génétique , Simulation numérique , ADN fongique/génétique , Électrophorèse en champ pulsé , Homozygote , Mutation de type INDEL/génétique , Caryotypage , Mutation/génétique , Polymorphisme génétique , Reproduction/génétique , Saccharomyces cerevisiae/cytologie , Analyse de séquence d'ADN , Spores fongiques/cytologie , Spores fongiques/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE