Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
1.
Nat Immunol ; 25(7): 1193-1206, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38834865

RÉSUMÉ

Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKß-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.


Sujet(s)
Mouvement cellulaire , Cellules dendritiques , Homéostasie , Noeuds lymphatiques , Souris de lignée C57BL , Récepteurs CCR7 , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/cytologie , Récepteurs CCR7/métabolisme , Souris , Mouvement cellulaire/immunologie , Forme de la cellule , Facteur de transcription NF-kappa B/métabolisme , Souris knockout , Transduction du signal/immunologie , I-kappa B Kinase/métabolisme , Complexe Arp-2-3/métabolisme
2.
Front Oncol ; 12: 958155, 2022.
Article de Anglais | MEDLINE | ID: mdl-36387192

RÉSUMÉ

Human TRIAP1 (TP53-regulated inhibitor of apoptosis 1; also known as p53CSV for p53-inducible cell survival factor) is the homolog of yeast Mdm35, a well-known chaperone that interacts with the Ups/PRELI family proteins and participates in the intramitochondrial transfer of lipids for the synthesis of cardiolipin (CL) and phosphatidylethanolamine. Although recent reports indicate that TRIAP1 is a prosurvival factor abnormally overexpressed in various types of cancer, knowledge about its molecular and metabolic function in human cells is still elusive. It is therefore critical to understand the metabolic and proliferative advantages that TRIAP1 expression provides to cancer cells. Here, in a colorectal cancer cell model, we report that the expression of TRIAP1 supports cancer cell proliferation and tumorigenesis. Depletion of TRIAP1 perturbed the mitochondrial ultrastructure, without a major impact on CL levels and mitochondrial activity. TRIAP1 depletion caused extramitochondrial perturbations resulting in changes in the endoplasmic reticulum-dependent lipid homeostasis and induction of a p53-mediated stress response. Furthermore, we observed that TRIAP1 depletion conferred a robust p53-mediated resistance to the metabolic stress caused by glutamine deprivation. These findings highlight the importance of TRIAP1 in tumorigenesis and indicate that the loss of TRIAP1 has extramitochondrial consequences that could impact on the metabolic plasticity of cancer cells and their response to conditions of nutrient deprivation.

3.
Nat Commun ; 12(1): 6564, 2021 11 12.
Article de Anglais | MEDLINE | ID: mdl-34772931

RÉSUMÉ

Hybrids between diverged lineages contain novel genetic combinations but an impaired meiosis often makes them evolutionary dead ends. Here, we explore to what extent an aborted meiosis followed by a return-to-growth (RTG) promotes recombination across a panel of 20 Saccharomyces cerevisiae and S. paradoxus diploid hybrids with different genomic structures and levels of sterility. Genome analyses of 275 clones reveal that RTG promotes recombination and generates extensive regions of loss-of-heterozygosity in sterile hybrids with either a defective meiosis or a heavily rearranged karyotype, whereas RTG recombination is reduced by high sequence divergence between parental subgenomes. The RTG recombination preferentially arises in regions with low local heterozygosity and near meiotic recombination hotspots. The loss-of-heterozygosity has a profound impact on sexual and asexual fitness, and enables genetic mapping of phenotypic differences in sterile lineages where linkage analysis would fail. We propose that RTG gives sterile yeast hybrids access to a natural route for genome recombination and adaptation.


Sujet(s)
Diploïdie , Hybridation génétique , Infertilité/génétique , Méiose , Saccharomyces cerevisiae/génétique , Cartographie chromosomique , Évolution moléculaire , Génome fongique , Recombinaison homologue , Phénotype , Protéines de Saccharomyces cerevisiae/métabolisme
4.
NPJ Breast Cancer ; 7(1): 115, 2021 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-34504096

RÉSUMÉ

Circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) are two cancer-derived blood biomarkers that inform on patient prognosis and treatment efficacy in breast cancer. We prospectively evaluated the clinical validity of quantifying both CTCs (CellSearch) and ctDNA (targeted next-generation sequencing). Their combined value as prognostic and early monitoring markers was assessed in 198 HER2-negative metastatic breast cancer patients. All patients were included in the prospective multicenter UCBG study COMET (NCT01745757) and treated by first-line chemotherapy with weekly paclitaxel and bevacizumab. Blood samples were obtained at baseline and before the second cycle of chemotherapy. At baseline, CTCs and ctDNA were respectively detected in 72 and 74% of patients and were moderately correlated (Kendall's τ = 0.3). Only 26 (13%) patients had neither detectable ctDNA nor CTCs. Variants were most frequently observed in TP53 and PIK3CA genes. KMT2C/MLL3 variants detected in ctDNA were significantly associated with a lower CTC count, while the opposite trend was seen with GATA3 alterations. Both CTC and ctDNA levels at baseline and after four weeks of treatment were correlated with survival. For progression-free and overall survival, the best multivariate prognostic model included tumor subtype (triple negative vs other), grade (grade 3 vs other), ctDNA variant allele frequency (VAF) at baseline (per 10% increase), and CTC count at four weeks (≥5CTC/7.5 mL). Overall, this study demonstrates that CTCs and ctDNA have nonoverlapping detection profiles and complementary prognostic values in metastatic breast cancer patients. A comprehensive liquid-biopsy approach may involve simultaneous detection of ctDNA and CTCs.

