Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
Radiat Oncol ; 19(1): 85, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956684

RÉSUMÉ

BACKGROUND: Radiotherapy is essential in the treatment of prostate cancer. An alternative to conventional photon radiotherapy is the application of carbon ions, which provide a superior intratumoral dose distribution and less induced damage to adjacent healthy tissue. A common characteristic of prostate cancer cells is their dependence on androgens which is exploited therapeutically by androgen deprivation therapy in the advanced prostate cancer stage. Here, we aimed to analyze the transcriptomic response of prostate cancer cells to irradiation by photons in comparison to carbon ions, focusing on DNA damage, DNA repair and androgen receptor signaling. METHODS: Prostate cancer cell lines LNCaP (functional TP53 and androgen receptor signaling) and DU145 (dysfunctional TP53 and androgen receptor signaling) were irradiated by photons or carbon ions and the subsequent DNA damage was assessed by immuno-cytofluorescence. Furthermore, the cells were treated with an androgen-receptor agonist. The effects of irradiation and androgen treatment on the gene regulation and the transcriptome were investigated by RT-qPCR and RNA sequencing, followed by bioinformatic analysis. RESULTS: Following photon or carbon ion irradiation, both LNCaP and DU145 cells showed a dose-dependent amount of visible DNA damage that decreased over time, indicating occurring DNA repair. In terms of gene regulation, mRNAs involved in the TP53-dependent DNA damage response were significantly upregulated by photons and carbon ions in LNCaP but not in DU145 cells, which generally showed low levels of gene regulation after irradiation. Both LNCaP and DU145 cells responded to photons and carbon ions by downregulation of genes involved in DNA repair and cell cycle, partially resembling the transcriptome response to the applied androgen receptor agonist. Neither photons nor carbon ions significantly affected canonical androgen receptor-dependent gene regulation. Furthermore, certain genes that were specifically regulated by either photon or carbon ion irradiation were identified. CONCLUSION: Photon and carbon ion irradiation showed a significant congruence in terms of induced signaling pathways and transcriptomic responses. These responses were strongly impacted by the TP53 status. Nevertheless, irradiation mode-dependent distinct gene regulations with undefined implication for radiotherapy outcome were revealed. Androgen receptor signaling and irradiations shared regulation of certain genes with respect to DNA-repair and cell-cycle.


Sujet(s)
Photons , Tumeurs de la prostate , Récepteurs aux androgènes , Transduction du signal , Transcriptome , Protéine p53 suppresseur de tumeur , Humains , Mâle , Tumeurs de la prostate/radiothérapie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Transcriptome/effets des radiations , Transduction du signal/effets des radiations , Altération de l'ADN/effets des radiations , Radiothérapie par ions lourds , Réparation de l'ADN , Régulation de l'expression des gènes tumoraux/effets des radiations , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Carbone/pharmacologie
2.
Mol Cancer ; 21(1): 191, 2022 10 03.
Article de Anglais | MEDLINE | ID: mdl-36192757

RÉSUMÉ

BACKGROUND: In vivo gene editing of somatic cells with CRISPR nucleases has facilitated the generation of autochthonous mouse tumors, which are initiated by genetic alterations relevant to the human disease and progress along a natural timeline as in patients. However, the long and variable, orthotopic tumor growth in inner organs requires sophisticated, time-consuming and resource-intensive imaging for longitudinal disease monitoring and impedes the use of autochthonous tumor models for preclinical studies. METHODS: To facilitate a more widespread use, we have generated a reporter mouse that expresses a Cre-inducible luciferase from Gaussia princeps (GLuc), which is secreted by cells in an energy-consuming process and can be measured quantitatively in the blood as a marker for the viable tumor load. In addition, we have developed a flexible, complementary toolkit to rapidly assemble recombinant adenoviruses (AVs) for delivering Cre recombinase together with CRISPR nucleases targeting cancer driver genes. RESULTS: We demonstrate that intratracheal infection of GLuc reporter mice with CRISPR-AVs efficiently induces lung tumors driven by mutations in the targeted cancer genes and simultaneously activates the GLuc transgene, resulting in GLuc secretion into the blood by the growing tumor. GLuc blood levels are easily and robustly quantified in small-volume blood samples with inexpensive equipment, enable tumor detection already several months before the humane study endpoint and precisely mirror the kinetics of tumor development specified by the inducing gene combination. CONCLUSIONS: Our study establishes blood-based GLuc monitoring as an inexpensive, rapid, high-throughput and animal-friendly method to longitudinally monitor autochthonous tumor growth in preclinical studies.


