Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Nat Commun ; 15(1): 1165, 2024 Feb 07.
Article de Anglais | MEDLINE | ID: mdl-38326311

RÉSUMÉ

The t(X,17) chromosomal translocation, generating the ASPSCR1::TFE3 fusion oncoprotein, is the singular genetic driver of alveolar soft part sarcoma (ASPS) and some Xp11-rearranged renal cell carcinomas (RCCs), frustrating efforts to identify therapeutic targets for these rare cancers. Here, proteomic analysis identifies VCP/p97, an AAA+ ATPase with known segregase function, as strongly enriched in co-immunoprecipitated nuclear complexes with ASPSCR1::TFE3. We demonstrate that VCP is a likely obligate co-factor of ASPSCR1::TFE3, one of the only such fusion oncoprotein co-factors identified in cancer biology. Specifically, VCP co-distributes with ASPSCR1::TFE3 across chromatin in association with enhancers genome-wide. VCP presence, its hexameric assembly, and its enzymatic function orchestrate the oncogenic transcriptional signature of ASPSCR1::TFE3, by facilitating assembly of higher-order chromatin conformation structures demonstrated by HiChIP. Finally, ASPSCR1::TFE3 and VCP demonstrate co-dependence for cancer cell proliferation and tumorigenesis in vitro and in ASPS and RCC mouse models, underscoring VCP's potential as a novel therapeutic target.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Animaux , Souris , Humains , Protéomique , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Translocation génétique , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Tumeurs du rein/génétique , Chromatine/génétique , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Chromosomes X humains/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéine contenant la valosine/génétique
2.
bioRxiv ; 2023 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-37873234

RÉSUMÉ

The t(X,17) chromosomal translocation, generating the ASPSCR1-TFE3 fusion oncoprotein, is the singular genetic driver of alveolar soft part sarcoma (ASPS) and some Xp11-rearranged renal cell carcinomas (RCC), frustrating efforts to identify therapeutic targets for these rare cancers. Proteomic analysis showed that VCP/p97, an AAA+ ATPase with known segregase function, was strongly enriched in co-immunoprecipitated nuclear complexes with ASPSCR1-TFE3. We demonstrate that VCP is a likely obligate co-factor of ASPSCR1-TFE3, one of the only such fusion oncoprotein co-factors identified in cancer biology. Specifically, VCP co-distributed with ASPSCR1-TFE3 across chromatin in association with enhancers genome-wide. VCP presence, its hexameric assembly, and its enzymatic function orchestrated the oncogenic transcriptional signature of ASPSCR1-TFE3, by facilitating assembly of higher-order chromatin conformation structures as demonstrated by HiChIP. Finally, ASPSCR1-TFE3 and VCP demonstrated co-dependence for cancer cell proliferation and tumorigenesis in vitro and in ASPS and RCC mouse models, underscoring VCP's potential as a novel therapeutic target.

3.
J Pathol ; 258(4): 325-338, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36031730

RÉSUMÉ

Clear cell ovarian carcinoma (CCOC) is the second most common subtype of epithelial ovarian carcinoma. Late-stage CCOC is not responsive to gold-standard chemotherapy and results in suboptimal outcomes for patients. In-depth molecular insight is urgently needed to stratify the disease and drive therapeutic development. We conducted global proteomics for 192 cases of CCOC and compared these with other epithelial ovarian carcinoma subtypes. Our results showed distinct proteomic differences in CCOC compared with other epithelial ovarian cancer subtypes including alterations in lipid and purine metabolism pathways. Furthermore, we report potential clinically significant proteomic subgroups within CCOC, suggesting the biologic plausibility of stratified treatment for this cancer. Taken together, our results provide a comprehensive understanding of the CCOC proteomic landscape to facilitate future understanding and research of this disease. © 2022 The Pathological Society of Great Britain and Ireland.


Sujet(s)
Adénocarcinome à cellules claires , Tumeurs de l'ovaire , Femelle , Humains , Carcinome épithélial de l'ovaire/anatomopathologie , Protéome , Protéomique , Adénocarcinome à cellules claires/anatomopathologie , Tumeurs de l'ovaire/métabolisme
4.
Nat Commun ; 13(1): 896, 2022 02 16.
Article de Anglais | MEDLINE | ID: mdl-35173148