5.
Acta Cytol ; 65(1): 88-98, 2021.
Article de Anglais | MEDLINE | ID: mdl-33011718

RÉSUMÉ

OBJECTIVE: Although transcriptomic assessments of small samples using high-throughput techniques are usually performed on fresh or frozen tissues, there is a growing demand for those performed on stained cellular specimens already used for diagnostic purposes. STUDY DESIGN: The possibility of detecting mRNAs and microRNAs (miRNAs) from routinely processed cytological samples using nCounter® technology was explored. Fresh samples from pleural and peritoneal effusions were analyzed using 2 parallel methods: samples were smeared and routinely stained using the May-Grünwald-Giemsa or Diff-Quik® method and mounted using conventional methods, and they were also studied following a snap freezing method, in which samples were maintained at -80°C until use. mRNAs and miRNAs were assessed and compared after total RNA extraction from both routinely processed samples and their matched frozen controls. RESULTS: A good concordance was found between the gene expression measured in routinely processed samples and their matched frozen controls for the majority of mRNAs and miRNAs tested. However, the standard deviation of low-expressed miRNA was high. CONCLUSIONS: Although nCounter® technology is a robust method to measure and characterize both mRNAs and miRNAs from routinely processed cytological samples, caution is recommended for the interpretation of low-expressed miRNA.


Sujet(s)
Techniques cytologiques/méthodes , Analyse de profil d'expression de gènes/méthodes , Expression des gènes/génétique , microARN/génétique , ARN messager/génétique , Colorants azurés/composition chimique , Éosine jaunâtre/composition chimique , Humains , Bleu de méthylène/composition chimique , Inclusion en paraffine/méthodes , Étude de validation de principe , Xanthènes/composition chimique
6.
J Cell Mol Med ; 24(24): 14453-14466, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33159500

RÉSUMÉ

In haemophilia, the recurrence of hemarthrosis leads to irreversible arthropathy termed haemophilic arthropathy (HA). However, HA is a unique form of arthropathy in which resident cells, such as fibroblast-like synoviocytes (FLS), come into direct contact with blood. Therefore, we hypothesized that FLS in HA could have a unique inflammatory signature as a consequence of their contact with blood. We demonstrated with ELISA and ELISPOT analyses that HA-FLS expressed a unique profile of cytokine secretion, which differed from that of non-HA-FLS, mainly consisting of cytokines involved in innate immunity. We showed that unstable cytokine mRNAs were involved in this process, especially through miRNA complexes as confirmed by DICER silencing. A miRNOME analysis revealed that 30 miRNAs were expressed differently between HA and non-HA-FLS, with most miRNAs involved in inflammatory control pathways or described in certain inflammatory diseases, such as rheumatoid arthritis or lupus. Analysis of transcriptomic networks, impacted by these miRNAs, revealed that protein processes and inflammatory pathways were particularly targeted in LPS-induced FLS, and in particular vascularization and osteoarticular modulation pathways in steady-state FLS. Our study demonstrates that the presence of blood in contact with FLS may induce durable miRNA changes that likely participate in HA pathophysiology.


Sujet(s)
Marqueurs biologiques , Hémarthrose/étiologie , Hémarthrose/métabolisme , microARN/génétique , Cellules synoviales/métabolisme , Communication cellulaire , Cytokines/métabolisme , Prédisposition aux maladies , Épigenèse génétique , Fibroblastes/métabolisme , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Hémarthrose/anatomopathologie , Hémophilie A/complications , Humains , Immunophénotypage , Médiateurs de l'inflammation/métabolisme , Protéomique/méthodes , Transduction du signal
7.
Nucleic Acids Res ; 46(21): 11405-11422, 2018 11 30.
Article de Anglais | MEDLINE | ID: mdl-30321409