Sujet(s)
Copepoda , Tumeurs du poumon , Animaux , Clustered regularly interspaced short palindromic repeats , Copepoda/génétique , Copepoda/métabolisme , Édition de gène , Gènes rapporteurs , Humains , Luciferases/génétique , Luciferases/métabolisme , Tumeurs du poumon/génétique , Souris
3.
Cell Death Differ ; 29(11): 2163-2176, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-35459909

RÉSUMÉ

The processes leading from disturbed B-cell development to adult B-cell progenitor acute lymphoblastic leukemia (BCP-ALL) remain poorly understood. Here, we describe Irf4-/- mice as prone to developing BCP-ALL with age. Irf4-/- preB-I cells exhibited impaired differentiation but enhanced proliferation in response to IL-7, along with reduced retention in the IL-7 providing bone marrow niche due to decreased CXCL12 responsiveness. Thus selected, preB-I cells acquired Jak3 mutations, probably following irregular AID activity, resulting in malignant transformation. We demonstrate heightened IL-7 sensitivity due to Jak3 mutants, devise a model to explain it, and describe structural and functional similarities to Jak2 mutations often occurring in human Ph-like ALL. Finally, targeting JAK signaling with Ruxolitinib in vivo prolonged survival of mice bearing established Irf4-/- leukemia. Intriguingly, organ infiltration including leukemic meningeosis was selectively reduced without affecting blood blast counts. In this work, we present spontaneous leukemogenesis following IRF4 deficiency with potential implications for high-risk BCP-ALL in adult humans.


Sujet(s)
Lymphome de Burkitt , Leucémie-lymphome lymphoblastique à précurseurs B et T , Adulte , Animaux , Humains , Souris , Lymphocytes B , Lymphome de Burkitt/anatomopathologie , Interleukine-7/génétique , Janus kinase 3/génétique , Mutation/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Transduction du signal
4.
Oncogene ; 41(7): 1011-1023, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34907344

RÉSUMÉ

The tumor suppressive transcription factor p53 is frequently inactivated in cancer cells by missense mutations that cluster in the DNA binding domain. 30% hit mutational hotspot residues, resulting in a complete loss of transcriptional activity and mutant p53-driven chemotherapy resistance. Of the remaining 70% of non-hotspot mutants, many are partial loss-of-function (partial-LOF) mutants with residual transcriptional activity. The therapeutic consequences of a partial-LOF have remained largely elusive. Using a p53 mutation engineered to reduce DNA binding, we demonstrate that partial-LOF is sufficient to enhance oncogene-driven tumorigenesis in mouse models of lung and pancreatic ductal adenocarcinoma and acute myeloid leukemia. Interestingly, mouse and human tumors with partial-LOF mutations showed mutant p53 protein accumulation similar as known for hotspot mutants. Different from the chemotherapy resistance caused by p53-loss, the partial-LOF mutant sensitized to an apoptotic chemotherapy response and led to a survival benefit. Mechanistically, the pro-apoptotic transcriptional activity of mouse and human partial-LOF mutants was rescued at high mutant protein levels, suggesting that accumulation of partial-LOF mutants enables the observed apoptotic chemotherapy response. p53 non-hotspot mutants with partial-LOF, therefore, represent tumorigenic p53 mutations that need to be distinguished from other mutations because of their beneficial impact on survival in a therapy context.


Sujet(s)
Protéine p53 suppresseur de tumeur
5.
Cancers (Basel) ; 13(19)2021 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-34638442

RÉSUMÉ

The impact of aspirin use after the diagnosis of colorectal cancer is unknown. Among others, PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha) mutational status was proposed as a molecular biomarker for the response to adjuvant aspirin therapy. However, prognostic data on aspirin use after a colorectal cancer diagnosis in relation to KRAS mutational status is limited. In a single-center retrospective study, we obtained KRAS and PIK3CA mutational status in a cohort of 153 patients with a first diagnosis of colorectal cancer receiving tumor surgery with curative intent. PIK3CA mutational status was determined by pyrosequencing, and KRAS mutational status was determined by next-generation sequencing. Clinicopathological data and survival data were assessed using patient records and reporting registers. We observed a significant 10-year overall survival benefit in patients with aspirin use and combined wild-type PIK3CA and mutated-KRAS tumors (HR = 0.38; 95% CI = 0.17-0.87; p = 0.02), but not in patients without aspirin use. Our data indicate a benefit of aspirin usage particularly for patients with combined wild-type PIK3CA and mutated-KRAS tumor characteristics.

6.
Cancer Res ; 80(23): 5231-5244, 2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-32873634

RÉSUMÉ

Posttranslational modifications are essential for regulating the transcription factor p53, which binds DNA in a highly cooperative manner to control expression of a plethora of tumor-suppressive programs. Here we show at the biochemical, cellular, and organismal level that the cooperative nature of DNA binding is reduced by phosphorylation of highly conserved serine residues (human S183/S185, mouse S180) in the DNA-binding domain. To explore the role of this inhibitory phosphorylation in vivo, new phosphorylation-deficient p53-S180A knock-in mice were generated. Chromatin immunoprecipitation sequencing and RNA sequencing studies of S180A knock-in cells demonstrated enhanced DNA binding and increased target gene expression. In vivo, this translated into a tissue-specific vulnerability of the bone marrow that caused depletion of hematopoietic stem cells and impaired proper regeneration of hematopoiesis after DNA damage. Median lifespan was significantly reduced by 20% from 709 days in wild type to only 568 days in S180A littermates. Importantly, lifespan was reduced by a loss of general fitness and increased susceptibility to age-related diseases, not by increased cancer incidence as often seen in other p53-mutant mouse models. For example, S180A knock-in mice showed markedly reduced spontaneous tumorigenesis and increased resistance to Myc-driven lymphoma and Eml4-Alk-driven lung cancer. Preventing phosphorylation of S183/S185 in human cells boosted p53 activity and allowed tumor cells to be killed more efficiently. Together, our data identify p53 DNA-binding domain phosphorylation as a druggable mechanism that balances tumorigenesis and aging. SIGNIFICANCE: These findings demonstrate that p53 tumor suppressor activity is reduced by DNA-binding domain phosphorylation to prevent aging and identify this phosphorylation as a potential target for cancer therapy.See related commentary by Horikawa, p. 5164.