RÉSUMÉ

Despite advances in genomic classification of breast cancer, current clinical tests and treatment decisions are commonly based on protein level information. Formalin-fixed paraffin-embedded (FFPE) tissue specimens with extended clinical outcomes are widely available. Here, we perform comprehensive proteomic profiling of 300 FFPE breast cancer surgical specimens, 75 of each PAM50 subtype, from patients diagnosed in 2008-2013 (n = 178) and 1986-1992 (n = 122) with linked clinical outcomes. These two cohorts are analyzed separately, and we quantify 4214 proteins across all 300 samples. Within the aggressive PAM50-classified basal-like cases, proteomic profiling reveals two groups with one having characteristic immune hot expression features and highly favorable survival. Her2-Enriched cases separate into heterogeneous groups differing by extracellular matrix, lipid metabolism, and immune-response features. Within 88 triple-negative breast cancers, four proteomic clusters display features of basal-immune hot, basal-immune cold, mesenchymal, and luminal with disparate survival outcomes. Our proteomic analysis characterizes the heterogeneity of breast cancer in a clinically-applicable manner, identifies potential biomarkers and therapeutic targets, and provides a resource for clinical breast cancer classification.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Protéome/métabolisme , Tumeurs du sein triple-négatives/classification , Tumeurs du sein triple-négatives/anatomopathologie , Région mammaire/anatomopathologie , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/génétique , Humains , Protéomique , Résultat thérapeutique , Tumeurs du sein triple-négatives/mortalité
5.
Autophagy ; 18(11): 2547-2560, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35220892

RÉSUMÉ

Chloroquine (CQ), a lysosomotropic agent, is commonly used to inhibit lysosomal degradation and macroautophagy/autophagy. Here we investigated the cell-extrinsic effects of CQ on secretion. We showed that lysosomal and autophagy inhibition by CQ altered the secretome, and induced the release of Atg8 orthologs and autophagy receptors. Atg8-family proteins, in particular, were secreted inside small extracellular vesicles (sEVs) in a lipidation-dependent manner. CQ treatment enhanced the release of Atg8-family proteins inside sEVs. Using full-length ATG16L1 and an ATG16L1 mutant that enables Atg8-family protein lipidation on double but not on single membranes, we demonstrated that LC3B is released in two distinct sEV populations: one enriched with SDCBP/Syntenin-1, CD63, and endosomal lipidated LC3B, and another that contains LC3B but is not enriched with SDCBP/Syntenin-1 or CD63, and which our data supports as originating from a double-membrane source. Our findings underscore the context-dependency of sEV heterogeneity and composition, and illustrate the integration of autophagy and sEV composition in response to lysosomal inhibition.Abbreviations: ACTB: actin beta; ANOVA: analysis of variance; ATG4B: autophagy related 4B cysteine peptidase; Atg8: autophagy related 8; ATG16L1: autophagy related 16 like 1; ATP5F1A/ATP5a: ATP synthase F1 subunit alpha; CALCOCO2: calcium binding and coiled-coil domain 2; CASP3: caspase 3; CASP7: caspase 7; CQ: chloroquine; CD9: CD9 molecule; CD63: CD63 molecule; DAPI: 4',6-diamidino-2-phenylindole; DQ-BSA: dye quenched-bovine serum albumin; ER: endoplasmic reticulum; ERN1/IRE1a: endoplasmic reticulum to nucleus signaling 1; EV: extracellular vesicles; FBS: fetal bovine serum; FDR: false discovery rate; GABARAP: GABA type A receptor-associated protein; GABARAPL2: GABA type A receptor associated protein like 2; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GO: gene ontology; HCQ: hydroxychloroquine; HSP90AA1: heat shock protein 90 alpha family class A member 1; IP: immunoprecipitation; KO: knockout; LAMP2: lysosomal associated membrane protein 2; LIR: LC3-interacting region; LMNA: lamin A/C; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MS: mass spectrometry; NBR1: NBR1 autophagy cargo receptor; NCOA4: nuclear receptor coactivator 4; NTA: nanoparticle tracking analysis; PE: phosphatidylethanolamine; PECA: probe-level expression change averaging; SDCBP/syntenin-1: syndecan binding protein; SD: standard deviation; SE: secreted; sEV: small extracellular vesicles; SQSTM1/p62: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TEM: transmission electron microscopy; TMT: tandem-mass tag; TSG101: tumor susceptibility 101; ULK1: unc-51 like autophagy activating kinase 1; WC: whole cell.


Sujet(s)
Vésicules extracellulaires , Synténines , Famille de la protéine-8 associée à l'autophagie/métabolisme , Protéines associées à l'autophagie/métabolisme , Synténines/métabolisme , Chloroquine/pharmacologie , Autophagie/physiologie , Protéines régulatrices de l'apoptose/métabolisme , Vésicules extracellulaires/métabolisme , Acide gamma-amino-butyrique
6.
Cell Death Differ ; 29(1): 54-64, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34294889

RÉSUMÉ

Breast cancer heterogeneity has made it challenging to identify mechanisms critical to the initial stages of their genesis in vivo. Here, we sought to interrogate the role of YB-1 in newly arising human breast cancers as well as in established cell lines. In a first series of experiments, we found that short-hairpin RNA-mediated knockdown of YB-1 in MDA-MB-231 cells blocked both their local tumour-forming and lung-colonising activity in immunodeficient mice. Conversely, upregulated expression of YB-1 enhanced the poor in vivo tumorigenicity of T47D cells. We then found that YB-1 knockdown also inhibits the initial generation in mice of invasive ductal carcinomas and ductal carcinomas in situ from freshly isolated human mammary cells transduced, respectively, with KRASG12D or myristoylated-AKT1. Interestingly, increased expression of HIF1α and G3BP1, two YB-1 translational targets and elements of a stress-adaptive programme, mirrored the levels of YB-1 in both transformed primary and established MDA-MB-231 breast cancer cells.