RÉSUMÉ

Tauopathies such as Alzheimer's Disease (AD) are neurodegenerative disorders for which there is presently no cure. They are named after the abnormal oligomerization/aggregation of the neuronal microtubule-associated Tau protein. Besides its role as a microtubule-associated protein, a DNA-binding capacity and a nuclear localization for Tau protein has been described in neurons. While questioning the potential role of Tau-DNA binding in the development of tauopathies, we have carried out a large-scale analysis of the interaction of Tau protein with the neuronal genome under physiological and heat stress conditions using the ChIP-on-chip technique that combines Chromatin ImmunoPrecipitation (ChIP) with DNA microarray (chip). Our findings show that Tau protein specifically interacts with genic and intergenic DNA sequences of primary culture of neurons with a preference for DNA regions positioned beyond the ±5000 bp range from transcription start site. An AG-rich DNA motif was found recurrently present within Tau-interacting regions and 30% of Tau-interacting regions overlapped DNA sequences coding for lncRNAs. Neurological processes affected in AD were enriched among Tau-interacting regions with in vivo gene expression assays being indicative of a transcriptional repressor role for Tau protein, which was exacerbated in neurons displaying nuclear pathological oligomerized forms of Tau protein.


Sujet(s)
ADN intergénique/génétique , ADN/composition chimique , Neurones/métabolisme , Protéines tau/génétique , Maladie d'Alzheimer/génétique , Animaux , Encéphale/embryologie , Immunoprécipitation de la chromatine , Hyperthermie provoquée , Souris , Souris knockout , Séquençage par oligonucléotides en batterie , Phosphorylation , Liaison aux protéines , Tauopathies , Facteurs de transcription/métabolisme , Ubiquitin-protein ligases/métabolisme
8.
Lancet Oncol ; 19(4): 549-561, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29475724

RÉSUMÉ

BACKGROUND: Patients with follicular lymphoma have heterogeneous outcomes. Predictor models to distinguish, at diagnosis, between patients at high and low risk of progression are needed. The objective of this study was to use gene-expression profiling data to build and validate a predictive model of outcome for patients treated in the rituximab era. METHODS: A training set of fresh-frozen tumour biopsies was prospectively obtained from 160 untreated patients with high-tumour-burden follicular lymphoma enrolled in the phase 3 randomised PRIMA trial, in which rituximab maintenance was evaluated after rituximab plus chemotherapy induction (median follow-up 6·6 years [IQR 6·0-7·0]). RNA of sufficient quality was obtained for 149 of 160 cases, and Affymetrix U133 Plus 2.0 microarrays were used for gene-expression profiling. We did a multivariate Cox regression analysis to identify genes with expression levels associated with progression-free survival independently of maintenance treatment in a subgroup of 134 randomised patients. Expression levels from 95 curated genes were then determined by digital expression profiling (NanoString technology) in 53 formalin-fixed paraffin-embedded samples of the training set to compare the technical reproducibility of expression levels for each gene between technologies. Genes with high correlation (>0·75) were included in an L2-penalised Cox model adjusted on rituximab maintenance to build a predictive score for progression-free survival. The model was validated using NanoString technology to digitally quantify gene expression in 488 formalin-fixed, paraffin-embedded samples from three independent international patient cohorts from the PRIMA trial (n=178; distinct from the training cohort), the University of Iowa/Mayo Clinic Lymphoma SPORE project (n=201), and the Barcelona Hospital Clinic (n=109). All tissue samples consisted of pretreatment diagnostic biopsies and were confirmed as follicular lymphoma grade 1-3a. The patients were all treated with regimens containing rituximab and chemotherapy, possibly followed by either rituximab maintenance or ibritumomab-tiuxetan consolidation. We determined an optimum threshold on the score to predict patients at low risk and high risk of progression. The model, including the multigene score and the threshold, was initially evaluated in the three validation cohorts separately. The sensitivity and specificity of the score for the prediction of the risk of lymphoma progression at 2 years were assessed on the combined validation cohorts. FINDINGS: In the training cohort, the expression levels of 395 genes were associated with a risk of progression. 23 genes reflecting both B-cell biology and tumour microenvironment with correlation coefficients greater than 0·75 between the two technologies and sample types were retained to build a predictive model that identified a population at an increased risk of progression (p<0·0001). In a multivariate Cox model for progression-free survival adjusted on rituximab maintenance treatment and Follicular Lymphoma International Prognostic Index 1 (FLIPI-1) score, this predictor independently predicted progression (adjusted hazard ratio [aHR] of the high-risk group compared with the low-risk group 3·68, 95% CI 2·19-6·17 [p<0·0001]). The 5-year progression-free survival was 26% (95% CI 16-43) in the high-risk group and 73% (64-83) in the low-risk group. The predictor performances were confirmed in each of the individual validation cohorts (aHR comparing high-risk to low-risk groups 2·57 [95% CI 1·65-4·01] in cohort 1; 2·12 [1·32-3·39] in cohort 2; and 2·11 [1·01-4·41] in cohort 3). In the combined validation cohort, the median progression-free survival was 3·1 years (95% CI 2·4-4·8) in the high-risk group and 10·8 years (10·1-not reached) in the low-risk group (p<0·0001). The risk of lymphoma progression at 2 years was 38% (95% CI 29-46) in the high-risk group and 19% (15-24) in the low-risk group. In a multivariate analysis, the score predicted progression-free survival independently of anti-CD20 maintenance treatment and of the FLIPI score (aHR for the combined cohort 2·30, 95% CI 1·72-3·07). INTERPRETATION: We developed and validated a robust 23-gene expression-based predictor of progression-free survival that is applicable to routinely available formalin-fixed, paraffin-embedded tumour biopsies from patients with follicular lymphoma at time of diagnosis. Applying this score could allow individualised therapy for patients according to their risk category. FUNDING: Roche, SIRIC Lyric, LYSARC, National Institutes of Health, the Henry J Predolin Foundation, and the Spanish Plan Nacional de Investigacion.