Sujet(s)
Tumeurs , Protéine p53 suppresseur de tumeur , Vieillissement/génétique , Animaux , Carcinogenèse/génétique , ADN , Altération de l'ADN , Longévité/génétique , Souris , Tumeurs/génétique , Phosphorylation , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
7.
Nature ; 586(7827): 133-138, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32728212

RÉSUMÉ

Somatic mutations in p53, which inactivate the tumour-suppressor function of p53 and often confer oncogenic gain-of-function properties, are very common in cancer1,2. Here we studied the effects of hotspot gain-of-function mutations in Trp53 (the gene that encodes p53 in mice) in mouse models of WNT-driven intestinal cancer caused by Csnk1a1 deletion3,4 or ApcMin mutation5. Cancer in these models is known to be facilitated by loss of p533,6. We found that mutant versions of p53 had contrasting effects in different segments of the gut: in the distal gut, mutant p53 had the expected oncogenic effect; however, in the proximal gut and in tumour organoids it had a pronounced tumour-suppressive effect. In the tumour-suppressive mode, mutant p53 eliminated dysplasia and tumorigenesis in Csnk1a1-deficient and ApcMin/+ mice, and promoted normal growth and differentiation of tumour organoids derived from these mice. In these settings, mutant p53 was more effective than wild-type p53 at inhibiting tumour formation. Mechanistically, the tumour-suppressive effects of mutant p53 were driven by disruption of the WNT pathway, through preventing the binding of TCF4 to chromatin. Notably, this tumour-suppressive effect was completely abolished by the gut microbiome. Moreover, a single metabolite derived from the gut microbiota-gallic acid-could reproduce the entire effect of the microbiome. Supplementing gut-sterilized p53-mutant mice and p53-mutant organoids with gallic acid reinstated the TCF4-chromatin interaction and the hyperactivation of WNT, thus conferring a malignant phenotype to the organoids and throughout the gut. Our study demonstrates the substantial plasticity of a cancer mutation and highlights the role of the microenvironment in determining its functional outcome.


Sujet(s)
Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Microbiome gastro-intestinal/génétique , Gènes suppresseurs de tumeur , Mutation , Oncogènes/génétique , Protéine p53 suppresseur de tumeur/génétique , Animaux , Antibactériens/pharmacologie , Carcinogenèse/effets des médicaments et des substances chimiques , Femelle , Acide gallique/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Humains , Mâle , Souris , Organoïdes/métabolisme , Transcription génétique , Protéine p53 suppresseur de tumeur/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques
8.
Cancers (Basel) ; 12(5)2020 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-32357518

RÉSUMÉ

Survivin is a drug target and its suppressant YM155 a drug candidate mainly investigated for high-risk neuroblastoma. Findings from one YM155-adapted subline of the neuroblastoma cell line UKF-NB-3 had suggested that increased ABCB1 (mediates YM155 efflux) levels, decreased SLC35F2 (mediates YM155 uptake) levels, decreased survivin levels, and TP53 mutations indicate YM155 resistance. Here, the investigation of 10 additional YM155-adapted UKF-NB-3 sublines only confirmed the roles of ABCB1 and SLC35F2. However, cellular ABCB1 and SLC35F2 levels did not indicate YM155 sensitivity in YM155-naïve cells, as indicated by drug response data derived from the Cancer Therapeutics Response Portal (CTRP) and the Genomics of Drug Sensitivity in Cancer (GDSC) databases. Moreover, the resistant sublines were characterized by a remarkable heterogeneity. Only seven sublines developed on-target resistance as indicated by resistance to RNAi-mediated survivin depletion. The sublines also varied in their response to other anti-cancer drugs. In conclusion, cancer cell populations of limited intrinsic heterogeneity can develop various resistance phenotypes in response to treatment. Therefore, individualized therapies will require monitoring of cancer cell evolution in response to treatment. Moreover, biomarkers can indicate resistance formation in the acquired resistance setting, even when they are not predictive in the intrinsic resistance setting.