Sujet(s)
Tumeurs du sein , Animaux , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Helicase/métabolisme , Femelle , Humains , Souris , Protéines liant le poly-adp-ribose , RNA helicases/métabolisme , Protéines à motif de reconnaissance de l'ARN , Facteurs de transcription/métabolisme , Protéine-1 de liaison à la boîte Y/génétique , Protéine-1 de liaison à la boîte Y/métabolisme
7.
Cell Rep ; 37(2): 109817, 2021 10 12.
Article de Anglais | MEDLINE | ID: mdl-34644566

RÉSUMÉ

Pancreatic neuroendocrine neoplasms (PNENs) are biologically and clinically heterogeneous. Here, we use a multi-omics approach to uncover the molecular factors underlying this heterogeneity. Transcriptomic analysis of 84 PNEN specimens, drawn from two cohorts, is substantiated with proteomic profiling and identifies four subgroups: Proliferative, PDX1-high, Alpha cell-like and Stromal/Mesenchymal. The Proliferative subgroup, consisting of both well- and poorly differentiated specimens, is associated with inferior overall survival probability. The PDX1-high and Alpha cell-like subgroups partially resemble previously described subtypes, and we further uncover distinctive metabolism-related features in the Alpha cell-like subgroup. The Stromal/Mesenchymal subgroup exhibits molecular characteristics of YAP1/WWTR1(TAZ) activation suggestive of Hippo signaling pathway involvement in PNENs. Whole-exome sequencing reveals subgroup-enriched mutational differences, supported by activity inference analysis, and identifies hypermorphic proto-oncogene variants in 14.3% of sequenced PNENs. Our study reveals differences in cellular signaling axes that provide potential directions for PNEN patient stratification and treatment strategies.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protéome , Transcriptome , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Différenciation cellulaire , Prolifération cellulaire , Protéines corépressives/génétique , Protéines corépressives/métabolisme , Bases de données génétiques , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Hétérogénéité génétique , Humains , Mâle , Chaperons moléculaires/génétique , Chaperons moléculaires/métabolisme , Mutation , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/thérapie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/thérapie , Pronostic , Protéomique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Transduction du signal , Transcriptional coactivator with PDZ-binding motif proteins/génétique , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Protéines de signalisation YAP/génétique , Protéines de signalisation YAP/métabolisme
8.
Cancer Discov ; 11(11): 2884-2903, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34021002

RÉSUMÉ

Cancer cells must overcome anoikis (detachment-induced death) to successfully metastasize. Using proteomic screens, we found that distinct oncoproteins upregulate IL1 receptor accessory protein (IL1RAP) to suppress anoikis. IL1RAP is directly induced by oncogenic fusions of Ewing sarcoma, a highly metastatic childhood sarcoma. IL1RAP inactivation triggers anoikis and impedes metastatic dissemination of Ewing sarcoma cells. Mechanistically, IL1RAP binds the cell-surface system Xc - transporter to enhance exogenous cystine uptake, thereby replenishing cysteine and the glutathione antioxidant. Under cystine depletion, IL1RAP induces cystathionine gamma lyase (CTH) to activate the transsulfuration pathway for de novo cysteine synthesis. Therefore, IL1RAP maintains cyst(e)ine and glutathione pools, which are vital for redox homeostasis and anoikis resistance. IL1RAP is minimally expressed in pediatric and adult normal tissues, and human anti-IL1RAP antibodies induce potent antibody-dependent cellular cytotoxicity of Ewing sarcoma cells. Therefore, we define IL1RAP as a new cell-surface target in Ewing sarcoma, which is potentially exploitable for immunotherapy. SIGNIFICANCE: Here, we identify cell-surface protein IL1RAP as a key driver of metastasis in Ewing sarcoma, a highly aggressive childhood sarcoma. Minimal expression in pediatric and adult normal tissues nominates IL1RAP as a promising target for immunotherapy.See related commentary by Yoon and DeNicola, p. 2679.This article is highlighted in the In This Issue feature, p. 2659.