Sujet(s)
Analyse de profil d'expression de gènes , Lymphome folliculaire/traitement médicamenteux , Lymphome folliculaire/génétique , ARN tumoral/analyse , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Essais cliniques de phase III comme sujet , Femelle , Humains , Internationalité , Chimiothérapie de maintenance , Mâle , Adulte d'âge moyen , Séquençage par oligonucléotides en batterie , Valeur prédictive des tests , Survie sans progression , Modèles des risques proportionnels , Essais contrôlés randomisés comme sujet , Études rétrospectives , Appréciation des risques/méthodes , Rituximab/administration et posologie
9.
Proc Natl Acad Sci U S A ; 115(3): E488-E497, 2018 01 16.
Article de Anglais | MEDLINE | ID: mdl-29282317

RÉSUMÉ

The contribution of host genetic and nongenetic factors to immunological differences in humans remains largely undefined. Here, we generated bacterial-, fungal-, and viral-induced immune transcriptional profiles in an age- and sex-balanced cohort of 1,000 healthy individuals and searched for the determinants of immune response variation. We found that age and sex affected the transcriptional response of most immune-related genes, with age effects being more stimulus-specific relative to sex effects, which were largely shared across conditions. Although specific cell populations mediated the effects of age and sex on gene expression, including CD8+ T cells for age and CD4+ T cells and monocytes for sex, we detected a direct effect of these intrinsic factors for the majority of immune genes. The mapping of expression quantitative trait loci (eQTLs) revealed that genetic factors had a stronger effect on immune gene regulation than age and sex, yet they affected a smaller number of genes. Importantly, we identified numerous genetic variants that manifested their regulatory effects exclusively on immune stimulation, including a Candida albicans-specific master regulator at the CR1 locus. These response eQTLs were enriched in disease-associated variants, particularly for autoimmune and inflammatory disorders, indicating that differences in disease risk may result from regulatory variants exerting their effects only in the presence of immune stress. Together, this study quantifies the respective effects of age, sex, genetics, and cellular heterogeneity on the interindividual variability of immune responses and constitutes a valuable resource for further exploration in the context of different infection risks or disease outcomes.


Sujet(s)
Vieillissement , Régulation de l'expression des gènes/immunologie , Variation génétique , Adulte , Sujet âgé , Bactéries/immunologie , Études de cohortes , Entérotoxines/immunologie , Femelle , Champignons/immunologie , Génotype , Humains , Virus de la grippe A/immunologie , Mâle , Adulte d'âge moyen , Locus de caractère quantitatif , Jeune adulte
10.
PLoS One ; 12(10): e0185753, 2017.
Article de Anglais | MEDLINE | ID: mdl-29045452