9.
J Biol Chem ; 295(33): 11388-11407, 2020 08 14.
Article de Anglais | MEDLINE | ID: mdl-32303635

RÉSUMÉ

Cleavage of influenza virus hemagglutinin (HA) by host proteases is essential for virus infectivity. HA of most influenza A and B (IAV/IBV) viruses is cleaved at a monobasic motif by trypsin-like proteases. Previous studies have reported that transmembrane serine protease 2 (TMPRSS2) is essential for activation of H7N9 and H1N1pdm IAV in mice but that H3N2 IAV and IBV activation is independent of TMPRSS2 and carried out by as-yet-undetermined protease(s). Here, to identify additional H3 IAV- and IBV-activating proteases, we used RNA-Seq to investigate the protease repertoire of murine lower airway tissues, primary type II alveolar epithelial cells (AECIIs), and the mouse lung cell line MLE-15. Among 13 candidates identified, TMPRSS4, TMPRSS13, hepsin, and prostasin activated H3 and IBV HA in vitro IBV activation and replication was reduced in AECIIs from Tmprss2/Tmprss4-deficient mice compared with WT or Tmprss2-deficient mice, indicating that murine TMPRSS4 is involved in IBV activation. Multicycle replication of H3N2 IAV and IBV in AECIIs of Tmprss2/Tmprss4-deficient mice varied in sensitivity to protease inhibitors, indicating that different, but overlapping, sets of murine proteases facilitate H3 and IBV HA cleavages. Interestingly, human hepsin and prostasin orthologs did not activate H3, but they did activate IBV HA in vitro Our results indicate that TMPRSS4 is an IBV-activating protease in murine AECIIs and suggest that TMPRSS13, hepsin, and prostasin cleave H3 and IBV HA in mice. They further show that hepsin and prostasin orthologs might contribute to the differences observed in TMPRSS2-independent activation of H3 in murine and human airways.


Sujet(s)
Sous-type H3N2 du virus de la grippe A/physiologie , Virus influenza B/physiologie , Grippe humaine/enzymologie , Infections à Orthomyxoviridae/enzymologie , Peptide hydrolases/métabolisme , Activation virale , Animaux , Lignée cellulaire , Chiens , Activation enzymatique/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes , Cellules HEK293 , Interactions hôte-pathogène/effets des médicaments et des substances chimiques , Humains , Sous-type H3N2 du virus de la grippe A/effets des médicaments et des substances chimiques , Virus influenza B/effets des médicaments et des substances chimiques , Grippe humaine/traitement médicamenteux , Grippe humaine/génétique , Grippe humaine/virologie , Poumon/enzymologie , Poumon/métabolisme , Poumon/virologie , Cellules rénales canines Madin-Darby , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris , Infections à Orthomyxoviridae/traitement médicamenteux , Infections à Orthomyxoviridae/génétique , Infections à Orthomyxoviridae/virologie , Peptide hydrolases/génétique , Inhibiteurs de protéases/pharmacologie , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , Activation virale/effets des médicaments et des substances chimiques
10.
Proc Natl Acad Sci U S A ; 116(44): 22288-22293, 2019 10 29.
Article de Anglais | MEDLINE | ID: mdl-31611375

RÉSUMÉ

Cancer development is driven by activated oncogenes and loss of tumor suppressors. While oncogene inhibitors have entered routine clinical practice, tumor suppressor reactivation therapy remains to be established. For the most frequently inactivated tumor suppressor p53, genetic mouse models have demonstrated regression of p53-null tumors upon p53 reactivation. While this was shown in tumor models driven by p53 loss as the initiating lesion, many human tumors initially develop in the presence of wild-type p53, acquire aberrations in the p53 pathway to bypass p53-mediated tumor suppression, and inactivate p53 itself only at later stages during metastatic progression or therapy. To explore the efficacy of p53 reactivation in this scenario, we used a reversibly switchable p53 (p53ERTAM) mouse allele to generate Eµ-Myc-driven lymphomas in the presence of active p53 and, after full lymphoma establishment, switched off p53 to model late-stage p53 inactivation. Although these lymphomas had evolved in the presence of active p53, later loss and subsequent p53 reactivation surprisingly activated p53 target genes triggering massive apoptosis, tumor regression, and long-term cure of the majority of animals. Mechanistically, the reactivation response was dependent on Cdkn2a/p19Arf, which is commonly silenced in p53 wild-type lymphomas, but became reexpressed upon late-stage p53 inactivation. Likewise, human p53 wild-type tumor cells with CRISPR-engineered switchable p53ERTAM alleles responded to p53 reactivation when CDKN2A/p14ARF function was restored or mimicked with Mdm2 inhibitors. Together, these experiments provide genetic proof of concept that tumors can respond, in an ARF-dependent manner, to p53 reactivation even if p53 inactivation has occurred late during tumor evolution.