Sujet(s)
Anoïkis , Protéine accessoire du récepteur à l'interleukine-1 , Sarcome d'Ewing , Adulte , Lignée cellulaire tumorale , Enfant , Humains , Protéomique , Récepteurs à l'interleukine-1 , Sarcome d'Ewing/génétique , Sarcome d'Ewing/métabolisme , Sarcome d'Ewing/anatomopathologie
9.
J Proteomics ; 239: 104182, 2021 05 15.
Article de Anglais | MEDLINE | ID: mdl-33705978

RÉSUMÉ

Protein aggregation is indicative of failing protein quality control systems. These systems are responsible for the refolding or degradation of aberrant and misfolded proteins. Heat stress can cause proteins to misfold, triggering cellular responses including a marked increase in the ubiquitination of proteins. This response has been characterized in yeast, however more studies are needed within mammalian cells. Herein, we examine proteins that become ubiquitinated during heat shock in human tissue culture cells using diGly enrichment coupled with mass spectrometry. A majority of these proteins are localized in the nucleus or cytosol. Proteins which are conjugated under stress display longer sequence lengths, more interaction partners, and more hydrophobic patches than controls but do not show lower melting temperatures. Furthermore, heat-induced conjugation sites occur less frequently in disordered regions and are closer to hydrophobic patches than other ubiquitination sites; perhaps providing novel insight into the molecular mechanism mediating this response. Nuclear and cytosolic pools of modified proteins appear to have different protein features. Using a pulse-SILAC approach, we found that both long-lived and newly-synthesized proteins are conjugated under stress. Modified long-lived proteins are predominately nuclear and were distinct from newly-synthesized proteins, indicating that different pathways may mediate the heat-induced increase of polyubiquitination. SIGNIFICANCE: The maintenance of protein homeostasis requires a balance of protein synthesis, folding, and degradation. Under stress conditions, the cell must rapidly adapt by increasing its folding capacity to eliminate aberrant proteins. A major pathway for proteolysis is mediated by the ubiquitin proteasome system. While increased ubiquitination after heat stress was observed over 30 years ago, it remains unclear which proteins are conjugated during heat shock in mammalian cells and by what means this conjugation occurs. In this study, we combined SILAC-based mass spectrometry with computational analyses to reveal features associated to proteins ubiquitinated while under heat shock. Interestingly, we found that conjugation sites induced by the stress are less often located within disordered regions and more often located near hydrophobic patches. Our study showcases how proteomics can reveal distinct feature associated to a cohort of proteins that are modified post translationally and how the ubiquitin conjugation sites are preferably selected in these conditions. Our work opens a new path for delineating the molecular mechanisms leading to the heat stress response and the regulation of protein homeostasis.


Sujet(s)
Réaction de choc thermique , Ubiquitine , Animaux , Humains , Proteasome endopeptidase complex/métabolisme , Pliage des protéines , Saccharomyces cerevisiae/métabolisme , Ubiquitine/métabolisme , Ubiquitination
10.
Cancer Res ; 81(7): 1681-1694, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33441310

RÉSUMÉ

Low-grade serous ovarian carcinoma (LGSOC) is a rare tumor subtype with high case fatality rates in patients with metastatic disease. There is a pressing need to develop effective treatments using newly available preclinical models for therapeutic discovery and drug evaluation. Here, we use multiomics integration of whole-exome sequencing, RNA sequencing, and mass spectrometry-based proteomics on 14 LGSOC cell lines to elucidate novel biomarkers and therapeutic vulnerabilities. Comparison of LGSOC cell line data with LGSOC tumor data enabled predictive biomarker identification of MEK inhibitor (MEKi) efficacy, with KRAS mutations found exclusively in MEKi-sensitive cell lines and NRAS mutations found mostly in MEKi-resistant cell lines. Distinct patterns of Catalogue of Somatic Mutations in Cancer mutational signatures were identified in MEKi-sensitive and MEKi-resistant cell lines. Deletions of CDKN2A/B and MTAP genes were more frequent in cell lines than tumor samples and possibly represent key driver events in the absence of KRAS/NRAS/BRAF mutations. These LGSOC cell lines were representative models of the molecular aberrations found in LGSOC tumors. For prediction of in vitro MEKi efficacy, proteomic data provided better discrimination than gene expression data. Condensin, minichromosome maintenance, and replication factor C protein complexes were identified as potential treatment targets in MEKi-resistant cell lines. This study suggests that CDKN2A/B or MTAP deficiency may be exploited using synthetically lethal treatment strategies, highlighting the importance of using proteomic data as a tool for molecular drug prediction. Multiomics approaches are crucial to improving our understanding of the molecular underpinnings of LGSOC and applying this information to develop new therapies. SIGNIFICANCE: These findings highlight the utility of global multiomics to characterize LGSOC cell lines as research models, to determine biomarkers of MEKi resistance, and to identify potential novel therapeutic targets.