RÉSUMÉ

PURPOSE: The Prosigna® breast cancer prognostic gene signature assay identifies a gene-expression profile that permits the classification of tumors into subtypes and gives a score for the risk of recurrence (ROR) at 10 years. The primary objective of this multicenter study was to evaluate the impact of Prosigna's assay information on physicians' adjuvant treatment decisions in patients with early-stage breast cancer. Secondary objectives were to assess confidence of practitioners in their therapeutic recommendations before and after the added information provided by the Prosigna assay; and to evaluate the emotional state of patients before and after the Prosigna test results. METHODS: Consecutive patients with invasive early-stage breast cancer were enrolled in a prospective, observational, multicenter study carried out in 8 hospitals in France. The Prosigna test was carried out on surgical specimens using the nCounter® Analysis System located at the Institut Curie. Both before and after receiving the Prosigna test results, physicians completed treatment confidence questionnaires and patients completed questionnaires concerning their state of anxiety, the difficulties felt in face of the therapy and quality of life. Information was also collected at 6 months regarding the physicians' opinion on the test results and the patients' degree of anxiety, difficulties with therapy and quality of life. RESULTS: Between March 2015 and January 2016, 8 study centers in France consecutively enrolled 210 postmenopausal women with estrogen receptor (ER) positive, human epidermal growth hormone-2 (HER-2) negative, and node negative tumors, either stage 1 or stage 2. Intrinsic tumor subtypes as assessed by the Prosigna test were 114 (58.2%) Luminal A, 79 (40.3%) Luminal B, 1 (0.5%) HER-2 enriched (HER-2E), and 2 (1.0%) basal-like. Before receiving the Prosigna test results, physicians categorized tumor subtypes based on immunohistochemistry (IHC) as Luminal A in 126 (64%) patients and Luminal B in 70 (36%) patients, an overall discordance rate of 25%. The availability of Prosigna assay results was significantly associated with the likelihood of change in treatment recommendations, with 34 patients (18%) having their treatment plan changed from Adjuvant Chemotherapy to No Adjuvant Chemotherapy or vice versa (p<0.001, Fisher's exact test). Prosigna test results also decreased patients' anxiety about the chosen adjuvant therapy, and improved emotional well-being and measures of personal perceptions of uncertainty. CONCLUSIONS: The results of this prospective decision impact study are consistent with 2 previous, identically designed studies carried out in Spain and Germany. The availability of Prosigna test results increased the confidence of treating physicians in their adjuvant treatment decisions, and led to an 18% change in chemotherapy treatment plan (from Adjuvant Chemotherapy to No Adjuvant Chemotherapy or vice versa). Prosigna testing decreased anxiety and improved measures of health-related quality of life in patients facing adjuvant therapy. The 25% discordance between Prosigna test and IHC subtyping underlines the importance of molecular testing for optimal systemic therapy indications in early breast cancer.


Sujet(s)
Tumeurs du sein/diagnostic , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Traitement médicamenteux adjuvant , Femelle , France , Directives de santé publique , Humains , Adulte d'âge moyen , Récidive tumorale locale/anatomopathologie , Stadification tumorale , Médecins , Études prospectives , Résultat thérapeutique
11.
Oncotarget ; 8(21): 34245-34257, 2017 May 23.
Article de Anglais | MEDLINE | ID: mdl-28427232

RÉSUMÉ

Extra-cranial rhabdoid tumors (RT) are highly aggressive malignancies of infancy, characterized by undifferentiated histological features and loss of SMARCB1 expression. The diagnosis is all the more challenging that other poorly differentiated cancers lose SMARCB1 expression, such as epithelioid sarcomas (ES), renal medullary carcinomas (RMC) or undifferentiated chordomas (UC). Moreover, late cases occurring in adults are now increasingly reported, raising the question of differential diagnoses and emphasizing nosological issues. To address this issue, we have analyzed the expression profiles of a training set of 32 SMARCB1-deficient tumors (SDT), with ascertained diagnosis of RT (n = 16, all < 5 years of age), ES (n = 8, all > 10 years of age), UC (n = 3) and RMC (n = 5). As compared with other SDT, RT are characterized by an embryonic signature, and up-regulation of key-actors of de novo DNA methylation processes. Using this signature, we then analysed the expression profiling of 37 SDT to infer the appropriate diagnosis. Thirteen adult onset tumors showed strong similarity with pediatric RT, in spite of older age; by exome sequencing, these tumors also showed genomic features indistinguishable from pediatric RT. In contrary, 8 tumors were reclassified within carcinoma, ES or UC categories, while the remaining could not be related to any of those entities. Our results demonstrate that embryonic signature is shared by all RT, whatever the age at diagnosis; they also illustrate that many adult-onset SDT of ambiguous histological diagnosis are clearly different from RT. Finally, our study paves the way for the routine use of expression-based signatures to give accurate diagnosis of SDT.


Sujet(s)
/méthodes , Analyse de profil d'expression de gènes/méthodes , Tumeur rhabdoïde/diagnostic , Tumeur rhabdoïde/génétique , Protéine SMARCB1/déficit , Adolescent , Adulte , Âge de début , Enfant , Enfant d'âge préscolaire , Méthylation de l'ADN , Diagnostic différentiel , Réseaux de régulation génique , Humains , Nourrisson , Mâle , Jeune adulte
12.
Hum Mol Genet ; 26(1): 90-108, 2017 01 01.
Article de Anglais | MEDLINE | ID: mdl-28007902