Sujet(s)
Carcinogenèse/génétique , Thérapie génétique/méthodes , Lymphomes/génétique , Protéine p53 suppresseur de tumeur/génétique , Cellules 3T3 , Animaux , Apoptose , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Cellules HCT116 , Humains , Lymphomes/thérapie , Souris , Protéine p53 suppresseur de tumeur/métabolisme
11.
EMBO J ; 38(20): e102096, 2019 10 15.
Article de Anglais | MEDLINE | ID: mdl-31483066

RÉSUMÉ

Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53-/- mice. Surprisingly, stabilization of p53R178E in Mdm2-/- mice nevertheless triggers extensive apoptosis, indicative of residual wild-type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E ;Mdm2-/- embryos, it proves insufficient for suppression of spontaneous and oncogene-driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53-/- mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53-/- ones, resulting in enhanced eradication of p53-mutated tumor cells. Together, this provides genetic proof-of-principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Leucémie aigüe myéloïde/prévention et contrôle , Lymphomes/prévention et contrôle , Mutation , Protéines proto-oncogènes c-mdm2/physiologie , Protéine p53 suppresseur de tumeur/physiologie , Animaux , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Cycle cellulaire , Modèles animaux de maladie humaine , Évolution de la maladie , Femelle , Régulation de l'expression des gènes tumoraux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Lymphomes/génétique , Lymphomes/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Cellules cancéreuses en culture
12.
Oncotarget ; 10(20): 1975-1992, 2019 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-30956778

RÉSUMÉ

BACKGROUND: Genetic factors play a substantial role in breast cancer etiology. Genes encoding proteins that have key functions in the DNA damage response, such as p53 and its inhibitors MDM2 and MDMX, are most likely candidates to harbor allelic variants that influence breast cancer susceptibility. The aim of our study was to comprehensively analyze the impact of SNPs in the TP53, MDM2, and MDMX genes in conjunction with TP53 mutational status regarding the onset and progression of breast cancer. METHODS: In specimen from 815 breast cancer patients, five SNPs within the selected genes were analyzed: TP53 - Arg72Pro (rs1042522), MDM2 - SNP285 (rs2279744), SNP309 (rs117039649); MDMX - SNP31826 (rs1563828), and SNP34091 (rs4245739). Classification of the tumors was evaluated by histomorphology. Subtyping according hormone receptor status, HER2-status and proliferation rate enabled provision of the clinico-pathological surrogate of intrinsic subtypes. RESULTS: The homozygous C-allele of MDM2 SNP285 was significantly associated with a younger age-at-diagnosis of 44.2 years, in contrast to G/G- and G/C-patients (62.4, 62.7 yrs., respectively; p = 0.0007; log-Rank-test). In contrast, there was no difference regarding the age-at-diagnosis for patients with the respective genotypes of MDM2 SNP309 (p = 0.799; log-Rank-test). In patients with estrogen receptor (ER)-positive and TP53-mutated tumors, however, the T/T-genotype of the MDM2 SNP309 was significantly associated with an earlier average age-at-diagnosis compared with T/G+G/G-patients (53.5 vs. 68.2 yrs; p = 0.002; log-Rank-test). In the triple-negative subgroup, the G/G-patients had an average age-at-diagnosis of 51 years compared with 63 years for SNP309T carriers (p = 0.004; log-Rank-test) indicating a susceptibility of the G/G genotype for the development of triple negative breast cancer. Patients with the A/A-genotype of MDMX SNP31826 with ER-negative tumors were diagnosed 11 years earlier compared with patients and ER-positive tumors (53.2 vs. 64.4 yrs; p = 0.025, log-Rank-test). Furthermore, in luminal B-like patients (HER2-independent) the C/C-genotype of MDMX SNP34091 was significantly correlated with a decreased event-free survival compared with the A/A-genotype (p < 0.001; log-Rank-test). CONCLUSIONS: We showed that SNPs in the MDM2 and MDMX genes affect at least in part the onset and progression of breast cancer dependent on the ER-status. Our findings provide further evidence for the distinct etiological pathways in ER-negative and ER-positive breast cancers.

13.
Oncotarget ; 9(32): 22423-22435, 2018 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-29854289

RÉSUMÉ

Acute myeloid leukemia (AML) arises through clonal expansion of transformed myeloid progenitor cells. The SKI proto-oncogene is highly upregulated in different solid tumors and leukemic cells, but little is known about its transcriptional regulation during leukemogenesis. MYB is an important hematopoietic transcription factor involved in proliferation as well as differentiation and upregulated in most human acute leukemias. Here, we find that MYB protein binds within the regulatory region of the SKI gene in AML cells. Reporter gene assays using MYB binding sites present in the SKI gene locus show MYB-dependent transcriptional activation. SiRNA-mediated depletion of MYB in leukemic cell lines reveals that MYB is crucial for SKI gene expression. Consistently, we observed a positive correlation of MYB and SKI expression in leukemic cell lines and in samples of AML patients. Moreover, MYB and SKI both were downregulated by treatment with histone deacetylase inhibitors. Strikingly, differentiation of AML cells induced by depletion of MYB is attenuated by overexpression of SKI. Our findings identify SKI as a novel MYB target gene, relevant for the MYB-induced differentiation block in leukemic cells.