Sujet(s)
Biomarqueurs pharmacologiques/analyse , Cystadénocarcinome séreux/traitement médicamenteux , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Tumeurs de l'ovaire/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/isolement et purification , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire tumorale , Études de cohortes , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/métabolisme , Cystadénocarcinome séreux/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Génomique/méthodes , Humains , Métabolomique/méthodes , Grading des tumeurs , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Protéomique/méthodes , Intégration de systèmes
11.
Hum Pathol ; 108: 1-11, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33121982

RÉSUMÉ

Mesonephric carcinomas (MEs) and female adnexal tumors of probable Wolffian origin (FATWO) are derived from embryologic remnants of Wolffian/mesonephric ducts. Mesonephric-like carcinomas (MLCs) show identical morphology to ME of the cervix but occur in the uterus and ovary without convincing mesonephric remnants. ME, MLC, and FATWO are challenging to diagnose due to their morphologic similarities to Müllerian/paramesonephric tumors, contributing to a lack of evidence-based and tumor-specific treatments. We performed whole-proteomic analysis on 9 ME/MLC and 56 endometrial carcinomas (ECs) to identify potential diagnostic biomarkers. Although there were no convincing differences between ME and MLC, 543 proteins showed increased expression in ME/MLC relative to EC. From these proteins, euchromatic histone lysine methyltransferase 2 (EHMT2), glutathione S-transferase Mu 3 (GSTM3), eukaryotic translation elongation factor 1 alpha 2 (EEF1A2), and glycogen synthase kinase 3 beta were identified as putative biomarkers. Immunohistochemistry was performed on these candidates and GATA3 in 14 ME/MLC, 8 FATWO, 155 EC, and normal tissues. Of the candidates, only GATA3 and EHMT2 were highly expressed in mesonephric remnants and mesonephric-derived male tissues. GATA3 had the highest sensitivity and specificity for ME/MLC versus EC (93% and 99%) but was absent in FATWO. EHMT2 was 100% sensitive for ME/MLC & FATWO but was not specific (65%). Similarly, EEF1A2 was reasonably sensitive to ME/MLC (92%) and FATWO (88%) but was the least specific (38%). GSTM3 performed intermediately (sensitivity for ME/MLC and FATWO: 83% and 38%, respectively; specificity 67%). Although GATA3 remained the best diagnostic biomarker for ME/MLC, we have identified EHMT2, EEF1A2, and GSTM3 as proteins of interest in these cancers. FATWO's cell of origin is uncertain and remains an area for future research.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Glutathione transferase/analyse , Antigènes d'histocompatibilité/analyse , Histone-lysine N-methyltransferase/analyse , Mésonéphrome/diagnostic , Facteur-1 d'élongation de la chaîne peptidique/analyse , Femelle , Humains , Protéomique/méthodes
12.
Elife ; 92020 12 23.
Article de Anglais | MEDLINE | ID: mdl-33355532

RÉSUMÉ

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare and aggressive form of ovarian cancer. SCCOHT tumors have inactivating mutations in SMARCA4 (BRG1), one of the two mutually exclusive ATPases of the SWI/SNF chromatin remodeling complex. To address the role that BRG1 loss plays in SCCOHT tumorigenesis, we performed integrative multi-omic analyses in SCCOHT cell lines +/- BRG1 reexpression. BRG1 reexpression induced a gene and protein signature similar to an epithelial cell and gained chromatin accessibility sites correlated with other epithelial originating TCGA tumors. Gained chromatin accessibility and BRG1 recruited sites were strongly enriched for transcription-factor-binding motifs of AP-1 family members. Furthermore, AP-1 motifs were enriched at the promoters of highly upregulated epithelial genes. Using a dominant-negative AP-1 cell line, we found that both AP-1 DNA-binding activity and BRG1 reexpression are necessary for the gene and protein expression of epithelial genes. Our study demonstrates that BRG1 reexpression drives an epithelial-like gene and protein signature in SCCOHT cells that depends upon by AP-1 activity.


Sujet(s)
Carcinome à petites cellules/anatomopathologie , Helicase/génétique , Hypercalcémie/anatomopathologie , Protéines nucléaires/génétique , Tumeurs de l'ovaire/métabolisme , Facteur de transcription AP-1/métabolisme , Facteurs de transcription/génétique , Marqueurs biologiques tumoraux/analyse , Carcinome à petites cellules/génétique , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/génétique , Helicase/métabolisme , Femelle , Humains , Hypercalcémie/génétique , Mutation/génétique , Protéines nucléaires/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Ovaire/métabolisme , Ovaire/anatomopathologie , Facteur de transcription AP-1/génétique , Facteurs de transcription/métabolisme
13.
Hum Pathol ; 101: 40-52, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32360491