RÉSUMÉ

Human doublecortin (DCX) mutations are associated with severe brain malformations leading to aberrant neuron positioning (heterotopia), intellectual disability and epilepsy. The Dcx protein plays a key role in neuronal migration, and hippocampal pyramidal neurons in Dcx knockout (KO) mice are disorganized. The single CA3 pyramidal cell layer observed in wild type (WT) is present as two abnormal layers in the KO, and CA3 KO pyramidal neurons are more excitable than WT. Dcx KO mice also exhibit spontaneous epileptic activity originating in the hippocampus. It is unknown, however, how hyperexcitability arises and why two CA3 layers are observed.Transcriptome analyses were performed to search for perturbed postnatal gene expression, comparing Dcx KO CA3 pyramidal cell layers with WT. Gene expression changes common to both KO layers indicated mitochondria and Golgi apparatus anomalies, as well as increased cell stress. Intriguingly, gene expression analyses also suggested that the KO layers differ significantly from each other, particularly in terms of maturity. Layer-specific molecular markers and BrdU birthdating to mark the final positions of neurons born at distinct timepoints revealed inverted layering of the CA3 region in Dcx KO animals. Notably, many early-born 'outer boundary' neurons are located in an inner position in the Dcx KO CA3, superficial to other pyramidal neurons. This abnormal positioning likely affects cell morphology and connectivity, influencing network function. Dissecting this Dcx KO phenotype sheds light on coordinated developmental mechanisms of neuronal subpopulations, as well as gene expression patterns contributing to a bi-layered malformation associated with epilepsy.


Sujet(s)
Hippocampe/métabolisme , Hippocampe/anatomopathologie , Protéines associées aux microtubules/physiologie , Neurones/métabolisme , Neurones/anatomopathologie , Neuropeptides/physiologie , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Encéphale/ultrastructure , Région CA1 de l'hippocampe/métabolisme , Région CA1 de l'hippocampe/anatomopathologie , Région CA1 de l'hippocampe/ultrastructure , Région CA3 de l'hippocampe/métabolisme , Région CA3 de l'hippocampe/anatomopathologie , Région CA3 de l'hippocampe/ultrastructure , Protéines à domaine doublecortine , Protéine doublecortine , Femelle , Hippocampe/ultrastructure , Traitement d'image par ordinateur , Microdissection au laser , Mâle , Souris , Souris knockout , Microscopie confocale , Neurones/ultrastructure
13.
Cell Rep ; 16(10): 2777-2791, 2016 09 06.
Article de Anglais | MEDLINE | ID: mdl-27568558

RÉSUMÉ

Systems approaches for the study of immune signaling pathways have been traditionally based on purified cells or cultured lines. However, in vivo responses involve the coordinated action of multiple cell types, which interact to establish an inflammatory microenvironment. We employed standardized whole-blood stimulation systems to test the hypothesis that responses to Toll-like receptor ligands or whole microbes can be defined by the transcriptional signatures of key cytokines. We found 44 genes, identified using Support Vector Machine learning, that captured the diversity of complex innate immune responses with improved segregation between distinct stimuli. Furthermore, we used donor variability to identify shared inter-cellular pathways and trace cytokine loops involved in gene expression. This provides strategies for dimension reduction of large datasets and deconvolution of innate immune responses applicable for characterizing immunomodulatory molecules. Moreover, we provide an interactive R-Shiny application with healthy donor reference values for induced inflammatory genes.


Sujet(s)
Sang/métabolisme , Analyse de profil d'expression de gènes/méthodes , Immunité/génétique , Transcription génétique , Adulte , Bactéries/métabolisme , Cytokines/pharmacologie , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Immunité/effets des médicaments et des substances chimiques , Lymphocytes/métabolisme , Mâle , Récepteurs de type Toll/métabolisme , Transcription génétique/effets des médicaments et des substances chimiques
14.
Clin Cancer Res ; 20(16): 4314-25, 2014 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-24947930

RÉSUMÉ

PURPOSE: Patients with luminal breast cancer (LBC) often become endocrine resistant over time. We investigated the molecular changes associated with acquired hormonoresistances in patient-derived xenografts of LBC. EXPERIMENTAL DESIGN: Two LBC xenografts (HBCx22 and HBCx34) were treated with different endocrine treatments (ET) to obtain xenografts with acquired resistances to tamoxifen (TamR) and ovariectomy (OvaR). PI3K pathway activation was analyzed by Western blot analysis and IHC and responses to ET combined to everolimus were investigated in vivo. Gene expression analyses were performed by RT-PCR and Affymetrix arrays. RESULTS: HBCx22 TamR xenograft was cross-resistant to several hormonotherapies, whereas HBCx22 OvaR and HBCx34 TamR exhibited a treatment-specific resistance profile. PI3K pathway was similarly activated in parental and resistant xenografts but the addition of everolimus did not restore the response to tamoxifen in TamR xenografts. In contrast, the combination of fulvestrant and everolimus induced tumor regression in vivo in HBCx34 TamR, where we found a cross-talk between the estrogen receptor (ER) and PI3K pathways. Expression of several ER-controlled genes and ER coregulators was significantly changed in both TamR and OvaR tumors, indicating impaired ER transcriptional activity. Expression changes associated with hormonoresistance were both tumor and treatment specific and were enriched for genes involved in cell growth, cell death, and cell survival. CONCLUSIONS: PDX models of LBC with acquired resistance to endocrine therapies show a great diversity of resistance phenotype, associated with specific deregulations of ER-mediated gene transcription. These models offer a tool for developing anticancer therapies and to investigate the dynamics of resistance emerging during pharmacologic interventions. Clin Cancer Res; 20(16); 4314-25. ©2014 AACR.


Sujet(s)
Antinéoplasiques hormonaux/pharmacologie , Tumeurs du sein/traitement médicamenteux , Résistance aux médicaments antinéoplasiques , Récepteur alpha des oestrogènes/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Récepteur ErbB-2/métabolisme , Tamoxifène/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Technique de Western , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteur alpha des oestrogènes/génétique , Femelle , Humains , Techniques immunoenzymatiques , Souris , Souris nude , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , Récepteur ErbB-2/génétique , RT-PCR , Transduction du signal , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
15.
Carcinogenesis ; 34(5): 1173-80, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23358853

RÉSUMÉ

Telomere shortening is a major source of chromosome instability (CIN) at early stages during carcinogenesis. However, the mechanisms through which telomere-driven CIN (T-CIN) contributes to the acquisition of tumor phenotypes remain uncharacterized. We discovered that human epithelial kidney cells undergoing T-CIN display massive microRNA (miR) expression changes that are not related to local losses or gains. This widespread miR deregulation encompasses a miR-200-dependent epithelial-to-mesenchymal transition (EMT) that confers to immortalized pre-tumoral cells phenotypic traits of metastatic potential. Remarkably, a miR signature of these cells, comprising a downregulation of miRs with conserved expression in kidney, was retrieved in poorly differentiated aggressive renal cell carcinomas. Our results reveal an unanticipated connection between telomere crisis and the activation of the EMT program that occurs at pre-invasive stages of epithelial cancers, through mechanisms that involve miR deregulation. Thus, this study provides a new rational into how telomere instability contributes to the acquisition of the malignant phenotype.


Sujet(s)
Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , microARN/génétique , Télomère/génétique , Lignée cellulaire tumorale , Instabilité des chromosomes/génétique , Altération de l'ADN/génétique , Cellules épithéliales/anatomopathologie , Transition épithélio-mésenchymateuse/génétique , Cellules HEK293 , Humains , Transcription génétique/génétique
16.
J Virol ; 86(20): 11333-44, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22896612

RÉSUMÉ

Rift Valley fever virus (RVFV) is a highly pathogenic Phlebovirus that infects humans and ruminants. Initially confined to Africa, RVFV has spread outside Africa and presently represents a high risk to other geographic regions. It is responsible for high fatality rates in sheep and cattle. In humans, RVFV can induce hepatitis, encephalitis, retinitis, or fatal hemorrhagic fever. The nonstructural NSs protein that is the major virulence factor is found in the nuclei of infected cells where it associates with cellular transcription factors and cofactors. In previous work, we have shown that NSs interacts with the promoter region of the beta interferon gene abnormally maintaining the promoter in a repressed state. In this work, we performed a genome-wide analysis of the interactions between NSs and the host genome using a genome-wide chromatin immunoprecipitation combined with promoter sequence microarray, the ChIP-on-chip technique. Several cellular promoter regions were identified as significantly interacting with NSs, and the establishment of NSs interactions with these regions was often found linked to deregulation of expression of the corresponding genes. Among annotated NSs-interacting genes were present not only genes regulating innate immunity and inflammation but also genes regulating cellular pathways that have not yet been identified as targeted by RVFV. Several of these pathways, such as cell adhesion, axonal guidance, development, and coagulation were closely related to RVFV-induced disorders. In particular, we show in this work that NSs targeted and modified the expression of genes coding for coagulation factors, demonstrating for the first time that this hemorrhagic virus impairs the host coagulation cascade at the transcriptional level.