14.
Nucleic Acids Res ; 46(14): 7097-7107, 2018 08 21.
Article de Anglais | MEDLINE | ID: mdl-29860503

RÉSUMÉ

The two paralogous zinc finger factors CTCF and CTCFL differ in expression such that CTCF is ubiquitously expressed, whereas CTCFL is found during spermatogenesis and in some cancer types in addition to other cell types. Both factors share the highly conserved DNA binding domain and are bound to DNA sequences with an identical consensus. In contrast, both factors differ substantially in the number of bound sites in the genome. Here, we addressed the molecular features for this binding specificity. In contrast to CTCF we found CTCFL highly enriched at 'open' chromatin marked by H3K27 acetylation, H3K4 di- and trimethylation, H3K79 dimethylation and H3K9 acetylation plus the histone variant H2A.Z. CTCFL is enriched at transcriptional start sites and regions bound by transcription factors. Consequently, genes deregulated by CTCFL are highly cell specific. In addition to a chromatin-driven choice of binding sites, we determined nucleotide positions critical for DNA binding by CTCFL, but not by CTCF.


Sujet(s)
Facteur de liaison à la séquence CCCTC/métabolisme , Chromatine/métabolisme , Protéines de liaison à l'ADN/métabolisme , Animaux , Séquence nucléotidique , Sites de fixation , Lignée cellulaire , ADN/composition chimique , Humains , Cellules K562 , Souris , Cellules NIH 3T3
15.
J Neurosci ; 38(4): 858-877, 2018 01 24.
Article de Anglais | MEDLINE | ID: mdl-29217679

RÉSUMÉ

Schwann cell differentiation and myelination depends on chromatin remodeling, histone acetylation, and methylation, which all affect Schwann cell proliferation. We previously reported that the deletion of the POZ (POxvirus and Zinc finger) domain of the transcription factor Miz1 (Myc-interacting zinc finger protein; encoded by Zbtb17) in mouse Schwann cells (Miz1ΔPOZ) causes a neuropathy at 90 d after birth [postnatal day (P) 90], with a subsequent spontaneous regeneration. Here we show that RNA sequencing from Miz1ΔPOZ and control animals at P30 revealed a set of upregulated genes with a strong correlation to cell-cycle regulation. Consistently, a subset of Schwann cells did not exit the cell cycle as observed in control animals and the growth fraction increased over time. From the RNAseq gene list, two direct Miz1 target genes were identified, one of which encodes the histone H3K36me2 demethylase Kdm8. We show that the expression of Kdm8 is repressed by Miz1 and that its release in Miz1ΔPOZ cells induces a decrease of H3K36me2, especially in deregulated cell-cycle-related genes. The linkage between elevated Kdm8 expression, hypomethylation of H3K36 at cell-cycle-relevant genes, and the subsequent re-entering of adult Schwann cells into the cell cycle suggests that the release of Kdm8 repression in the absence of a functional Miz1 is a central issue in the development of the Miz1ΔPOZ phenotype.SIGNIFICANCE STATEMENT The deletion of the Miz1 (Myc-interacting zinc finger protein 1) POZ (POxvirus and Zinc finger) domain in Schwann cells causes a neuropathy. Here we report sustained Schwann cell proliferation caused by an increased expression of the direct Miz1 target gene Kdm8, encoding a H3K36me2 demethylase. Hence, the demethylation of H3K36 is linked to the pathogenesis of a neuropathy.


Sujet(s)
Maladies démyélinisantes/métabolisme , Jumonji Domain-Containing Histone Demethylases/métabolisme , Protéines nucléaires/métabolisme , Neuropathies périphériques/métabolisme , Inhibiteurs de STAT activés/métabolisme , Cellules de Schwann/métabolisme , Animaux , Prolifération cellulaire/physiologie , Méthylation de l'ADN/physiologie , Régulation de l'expression des gènes/physiologie , Histone/métabolisme , Souris , Souris knockout , Ubiquitin-protein ligases
16.
Proc Natl Acad Sci U S A ; 114(38): E8035-E8044, 2017 09 19.
Article de Anglais | MEDLINE | ID: mdl-28878021

RÉSUMÉ

Casein kinase 1α (CK1α), a component of the ß-catenin destruction complex, is a critical regulator of Wnt signaling; its ablation induces both Wnt and p53 activation. To characterize the role of CK1α (encoded by Csnk1a1) in skin physiology, we crossed mice harboring floxed Csnk1a1 with mice expressing K14-Cre-ERT2 to generate mice in which tamoxifen induces the deletion of Csnk1a1 exclusively in keratinocytes [single-knockout (SKO) mice]. As expected, CK1α loss was accompanied by ß-catenin and p53 stabilization, with the preferential induction of p53 target genes, but phenotypically most striking was hyperpigmentation of the skin, importantly without tumorigenesis, for at least 9 mo after Csnk1a1 ablation. The number of epidermal melanocytes and eumelanin levels were dramatically increased in SKO mice. To clarify the putative role of p53 in epidermal hyperpigmentation, we established K14-Cre-ERT2 CK1α/p53 double-knockout (DKO) mice and found that coablation failed to induce epidermal hyperpigmentation, demonstrating that it was p53-dependent. Transcriptome analysis of the epidermis revealed p53-dependent up-regulation of Kit ligand (KitL). SKO mice treated with ACK2 (a Kit-neutralizing antibody) or imatinib (a Kit inhibitor) abrogated the CK1α ablation-induced hyperpigmentation, demonstrating that it requires the KitL/Kit pathway. Pro-opiomelanocortin (POMC), a precursor of α-melanocyte-stimulating hormone (α-MSH), was not activated in the CK1α ablation-induced hyperpigmentation, which is in contrast to the mechanism of p53-dependent UV tanning. Nevertheless, acute sunburn effects were successfully prevented in the hyperpigmented skin of SKO mice. CK1α inhibition induces skin-protective eumelanin but no carcinogenic pheomelanin and may therefore constitute an effective strategy for safely increasing eumelanin via UV-independent pathways, protecting against acute sunburn.