RÉSUMÉ

The current World Health Organization classification does not distinguish transitional cell carcinoma of the ovary (TCC) from conventional tubo-ovarian high-grade serous carcinoma (HGSC), despite evidence suggesting improved prognosis for patients with TCC; instead, it is considered a morphologic variant of HGSC. The immunohistochemical (IHC) markers applied to date do not distinguish between TCC and HGSC. Therefore, we sought to compare the proteomic profiles of TCC and conventional HGSC to identify proteins enriched in TCC. Prognostic biomarkers in HGSC have proven to be elusive, and our aim was to identify biomarkers of TCC as a way of reliably and reproducibly identifying patients with a favorable prognosis and better response to chemotherapy compared with those with conventional HGSC. Quantitative global proteome analysis was performed on archival material of 12 cases of TCC and 16 cases of HGSC using SP3 (single-pot, solid phase-enhanced, sample preparation)-Clinical Tissue Proteomics, a recently described protocol for full-proteome analysis from formalin-fixed paraffin-embedded tissues. We identified 430 proteins that were significantly enriched in TCC over HGSC. Unsupervised co-clustering perfectly distinguished TCC from HGSC based on protein expression. Pathway analysis showed that proteins associated with cell death, necrosis, and apoptosis were highly expressed in TCCs, whereas proteins associated with DNA homologous recombination, cell mitosis, proliferation and survival, and cell cycle progression pathways had reduced expression. From the proteomic analysis, three potential biomarkers for TCC were identified, claudin-4 (CLDN4), ubiquitin carboxyl-terminal esterase L1 (UCHL1), and minichromosome maintenance protein 7 (MCM7), and tested by IHC analysis on tissue microarrays. In agreement with the proteomic analysis, IHC expression of those proteins was stronger in TCC than in HGSC (p < 0.0001). Using global proteomic analysis, we are able to distinguish TCC from conventional HGSC. Follow-up studies will be necessary to confirm that these molecular and morphologic differences are clinically significant.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Carcinome transitionnel/diagnostic , Cystadénocarcinome séreux/diagnostic , Tumeurs de la trompe de Fallope/diagnostic , Tumeurs de l'ovaire/diagnostic , Femelle , Humains , Protéomique/méthodes
14.
Clin Cancer Res ; 26(16): 4402-4413, 2020 08 15.
Article de Anglais | MEDLINE | ID: mdl-32409304

RÉSUMÉ

PURPOSE: Many rare ovarian cancer subtypes, such as small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT), have poor prognosis due to their aggressive nature and resistance to standard platinum- and taxane-based chemotherapy. The development of effective therapeutics has been hindered by the rarity of such tumors. We sought to identify targetable vulnerabilities in rare ovarian cancer subtypes. EXPERIMENTAL DESIGN: We compared the global proteomic landscape of six cases each of endometrioid ovarian cancer (ENOC), clear cell ovarian cancer (CCOC), and SCCOHT to the most common subtype, high-grade serous ovarian cancer (HGSC), to identify potential therapeutic targets. IHC of tissue microarrays was used as validation of arginosuccinate synthase (ASS1) deficiency. The efficacy of arginine-depriving therapeutic ADI-PEG20 was assessed in vitro using cell lines and patient-derived xenograft mouse models representing SCCOHT. RESULTS: Global proteomic analysis identified low ASS1 expression in ENOC, CCOC, and SCCOHT compared with HGSC. Low ASS1 levels were validated through IHC in large patient cohorts. The lowest levels of ASS1 were observed in SCCOHT, where ASS1 was absent in 12 of 31 cases, and expressed in less than 5% of the tumor cells in 9 of 31 cases. ASS1-deficient ovarian cancer cells were sensitive to ADI-PEG20 treatment regardless of subtype in vitro. Furthermore, in two cell line mouse xenograft models and one patient-derived mouse xenograft model of SCCOHT, once-a-week treatment with ADI-PEG20 (30 mg/kg and 15 mg/kg) inhibited tumor growth in vivo. CONCLUSIONS: Preclinical in vitro and in vivo studies identified ADI-PEG20 as a potential therapy for patients with rare ovarian cancers, including SCCOHT.


Sujet(s)
Argininosuccinate synthase/génétique , Carcinome à petites cellules/traitement médicamenteux , Hydrolases/pharmacologie , Tumeurs de l'ovaire/traitement médicamenteux , Polyéthylène glycols/pharmacologie , Animaux , Arginine/antagonistes et inhibiteurs , Arginine/génétique , Argininosuccinate synthase/déficit , Carcinome à petites cellules/génétique , Carcinome à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Souris , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Ovaire/métabolisme , Ovaire/anatomopathologie , Protéine apparentée à l'hormone parathyroïdienne/génétique , Protéine apparentée à l'hormone parathyroïdienne/immunologie , Protéomique , Tests d'activité antitumorale sur modèle de xénogreffe
15.
EMBO Rep ; 20(12): e48375, 2019 12 05.
Article de Anglais | MEDLINE | ID: mdl-31668005