Sujet(s)
Facteurs de la coagulation sanguine/génétique , ADN/génétique , Régions promotrices (génétique) , Séquences d'acides nucléiques régulatrices , Virus de la fièvre de la vallée du Rift/génétique , Virus de la fièvre de la vallée du Rift/métabolisme , Protéines virales non structurales/métabolisme , Animaux , Chlorocebus aethiops , Immunoprécipitation de la chromatine , ADN/métabolisme , Étude d'association pangénomique , Interactions hôte-pathogène/génétique , Interféron bêta/génétique , Analyse par réseau de protéines , ARN messager/génétique , Fièvre de la Vallée du Rift/génétique , Fièvre de la Vallée du Rift/anatomopathologie , Virus de la fièvre de la vallée du Rift/pathogénicité , Transcription génétique , Cellules Vero , Protéines virales non structurales/analyse
17.
Cancer Res ; 71(3): 666-74, 2011 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-21135111

RÉSUMÉ

A high percentage of uveal melanoma patients develop metastatic tumors predominantly in the liver. We studied the molecular profiles derived from gene expression microarrays and comparative genomic hybridization microarrays, to identify genes associated with metastasis in this aggressive cancer. We compared 28 uveal melanomas from patients who developed liver metastases within three years of enucleation with 35 tumors from patients without metastases or who developed metastases more than 3 years after enucleation. Protein tyrosine phosphatase type IV A member 3 (PTP4A3/PRL3), was identified as a strong predictor of metastasis occurrence. We demonstrated that the differential expression of this gene, which maps to 8q24.3, was not merely a consequence of 8q chromosome overrepresentation. PTP4A3 overexpression in uveal melanoma cell lines significantly increased cell migration and invasiveness in vivo, suggesting a direct role for this protein in metastasis. Our findings suggest that PTP4A3 or its cellular substrates could constitute attractive therapeutic targets to treat metastatic uveal melanomas.


Sujet(s)
Marqueurs biologiques tumoraux/biosynthèse , Tumeurs du foie/secondaire , Protéines tumorales/biosynthèse , Protein Tyrosine Phosphatases/biosynthèse , Animaux , Marqueurs biologiques tumoraux/génétique , Embryon de poulet , Chromosomes humains de la paire 8 , Énucléation oculaire , Femelle , Expression des gènes , Humains , Tumeurs du foie/enzymologie , Tumeurs du foie/génétique , Mâle , Mélanome/enzymologie , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/chirurgie , Adulte d'âge moyen , Protéines tumorales/génétique , Protein Tyrosine Phosphatases/génétique , Taux de survie , Tumeurs de l'uvée/enzymologie , Tumeurs de l'uvée/génétique , Tumeurs de l'uvée/anatomopathologie , Tumeurs de l'uvée/chirurgie
18.
Hum Pathol ; 35(7): 817-24, 2004 Jul.
Article de Anglais | MEDLINE | ID: mdl-15257544

RÉSUMÉ

The INK4a/ARF locus encodes 2 cell cycle regulatory proteins: p16 and p14(ARF). P16 inhibits the activities of cdks, which maintain the retinoblastoma protein (pRb) in its active hypophosphorylated state. P14(ARF) blocks MDM2-induced p53 degradation and transactivational silencing. In this study, we investigated the expression of p16 and p14(ARF) in reference human urothelium and in 51 urothelial carcinomas (UCs) of all stages and grades, by reverse transcription-polymerase chain reaction (RT-PCR). Patterns of p14(ARF) and p16 expression were compared with each other and then with patterns of p53 and pRb protein expression, respectively, as determined by immunohistochemistry. P14(ARF) and p16 mRNAs were present at low levels or were undetectable in reference urothelia and in most superficial tumors, whereas they were present at high levels in a subset of tumors of advanced stage and high grade. The expression profiles of these 2 mRNAs were correlated in all but 4 cases, indicating that the 2 INK4a products may have nonredundant functions. Forty-six of the 51 tumors (90%) presented changes to or a lack of activation of the p14(ARF)-p53 pathway and were p53 positive (n = 10), p14(ARF) negative (n = 23), or both p53 positive and p14(ARF) negative (n = 13), suggesting that these 2 components of the pathway may be altered or nonactivated. Markedly high levels of p16 mRNA (n = 5) were associated with the absence of pRb expression, with the exception of 1 case in which the p16 gene contained a deletion mutation. A lack of p16 mRNA or low levels of this mRNA were associated with pRb detection in all but 1 case. In invasive UCs, the p16-pRb pathway, the p14(ARF)-p53 pathway, or in many cases both pathways were altered or not activated, demonstrating the involvement of these pathways in invasive bladder tumorigenesis.


Sujet(s)
Carcinomes/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Protéine du rétinoblastome/métabolisme , Protéine p14(ARF) suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Tumeurs de la vessie urinaire/métabolisme , Urothélium/métabolisme , Carcinomes/génétique , Carcinomes/anatomopathologie , Inhibiteur p16 de kinase cycline-dépendante/génétique , Amorces ADN/composition chimique , Régulation de l'expression des gènes tumoraux , Humains , Immunohistochimie , ARN messager/génétique , ARN messager/métabolisme , ARN tumoral/analyse , Protéine du rétinoblastome/génétique , RT-PCR , Protéine p14(ARF) suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/génétique , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Urothélium/anatomie et histologie , Urothélium/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...