Sujet(s)
Casein kinase I/métabolisme , Kératinocytes/métabolisme , Pigmentation de la peau , Coup de soleil/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Casein kinase I/antagonistes et inhibiteurs , Casein kinase I/génétique , Épiderme/métabolisme , Épiderme/anatomopathologie , Kératinocytes/anatomopathologie , Mélanines/biosynthèse , Mélanines/génétique , Mélanocytes/métabolisme , Mélanocytes/anatomopathologie , Souris , Souris knockout , Coup de soleil/génétique , Coup de soleil/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , bêta-Caténine/génétique , bêta-Caténine/métabolisme
17.
Proc Natl Acad Sci U S A ; 113(52): E8433-E8442, 2016 12 27.
Article de Anglais | MEDLINE | ID: mdl-27956623

RÉSUMÉ

Mutations in the p53 tumor suppressor gene are the most frequent genetic alteration in cancer and are often associated with progression from benign to invasive stages with metastatic potential. Mutations inactivate tumor suppression by p53, and some endow the protein with novel gain of function (GOF) properties that actively promote tumor progression and metastasis. By comparative gene expression profiling of p53-mutated and p53-depleted cancer cells, we identified ectonucleoside triphosphate diphosphohydrolase 5 (ENTPD5) as a mutant p53 target gene, which functions as a uridine 5'-diphosphatase (UDPase) in the endoplasmic reticulum (ER) to promote the folding of N-glycosylated membrane proteins. A comprehensive pan-cancer analysis revealed a highly significant correlation between p53 GOF mutations and ENTPD5 expression. Mechanistically, mutp53 is recruited by Sp1 to the ENTPD5 core promoter to induce its expression. We show ENTPD5 to be a mediator of mutant p53 GOF activity in clonogenic growth, architectural tissue remodeling, migration, invasion, and lung colonization in an experimental metastasis mouse model. Our study reveals folding of N-glycosylated membrane proteins in the ER as a mechanism underlying the metastatic progression of tumors with mutp53 that could provide new possibilities for cancer treatment.


Sujet(s)
Réticulum endoplasmique/métabolisme , Métastase tumorale , Protéines oncogènes/métabolisme , Pyrophosphatases/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/physiologie , Animaux , Apoptose , Calnexine/métabolisme , Calréticuline/métabolisme , Carcinogenèse/métabolisme , Lignée cellulaire tumorale , Évolution de la maladie , Femelle , Glycoprotéines/métabolisme , Glycosylation , Humains , Mâle , Souris , Protéines mutantes/génétique , Protéines mutantes/physiologie , Mutation , Invasion tumorale , Pronostic , Interférence par ARN , Petit ARN interférent/métabolisme , Facteur de transcription Sp1/métabolisme
18.
Cell Death Dis ; 7(10): e2410, 2016 10 13.
Article de Anglais | MEDLINE | ID: mdl-27735941

RÉSUMÉ

Resistance formation after initial therapy response (acquired resistance) is common in high-risk neuroblastoma patients. YM155 is a drug candidate that was introduced as a survivin suppressant. This mechanism was later challenged, and DNA damage induction and Mcl-1 depletion were suggested instead. Here we investigated the efficacy and mechanism of action of YM155 in neuroblastoma cells with acquired drug resistance. The efficacy of YM155 was determined in neuroblastoma cell lines and their sublines with acquired resistance to clinically relevant drugs. Survivin levels, Mcl-1 levels, and DNA damage formation were determined in response to YM155. RNAi-mediated depletion of survivin, Mcl-1, and p53 was performed to investigate their roles during YM155 treatment. Clinical YM155 concentrations affected the viability of drug-resistant neuroblastoma cells through survivin depletion and p53 activation. MDM2 inhibitor-induced p53 activation further enhanced YM155 activity. Loss of p53 function generally affected anti-neuroblastoma approaches targeting survivin. Upregulation of ABCB1 (causes YM155 efflux) and downregulation of SLC35F2 (causes YM155 uptake) mediated YM155-specific resistance. YM155-adapted cells displayed increased ABCB1 levels, decreased SLC35F2 levels, and a p53 mutation. YM155-adapted neuroblastoma cells were also characterized by decreased sensitivity to RNAi-mediated survivin depletion, further confirming survivin as a critical YM155 target in neuroblastoma. In conclusion, YM155 targets survivin in neuroblastoma. Furthermore, survivin is a promising therapeutic target for p53 wild-type neuroblastomas after resistance acquisition (neuroblastomas are rarely p53-mutated), potentially in combination with p53 activators. In addition, we show that the adaptation of cancer cells to molecular-targeted anticancer drugs is an effective strategy to elucidate a drug's mechanism of action.