RÉSUMÉ

Outcomes for metastatic Ewing sarcoma and osteosarcoma are dismal and have not changed for decades. Oxidative stress attenuates melanoma metastasis, and melanoma cells must reduce oxidative stress to metastasize. We explored this in sarcomas by screening for oxidative stress sensitizers, which identified the class I HDAC inhibitor MS-275 as enhancing vulnerability to reactive oxygen species (ROS) in sarcoma cells. Mechanistically, MS-275 inhibits YB-1 deacetylation, decreasing its binding to 5'-UTRs of NFE2L2 encoding the antioxidant factor NRF2, thereby reducing NFE2L2 translation and synthesis of NRF2 to increase cellular ROS. By global acetylomics, MS-275 promotes rapid acetylation of the YB-1 RNA-binding protein at lysine-81, blocking binding and translational activation of NFE2L2, as well as known YB-1 mRNA targets, HIF1A, and the stress granule nucleator, G3BP1. MS-275 dramatically reduces sarcoma metastasis in vivo, but an MS-275-resistant YB-1K81-to-alanine mutant restores metastatic capacity and NRF2, HIF1α, and G3BP1 synthesis in MS-275-treated mice. These studies describe a novel function for MS-275 through enhanced YB-1 acetylation, thus inhibiting YB-1 translational control of key cytoprotective factors and its pro-metastatic activity.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Benzamides/usage thérapeutique , Tumeurs osseuses/traitement médicamenteux , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Pyridines/usage thérapeutique , Sarcome d'Ewing/traitement médicamenteux , Facteurs de transcription/métabolisme , Acétylation , Animaux , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Lignée cellulaire tumorale , Cellules cultivées , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Souris , Facteur-2 apparenté à NF-E2/métabolisme , Métastase tumorale , Stress oxydatif , Sarcome d'Ewing/métabolisme , Sarcome d'Ewing/anatomopathologie
16.
Neoplasia ; 21(8): 740-751, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31220736

RÉSUMÉ

Myxoid liposarcoma is a malignant lipogenic tumor that develops in deep soft tissues. While local control rates are good, current chemotherapy options remain ineffective against metastatic disease. Myxoid liposarcoma is characterized by the FUS-DDIT3 fusion oncoprotein that is proposed to function as an aberrant transcription factor, but its exact mechanism of action has remained unclear. To identify the key functional interacting partners of FUS-DDIT3, this study utilized immunoprecipitation-mass spectrometry (IP-MS) to identify the FUS-DDIT3 interactome in whole cell lysates of myxoid liposarcoma cells, and results showed an enrichment of RNA processing proteins. Further quantitative MS analyses of FUS-DDIT3 complexes isolated from nuclear lysates showed that members of several chromatin regulatory complexes were present in the FUS-DDIT3 interactome, including NuRD, SWI/SNF, PRC1, PRC2, and MLL1 COMPASS-like complexes. Co-immunoprecipitation validated the associations of FUS-DDIT3 with BRG1/SMARCA4, BAF155/SMARCC1, BAF57/SMARCE1, and KDM1A. Data from this study provides candidates for functional validation as potential therapeutic targets, particularly for emerging epigenetic drugs.


Sujet(s)
Protéines de transport/métabolisme , Liposarcome myxoïde/métabolisme , Protéines de fusion oncogènes/métabolisme , Protéines de transport/composition chimique , Lignée cellulaire tumorale , Humains , Liposarcome myxoïde/génétique , Protéines de fusion oncogènes/composition chimique , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Cartographie d'interactions entre protéines , Protéome , Protéomique/méthodes , Reproductibilité des résultats
17.
J Proteome Res ; 18(2): 700-708, 2019 02 01.
Article de Anglais | MEDLINE | ID: mdl-30462513

RÉSUMÉ

Optimizing the quality of proteomics data collected from a mass spectrometer (MS) requires careful selection of acquisition parameters and proper assessment of instrument performance. Software tools capable of extracting a broad set of information from raw files, including meta, scan, quantification, and identification data, are needed to provide guidance for MS system management. In this work, direct extraction and utilization of these data is demonstrated using RawTools, a standalone tool for extracting meta and scan data directly from raw MS files generated on Thermo Orbitrap instruments. RawTools generates summarized and detailed plain text outputs after parsing individual raw files, including scan rates and durations, duty cycle characteristics, precursor and reporter ion quantification, and chromatography performance. RawTools also contains a diagnostic module that includes an optional "preview" database search for facilitating informed decision-making related to optimization of MS performance based on a variety of metrics. RawTools has been developed in C# and utilizes the Thermo RawFileReader library and thus can process raw MS files with high speed and high efficiency on all major operating systems (Windows, MacOS, Linux). To demonstrate the utility of RawTools, the extraction of meta and scan data from both individual and large collections of raw MS files was carried out to identify problematic characteristics of instrument performance. Taken together, the combined rich feature-set of RawTools with the capability for interrogation of MS and experiment performance makes this software a valuable tool for proteomics researchers.