Sujet(s)
Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Imidazoles/pharmacologie , Protéines IAP/métabolisme , Naphtoquinones/pharmacologie , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Altération de l'ADN , Humains , Cinétique , Protéines de transport membranaire/métabolisme , Mutation/génétique , Pipérazines/pharmacologie , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-mdm2/métabolisme , Petit ARN interférent/métabolisme , Survivine , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
19.
Cell Tissue Res ; 366(3): 651-665, 2016 Dec.
Article de Anglais | MEDLINE | ID: mdl-27553638

RÉSUMÉ

Autophagy is a lysosomal degradation process involved in the turnover of organelles or other cell constituents, in providing sources for energy production under starving conditions and in cell metabolism. A key protein in the macroautophagic machinery is the autophagy-related protein (Atg) 7. Constitutive deletion of Atg7 is lethal at birth. A conditional deletion of Atg7 in hepatocytes leads to hepatomegaly and in aged animals to liver tumors. With this study, we aim at analyzing the hepatomegaly development in more detail. The 3- to 4-fold enlargement of the liver takes place between days 25 and 35 after birth (P25-P35) and persists at least until P90. This is accompanied by a change in the expression of enzymes involved in the glycogen/glucose metabolism. While glycogen synthesis is inhibited, glucose is preferentially kept as glucose-6-phosphate inside the cells, inducing a swelling of the cells caused by hyperosmolarity. An increase of lipogenic enzymes suggests that glucose-6-phosphate is delivered to lipogenic pathways, which is supported by the occurrence of a steatosis around P30. The development of hepatomegaly is accompanied by a polyploidisation of hepatocytes, an enhanced expression of genes related to inflammatory processes and an infiltration of macrophages and granulocytes. Our data provide evidence that the attenuation of macroautophagy in hepatocytes leads to a glucose retention that causes cell swelling. The resulting hepatomegaly, which develops in a time interval of about 10 days, perturbs liver perfusion and induces an inflammatory reaction together with polyploidisation.


Sujet(s)
Protéine-7 associée à l'autophagie/métabolisme , Délétion de gène , Glycogène/métabolisme , Hépatomégalie/métabolisme , Animaux , Mort cellulaire , Prolifération cellulaire , Hydrates de carbone alimentaires , Femelle , Régulation de l'expression des gènes , Glucose/métabolisme , Hépatomégalie/anatomopathologie , Inflammation/anatomopathologie , Foie/métabolisme , Mâle , Souris de lignée C57BL , Souris knockout , Spécificité d'organe , Polyploïdie
20.
BMC Cancer ; 16: 502, 2016 07 19.
Article de Anglais | MEDLINE | ID: mdl-27435226

RÉSUMÉ

BACKGROUND: The multifunctional ß-galactoside-binding protein galectin-3 is found in many distinct subcellular compartments including the cell nucleus. Expression and distribution of galectin-3 between the cell nucleus and the cytosol changes during cell differentiation and cancer development. Nuclear functions of galectin-3 and how they contribute to tumorigenesis are not understood. METHODS: In order to identify nuclear galectin-3 interaction partners, we used affinity chromatography and co-immunoprecipitation. Spatial proximity in the nucleus was assessed by immunofluorescence and proximity ligation assay. We also investigated the function of galectin-3 on mRNA-export by fluorescence in situ hybridization and on mRNA-processing by RNA-sequencing. RESULTS: The heterogeneous ribonucleoprotein particle component hnRNPA2B1 was identified as a novel galectin-3 binding protein that associates with the lectin in a lactose-dependent manner in the cell nucleus. Specific individual depletion of galectin-3 does not affect the mRNA distribution between cytoplasm and nucleus. A significant alteration of this distribution was observed after combined depletion of galectin-1 and -3. However, silencing of galectin-3 was sufficient to alter the splicing patterns of several genes. CONCLUSIONS: Galectin-3 and hnRNPA2B1 interact as members of the early splicing machinery. Galectin-3 and -1 have redundant functions in mRNA transport and at least in part in mRNA splicing. RNA-sequencing data points to a specific function of the hnRNPA2B1/galectin-3 interaction in the processing of transcripts coding for the nuclear oncoprotein SET.


Sujet(s)
Noyau de la cellule/génétique , Galectine -3/génétique , Ribonucléoprotéine nucléaire hétérogène du groupe A-B/génétique , ARN messager/génétique , Noyau de la cellule/métabolisme , Galectine -3/métabolisme , Cellules HeLa , Ribonucléoprotéine nucléaire hétérogène du groupe A-B/métabolisme , Humains , Immunotransfert , Immunoprécipitation , Hybridation fluorescente in situ , Microscopie de fluorescence , Liaison aux protéines , Interférence par ARN , Épissage des ARN , Transport des ARN , ARN messager/métabolisme , Analyse de séquence d'ARN/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...