Sujet(s)
Mémorisation et recherche des informations/méthodes , Protéomique/méthodes , Logiciel , Analyse de données , Systèmes de gestion de bases de données , Spectrométrie de masse/méthodes
18.
Mol Cancer Ther ; 17(12): 2767-2779, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30232145

RÉSUMÉ

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare but extremely lethal malignancy that mainly impacts young women. SCCOHT is characterized by a diploid genome with loss of SMARCA4 and lack of SMARCA2 expression, two mutually exclusive ATPases of the SWI/SNF chromatin-remodeling complex. We and others have identified the histone methyltransferase EZH2 as a promising therapeutic target for SCCOHT, suggesting that SCCOHT cells depend on the alternation of epigenetic pathways for survival. In this study, we found that SCCOHT cells were more sensitive to pan-HDAC inhibitors compared with other ovarian cancer lines or immortalized cell lines tested. Pan-HDAC inhibitors, such as quisinostat, reversed the expression of a group of proteins that were deregulated in SCCOHT cells due to SMARCA4 loss, leading to growth arrest, apoptosis, and differentiation in vitro and suppressed tumor growth of xenografted tumors of SCCOHT cells. Moreover, combined treatment of HDAC inhibitors and EZH2 inhibitors at sublethal doses synergistically induced histone H3K27 acetylation and target gene expression, leading to rapid induction of apoptosis and growth suppression of SCCOHT cells and xenografted tumors. Therefore, our preclinical study highlighted the therapeutic potential of combined treatment of HDAC inhibitors with EZH2 catalytic inhibitors to treat SCCOHT.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Carcinome à petites cellules/traitement médicamenteux , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Hypercalcémie/traitement médicamenteux , Tumeurs de l'ovaire/traitement médicamenteux , Animaux , Apoptose/effets des médicaments et des substances chimiques , Biocatalyse/effets des médicaments et des substances chimiques , Carcinome à petites cellules/complications , Carcinome à petites cellules/génétique , Carcinome à petites cellules/anatomopathologie , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Lignage cellulaire/effets des médicaments et des substances chimiques , Helicase/métabolisme , Synergie des médicaments , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Épigenèse génétique/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Acides hydroxamiques/pharmacologie , Acides hydroxamiques/usage thérapeutique , Hypercalcémie/complications , Souris , Protéines nucléaires/métabolisme , Tumeurs de l'ovaire/complications , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Protéome/métabolisme , Facteurs de transcription/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
19.
J Pathol ; 242(3): 371-383, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28444909

RÉSUMÉ

Small cell carcinoma of the ovary, hypercalcaemic type (SCCOHT) is a rare but aggressive and untreatable malignancy affecting young women. We and others recently discovered that SMARCA4, a gene encoding the ATPase of the SWI/SNF chromatin-remodelling complex, is the only gene recurrently mutated in the majority of SCCOHT. The low somatic complexity of SCCOHT genomes and the prominent role of the SWI/SNF chromatin-remodelling complex in transcriptional control of genes suggest that SCCOHT cells may rely on epigenetic rewiring for oncogenic transformation. Herein, we report that approximately 80% (19/24) of SCCOHT tumour samples have strong expression of the histone methyltransferase EZH2 by immunohistochemistry, with the rest expressing variable amounts of EZH2. Re-expression of SMARCA4 suppressed the expression of EZH2 in SCCOHT cells. In comparison to other ovarian cell lines, SCCOHT cells displayed hypersensitivity to EZH2 shRNAs and two selective EZH2 inhibitors, GSK126 and EPZ-6438. EZH2 inhibitors induced cell cycle arrest, apoptosis, and cell differentiation in SCCOHT cells, along with the induction of genes involved in cell cycle regulation, apoptosis, and neuron-like differentiation. EZH2 inhibitors suppressed tumour growth and improved the survival of mice bearing SCCOHT xenografts. Therefore, our data suggest that loss of SMARCA4 creates a dependency on the catalytic activity of EZH2 in SCCOHT cells and that pharmacological inhibition of EZH2 is a promising therapeutic strategy for treating this disease. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Sujet(s)
Carcinome à petites cellules/enzymologie , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Histone-lysine N-methyltransferase/métabolisme , Hypercalcémie/enzymologie , Tumeurs de l'ovaire/enzymologie , Animaux , Apoptose/physiologie , Carcinome épithélial de l'ovaire , Points de contrôle du cycle cellulaire/physiologie , Lignée cellulaire tumorale , Transformation cellulaire néoplasique , Helicase/déficit , Régulation négative , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Femelle , Histone méthyltransférases , Humains , Transplantation tumorale , Tumeurs épithéliales épidermoïdes et glandulaires/enzymologie , Protéines nucléaires/déficit , Facteurs de transcription/déficit , Transplantation hétérologue , Cellules cancéreuses en culture , Régulation positive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE