Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 44
Filtrer
1.
J Clin Oncol ; 40(26): 3077-3087, 2022 09 10.
Article de Anglais | MEDLINE | ID: mdl-35442716

RÉSUMÉ

PURPOSE: Cisplatin is the main systemic treatment modality for male type II germ cell tumors (GCTs). Although generally very effective, 5%-10% of patients suffer from cisplatin-resistant disease. Identification of the driving mechanisms of resistance will enable improved risk stratification and development of alternative treatments. METHODS: We developed and characterized cisplatin-resistant GCT cell line models and compared their molecular characteristics with patient samples with cisplatin resistance and/or a poor clinical outcome. Subsequently, the association between the overlapping genetic features and clinical data was assessed. Finally, we used Cox regression to determine the prognostic relevance of these features within the currently used risk classification. RESULTS: Gain of chromosome 3p25.3 was detected in all cisplatin-resistant cell lines, and copy number of this region correlated with the level of resistance (R = 0.96, P = 1.5e-04). Gain of this region was detected at low frequencies in primary tumors and at higher frequencies in relapsed and/or cisplatin-resistant tumors. Chromosome 3p25.3 gain was associated with shorter progression-free survival and overall survival, with the strongest association observed in nonseminomas excluding pure teratomas. 3p25.3 gain was more frequently observed in tumors with yolk sac tumor histology and predicted adverse outcome independent of the International Germ Cell Cancer Collaborative Group risk classification and the presence of TP53/MDM2 alterations. CONCLUSION: On the basis of both in vitro analyses and clinical data, we found 3p25.3 to be strongly associated with cisplatin resistance and poor clinical outcome in male type II GCTs. Using genomic profiling, 3p25.3 status could help to improve risk stratification in male patients with type II GCT. Further characterization of this locus and underlying mechanisms of resistance is warranted to guide development of novel treatment approaches for cisplatin-resistant disease.


Sujet(s)
Tumeurs embryonnaires et germinales , Tumeurs du testicule , Aberrations des chromosomes , Chromosomes/métabolisme , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Résistance aux médicaments antinéoplasiques/génétique , Humains , Mâle , Tumeurs embryonnaires et germinales/traitement médicamenteux , Tumeurs embryonnaires et germinales/génétique , Tumeurs du testicule/traitement médicamenteux , Tumeurs du testicule/génétique , Tumeurs du testicule/anatomopathologie
2.
Cancer Cell Int ; 20: 364, 2020.
Article de Anglais | MEDLINE | ID: mdl-32774158

RÉSUMÉ

BACKGROUND: Cisplatin resistance of ovarian yolk sac tumors (oYST) is a clinical challenge due to dismal patient prognosis, even though the disease is extremely rare. We investigated potential association between cisplatin resistance and cancer stem cell (CSC) markers in chemoresistant oYST cells and targeting strategies to overcome resistance in oYST. METHODS: Chemoresistant cells were derived from chemosensitive human oYST cells by cultivation in cisplatin in vitro. Derivative cells were characterized by chemoresistance, functional assays, flow cytometry, gene expression and protein arrays focused on CSC markers. RNAseq, methylation and microRNA profiling were performed. Quail chorioallantoic membranes (CAM) with implanted oYST cells were used to analyze the micro-tumor extent and interconnection with the CAM. Tumorigenicity in vivo was determined on immunodeficient mouse model. Chemoresistant cells were treated by inhibitors intefering with the CSC properties to examine the chemosensitization to cisplatin. RESULTS: Long-term cisplatin exposure resulted in seven-fold higher IC50 value in resistant cells, cross-resistance to oxaliplatin and carboplatin, and increased migratory capacity, invasiveness and tumorigenicity, associated with hypomethylation of differentially methylated genes/promotors. Resistant cells exhibited increased expression of prominin-1 (CD133), ATP binding cassette subfamily G member 2 (ABCG2), aldehyde dehydrogenase 3 isoform A1 (ALDH3A1), correlating with reduced gene and promoter methylation, as well as increased expression of ALDH1A3 and higher overall ALDH enzymatic activity, rendering them cross-resistant to DEAB, disulfiram and napabucasin. Salinomycin and tunicamycin were significantly more toxic to resistant cells. Pretreatment with napabucasin resensitized the cells to cisplatin and reduced their tumorigenicity in vivo. CONCLUSIONS: The novel chemoresistant cells represent unique model of refractory oYST. CSC markers are associated with cisplatin resistance being possible targets in chemorefractory oYST.

3.
PLoS One ; 15(3): e0230253, 2020.
Article de Anglais | MEDLINE | ID: mdl-32176716

RÉSUMÉ

Autologous transplantation of spermatogonial stem cells is a promising new avenue to restore fertility in infertile recipients. Expansion of the initial spermatogonial stem cell pool through cell culturing is a necessary step to obtain enough cells for effective repopulation of the testis after transplantation. Since in vitro propagation can lead to (epi-)genetic mutations and possibly malignant transformation of the starting cell population, we set out to investigate genome-wide DNA methylation status in uncultured and cultured primary testicular ITGA6+ sorted cells and compare them with germ cell tumor samples of the seminoma subtype. Seminomas displayed a severely global hypomethylated profile, including loss of genomic imprinting, which we did not detect in cultured primary testicular ITGA6+ cells. Differential methylation analysis revealed altered regulation of gamete formation and meiotic processes in cultured primary testicular ITGA6+ cells but not in seminomas. The pivotal POU5F1 marker was hypomethylated in seminomas but not in uncultured or cultured primary testicular ITGA6+ cells, which is reflected in the POU5F1 mRNA expression levels. Lastly, seminomas displayed a number of characteristic copy number variations that were not detectable in primary testicular ITGA6+ cells, either before or after culture. Together, the data show a distinct DNA methylation patterns in cultured primary testicular ITGA6+ cells that does not resemble the pattern found in seminomas, but also highlight the need for more sensitive methods to fully exclude the presence of malignant cells after culture and to further study the epigenetic events that take place during in vitro culture.


Sujet(s)
Variations de nombre de copies de segment d'ADN/génétique , Méthylation de l'ADN/génétique , Instabilité du génome/génétique , Intégrine alpha6/génétique , Séminome/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques/métabolisme , Cellules cultivées , Épigenèse génétique/génétique , Empreinte génomique/génétique , Humains , Mâle , Adulte d'âge moyen , Mutation/génétique , Tumeurs embryonnaires et germinales , Facteur de transcription Oct-3/génétique , Tumeurs du testicule/génétique , Testicule/métabolisme
5.
Cells ; 8(12)2019 12 14.
Article de Anglais | MEDLINE | ID: mdl-31847394

RÉSUMÉ

Liquid biopsy-based biomarkers, such as microRNAs, represent valuable tools for patient management, but often do not make it to integration in the clinic. We aim to explore issues impeding this transition, in the setting of germ cell tumors, for which novel biomarkers are needed. We describe a model for identifying and validating clinically relevant microRNAs for germ cell tumor patients, using both in vitro, in vivo (mouse model) and patient-derived data. Initial wide screening of candidate microRNAs is performed, followed by targeted profiling of potentially relevant biomarkers. We demonstrate the relevance of appropriate (negative) controls, experimental conditions (proliferation), and issues related to sample origin (serum, plasma, cerebral spinal fluid) and pre-analytical variables (hemolysis, contaminants, temperature), all of which could interfere with liquid biopsy-based studies and their conclusions. Finally, we show the value of our identification model in a specific scenario, contradicting the presumed role of miR-375 as marker of teratoma histology in liquid biopsy setting. Our findings indicate other putative microRNAs (miR-885-5p, miR-448 and miR-197-3p) fulfilling this clinical need. The identification model is informative to identify the best candidate microRNAs to pursue in a clinical setting.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , microARN/génétique , Tumeurs embryonnaires et germinales/génétique , Adulte , Animaux , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/liquide cérébrospinal , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Humains , Biopsie liquide/méthodes , Mâle , Souris , Souris nude , microARN/analyse , Adulte d'âge moyen , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Br J Cancer ; 120(4): 444-452, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30739914

RÉSUMÉ

BACKGROUND: Testicular germ cell cancer (TGCC), being the most frequent malignancy in young Caucasian males, is initiated from an embryonic germ cell. This study determines intratumour heterogeneity to unravel tumour progression from initiation until metastasis. METHODS: In total, 42 purified samples of four treatment-resistant nonseminomatous (NS) TGCC were investigated, including the precursor germ cell neoplasia in situ (GCNIS) and metastatic specimens, using whole-genome and targeted sequencing. Their evolution was reconstructed. RESULTS: Intratumour molecular heterogeneity did not correspond to the supposed primary tumour histological evolution. Metastases after systemic treatment could be derived from cancer stem cells not identified in the primary cancer. GCNIS mostly lacked the molecular marks of the primary NS and comprised dominant clones that failed to progress. A BRCA-like mutational signature was observed without evidence for direct involvement of BRCA1 and BRCA2 genes. CONCLUSIONS: Our data strongly support the hypothesis that NS is initiated by whole-genome duplication, followed by chromosome copy number alterations in the cancer stem cell population, and accumulation of low numbers of somatic mutations, even in therapy-resistant cases. These observations of heterogeneity at all stages of tumourigenesis should be considered when treating patients with GCNIS-only disease, or with clinically overt NS.


Sujet(s)
Tumeurs embryonnaires et germinales/génétique , Tumeurs du testicule/génétique , Évolution moléculaire , Gène BRCA1 , Gène BRCA2 , Humains , Perte d'hétérozygotie , Mâle , Mutation , Métastase tumorale , Tumeurs embryonnaires et germinales/anatomopathologie , Tumeurs du testicule/anatomopathologie , Séquençage du génome entier
7.
Transplantation ; 103(2): 329-335, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30444806

RÉSUMÉ

BACKGROUND: Delayed graft function (DGF), a common complication after transplantation of deceased donor kidneys, affects both short- and long-term outcomes. Currently available biomarkers during graft preservation lack sensitivity in predicting risk for DGF. The aim of this study is to identify cell-free micro ribonucleic acid (miRNA) biomarkers in graft preservation fluid predictive of DGF after kidney transplantation. METHODS: Vascular bed preservation fluid was collected from 48 kidney grafts from donation after circulatory death (DCD) or donation after brain death (DBD) donors. miRNA profiles were determined by polymerase chain reaction (PCR) array (n = 8) and validated by reverse transcription and quantitative PCR (n = 40). Graft function posttransplantation was defined as immediate good function (IF) or DGF. RESULTS: A total of 223 miRNAs fulfilled the preset parameters (Ct < 40 in 3 or more samples) and were included in the analysis. Thirty-two miRNAs were significantly different between DGF and IF kidney grafts (P < 0.05) but, after correction for multiple testing, only miR-505-3p remained significant. The significant association of high miR-505-3p levels with DGF was confirmed in an independent validation cohort using conventional reverse transcription and quantitative PCR detection. Multivariate analyses showed miR-505-3p as an independent predictor for DGF (odds ratio, 1.12; P = 0.028). If stratified for donor type, miR-505-3p levels remained significantly different between IF and DGF in DCD grafts (P < 0.01), but not in DBD grafts. Receiver operating characteristic curve analysis showed a high sensitivity and specificity (area under the curve, 0.833). CONCLUSIONS: In DCD grafts, high levels of miR-505-3p in preservation fluid are associated with increased risk of DGF after kidney transplantation. Further study is required to confirm the utility of cell-free miR-505-3p as prognostic biomarker for DGF.


Sujet(s)
MicroARN circulant/analyse , Reprise retardée de fonction du greffon/étiologie , Transplantation rénale/effets indésirables , microARN/analyse , Adulte , Sujet âgé , Marqueurs biologiques , Femelle , Humains , Mâle , Adulte d'âge moyen , Courbe ROC
8.
Stem Cell Reports ; 11(6): 1493-1505, 2018 12 11.
Article de Anglais | MEDLINE | ID: mdl-30503260

RÉSUMÉ

Predicting developmental potency and risk of posttransplantation tumor formation by human pluripotent stem cells (hPSCs) and their derivatives largely rely on classical histological analysis of teratomas. Here, we investigated whether an assay based on microRNAs (miRNA) in blood plasma is able to detect potentially malignant elements. Several hPSCs and human malignant germ cell tumor (hGCT) lines were investigated in vitro and in vivo after mouse xenografting. The multiple conventional hPSC lines generated mature teratomas, while xenografts from induced hPSCs (hiPSCs) with reactivated reprogramming transgenes and hGCT lines contained undifferentiated and potentially malignant components. The presence of these elements was reflected in the mRNA and miRNA profiles of the xenografts with OCT3/4 mRNA and the miR-371 and miR-302 families readily detectable. miR-371 family members were also identified in mouse plasma faithfully reporting undifferentiated elements in the xenografts. This study demonstrated that undifferentiated and potentially malignant cells could be detected in vivo.


Sujet(s)
Dosage biologique/méthodes , Marqueurs biologiques tumoraux/sang , Différenciation cellulaire/génétique , microARN/sang , Cellules souches pluripotentes/métabolisme , Tératome/sang , Tératome/génétique , Animaux , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire , Régulation de l'expression des gènes tumoraux , Humains , Souris , microARN/génétique , Tumeurs embryonnaires et germinales/génétique , Tumeurs embryonnaires et germinales/anatomopathologie , Analyse en composantes principales , ARN messager/génétique , ARN messager/métabolisme , Facteurs temps , Tests d'activité antitumorale sur modèle de xénogreffe
9.
PLoS One ; 13(11): e0208002, 2018.
Article de Anglais | MEDLINE | ID: mdl-30496231

RÉSUMÉ

Most tumors are composed of a heterogeneous population of subclones. A more detailed insight into the subclonal evolution of these tumors can be helpful to study progression and treatment response. Problematically, tumor samples are typically very heterogeneous, making deconvolving individual tumor subclones a major challenge. To overcome this limitation, reducing heterogeneity, such as by means of microdissections, coupled with targeted sequencing, is a viable approach. However, computational methods that enable reconstruction of the evolutionary relationships require unbiased read depth measurements, which are commonly challenging to obtain in this setting. We introduce TargetClone, a novel method to reconstruct the subclonal evolution tree of tumors from single-nucleotide polymorphism allele frequency and somatic single-nucleotide variant measurements. Furthermore, our method infers copy numbers, alleles and the fraction of the tumor component in each sample. TargetClone was specifically designed for targeted sequencing data obtained from microdissected samples. We demonstrate that our method obtains low error rates on simulated data. Additionally, we show that our method is able to reconstruct expected trees in a testicular germ cell cancer and ovarian cancer dataset. The TargetClone package including tree visualization is written in Python and is publicly available at https://github.com/UMCUGenetics/targetclone.


Sujet(s)
Évolution clonale/génétique , Biologie informatique/méthodes , Tumeurs/génétique , Algorithmes , Allèles , Variations de nombre de copies de segment d'ADN/génétique , Fréquence d'allèle/génétique , Séquençage nucléotidique à haut débit/méthodes , Humains , Mutation/génétique , Polymorphisme de nucléotide simple/génétique , Logiciel
10.
PLoS One ; 13(4): e0194259, 2018.
Article de Anglais | MEDLINE | ID: mdl-29649216

RÉSUMÉ

BACKGROUND: Levothyroxine replacement treatment in hypothyroidism is unable to restore physiological thyroxine and triiodothyronine concentrations in serum and tissues completely. Normal serum thyroid stimulating hormone (TSH) concentrations reflect only pituitary euthyroidism and, therefore, novel biomarkers representing tissue-specific thyroid state are needed. MicroRNAs (miRNAs), small non-coding regulatory RNAs, exhibit tissue-specific expression patterns and can be detectable in serum. Previous studies have demonstrated differential expression of (precursors of) miRNAs in tissues under the influence of thyroid hormone. OBJECTIVE: To study if serum miRNA profiles are changed in different thyroid states. DESIGN AND METHODS: We studied 13 athyroid patients (6 males) during TSH suppressive therapy and after 4 weeks of thyroid hormone withdrawal. A magnetic bead capture system was used to isolate 384 defined miRNAs from serum. Subsequently, the TaqMan Array Card 3.0 platform was used for profiling after individual target amplification. RESULTS: Mean age of the subjects was 44.0 years (range 20-61 years). Median TSH levels were 88.9 mU/l during levothyroxine withdrawal and 0.006 mU/l during LT4 treatment with a median dosage of 2.1 µg/kg. After normalization to allow inter-sample analysis, a paired analysis did not demonstrate a significant difference in expression of any of the 384 miRNAs analyzed on and off LT4 treatment. CONCLUSION: Although we previously showed an up-regulation of pri-miRNAs 133b and 206 in hypothyroid state in skeletal muscle, the present study does not supply evidence that thyroid state also affects serum miRNAs in humans.


Sujet(s)
Hypothyroïdie/traitement médicamenteux , microARN/sang , Glande thyroide/chirurgie , Hormones thyroïdiennes/sang , Thyroxine/usage thérapeutique , Adulte , Marqueurs biologiques/sang , Femelle , Humains , Hypothyroïdie/sang , Mâle , Adulte d'âge moyen , Thyréostimuline/sang , Tri-iodothyronine/sang , Jeune adulte
11.
Sex Dev ; 12(1-3): 145-154, 2018.
Article de Anglais | MEDLINE | ID: mdl-28926831

RÉSUMÉ

We report on an adult male initially presenting with gynecomastia and a painless scrotal mass without additional genital anomalies. Hyperpigmentation of the skin following the Blaschko's lines was identified. He underwent gonadectomy because of suspected cancer. Histological analyses revealed an ovotestis with ovulatory activity confirmed by immunohistochemistry with multiple markers. Karyotyping of cultured peripheral blood lymphocytes and a buccal smear revealed a 46,XX/46,XY chimeric constitution with different percentages. Multiple molecular analyses as well as blood typing implied a tetragametic origin. After the unilateral gonadectomy, the patient developed recurrent painful cystic swellings of the remaining gonad. Because of the wish to preserve hormonal activity as well as future fertility, the patient underwent surgical resection of a cystic gonadal area. The removed tissue showed ovulation-related features in addition to both testicular and ovarian tissue, diagnosed as an ovotestis. Testosterone therapy was initiated to suppress the persistently elevated gonadotropins and thereby suppress ovarian activity. During treatment, the recurrent pain complaints and cystic swellings ceased, although gonadotropin levels were not fully suppressed. Based on these observations, the importance of a detailed genetic and pathological diagnosis and the clinical dilemmas including the pros and cons of personalized treatment with gonadal preservative surgery are discussed.


Sujet(s)
Troubles du développement sexuel de sujets 46, XX/anatomopathologie , Troubles du développement sexuel de sujets 46, XY/anatomopathologie , Troubles ovotesticulaires du développement sexuel/anatomopathologie , Ovulation , Troubles du développement sexuel de sujets 46, XX/sang , Troubles du développement sexuel de sujets 46, XX/génétique , Groupage sanguin et épreuve de compatibilité croisée , Troubles du développement sexuel de sujets 46, XY/sang , Troubles du développement sexuel de sujets 46, XY/génétique , Femelle , Gonades/anatomopathologie , Humains , Mâle , Troubles ovotesticulaires du développement sexuel/sang , Troubles ovotesticulaires du développement sexuel/génétique , Phénotype , Polymorphisme de nucléotide simple/génétique , Jeune adulte
12.
Oncotarget ; 8(61): 104123-104135, 2017 Nov 28.
Article de Anglais | MEDLINE | ID: mdl-29262626

RÉSUMÉ

INTRODUCTION: TP53 mutations are considered to be the driving factor in the initiation of esophageal adenocarcinoma (EAC). However, the impact of this gene and its encoded protein as a prognostic marker has not been definitely established yet. METHODS: In total, 204 chemoradiotherapy (CRT)-naive patients with EAC were included for p53 protein expression evaluation by immunohistochemistry (IHC) on the resection specimens, categorized as overexpression, heterogeneous or loss of expression, and correlated with disease free survival (DFS) and overall survival (OS) using multivariable Cox regression analysis. In a subset representing all three IHC subgroups mutational status of selected candidate genes (n=33) and high throughput methylation profiling (n=16) was assessed. RESULTS: Compared to heterogeneous p53 expression, loss and overexpression were both independently predictive for adverse DFS and OS. TP53 mutational status significantly correlated with the IHC categories (p=0.035). Most of the EAC with loss- or overexpression harbored TP53 mutations (18/20, representing nonsense and missense mutations respectively). In contrast, 6/13 EAC with heterogeneous expression were TP53 wild type, of which two demonstrated MDM4 or MDM2 amplification. Combined genomic hypomethylation and high frequency of intra-chromosomal breaks was found in a selection of EAC without p53 overexpression. CONCLUSION: P53 expression pattern is prognostic for DFS and OS in this historical cohort of CRT-naive EAC. P53 IHC is an informative readout for TP53 mutational status in EAC with either loss- or overexpression, but not in case of a heterogeneous p53 pattern. Different EAC pathogenesis might exist, related to p53 and other candidate gene status, DNA hypomethylation and intrachromosomal breaks.

13.
PLoS One ; 12(5): e0178169, 2017.
Article de Anglais | MEDLINE | ID: mdl-28542371

RÉSUMÉ

Adult male germline stem cells (spermatogonia) proliferate by mitosis and, after puberty, generate spermatocytes that undertake meiosis to produce haploid spermatozoa. Germ cells are under evolutionary constraint to curtail mutations and maintain genome integrity. Despite constant turnover, spermatogonia very rarely form tumors, so-called spermatocytic tumors (SpT). In line with the previous identification of FGFR3 and HRAS selfish mutations in a subset of cases, candidate gene screening of 29 SpTs identified an oncogenic NRAS mutation in two cases. To gain insights in the etiology of SpT and into properties of the male germline, we performed whole-genome sequencing of five tumors (4/5 with matched normal tissue). The acquired single nucleotide variant load was extremely low (~0.2 per Mb), with an average of 6 (2-9) non-synonymous variants per tumor, none of which is likely to be oncogenic. The observed mutational signature of SpTs is strikingly similar to that of germline de novo mutations, mostly involving C>T transitions with a significant enrichment in the ACG trinucleotide context. The tumors exhibited extensive aneuploidy (50-99 autosomes/tumor) involving whole-chromosomes, with recurrent gains of chr9 and chr20 and loss of chr7, suggesting that aneuploidy itself represents the initiating oncogenic event. We propose that SpT etiology recapitulates the unique properties of male germ cells; because of evolutionary constraints to maintain low point mutation rate, rare tumorigenic driver events are caused by a combination of gene imbalance mediated via whole-chromosome aneuploidy. Finally, we propose a general framework of male germ cell tumor pathology that accounts for their mutational landscape, timing and cellular origin.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Génome humain , Mutation germinale/génétique , Séquençage nucléotidique à haut débit/méthodes , Spermatocytes/anatomopathologie , Tumeurs du testicule/génétique , Variations de nombre de copies de segment d'ADN/génétique , Méthylation de l'ADN , Humains , Mâle , Récepteur de type 3 des facteurs de croissance fibroblastique , Maturation sexuelle , Spermatocytes/métabolisme , Tumeurs du testicule/anatomopathologie
14.
Genome Res ; 26(11): 1490-1504, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27803193

RÉSUMÉ

Testicular germ cell tumors (TGCTs) share germline ancestry but diverge phenotypically and clinically as seminoma (SE) and nonseminoma (NSE), the latter including the pluripotent embryonal carcinoma (EC) and its differentiated derivatives, teratoma (TE), yolk sac tumor (YST), and choriocarcinoma. Epigenomes from TGCTs may illuminate reprogramming in both normal development and testicular tumorigenesis. Herein we investigate pure-histological forms of 130 TGCTs for conserved and subtype-specific DNA methylation, including analysis of relatedness to pluripotent stem cell (ESC, iPSC), primordial germ cell (PGC), and differentiated somatic references. Most generally, TGCTs conserve PGC-lineage erasure of maternal and paternal genomic imprints and DPPA3 (also known as STELLA); however, like ESCs, TGCTs show focal recurrent imprinted domain hypermethylation. In this setting of shared physiologic erasure, NSEs harbor a malignancy-associated hypermethylation core, akin to that of a diverse cancer compendium. Beyond these concordances, we found subtype epigenetic homology with pluripotent versus differentiated states. ECs demonstrate a striking convergence of both CpG and CpH (non-CpG) methylation with pluripotent states; the pluripotential methyl-CpH signature crosses species boundaries and is distinct from neuronal methyl-CpH. EC differentiation to TE and YST entails reprogramming toward the somatic state, with loss of methyl-CpH but de novo methylation of pluripotency loci such as NANOG Extreme methyl-depletion among SE reflects the PGC methylation nadir. Adjacent to TGCTs, benign testis methylation profiles are determined by spermatogenetic proficiency measured by Johnsen score. In sum, TGCTs share collective entrapment in a PGC-like state of genomic-imprint and DPPA3 erasure, recurrent hypermethylation of cancer-associated targets, and subtype-dependent pluripotent, germline, or somatic methylation.


Sujet(s)
Reprogrammation cellulaire , Méthylation de l'ADN , Empreinte génomique , Tumeurs embryonnaires et germinales/génétique , Cellules souches pluripotentes/métabolisme , Protéines/génétique , Tumeurs du testicule/génétique , Lignage cellulaire , Protéines chromosomiques nonhistones , Ilots CpG , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Protéine homéotique Nanog/génétique , Protéine homéotique Nanog/métabolisme , Cellules souches pluripotentes/cytologie , Protéines/métabolisme
15.
PLoS One ; 10(8): e0136845, 2015.
Article de Anglais | MEDLINE | ID: mdl-26317614

RÉSUMÉ

BACKGROUND: Most breast cancers depend on estrogenic growth stimulation. Functional genetic screenings in in vitro cell models have identified genes, which override growth suppression induced by anti-estrogenic drugs like tamoxifen. Using that approach, we have previously identified Breast Cancer Anti-Estrogen Resistance 4 (BCAR4) as a mediator of cell proliferation and tamoxifen-resistance. Here, we show high level of expression and function of BCAR4 in human breast cancer. METHODS: BCAR4 mRNA expression was evaluated by (q)RT-PCR in a panel of human normal tissues, primary breast cancers and cell lines. A new antibody raised against C78-I97 of the putative BCAR4 protein and used for western blot and immunoprecipitation assays. Furthermore, siRNA-mediated gene silencing was implemented to study the function of BCAR4 and its downstream targets ERBB2/3. RESULTS: Except for placenta, all human normal tissues tested were BCAR4-negative. In primary breast cancers, BCAR4 expression was comparatively rare (10%), but associated with enhanced proliferation. Relative high BCAR4 mRNA expression was identified in IPH-926, a cell line derived from an endocrine-resistant lobular breast cancer. Moderate BCAR4 expression was evident in MDA-MB-134 and MDA-MB-453 breast cancer cells. BCAR4 protein was detected in breast cancer cells with ectopic (ZR-75-1-BCAR4) and endogenous (IPH-926, MDA-MB-453) BCAR4 mRNA expression. Knockdown of BCAR4 inhibited cell proliferation. A similar effect was observed upon knockdown of ERBB2/3 and exposure to lapatinib, implying that BCAR4 acts in an ERBB2/3-dependent manner. CONCLUSION: BCAR4 encodes a functional protein, which drives proliferation of endocrine-resistant breast cancer cells. Lapatinib, a clinically approved EGFR/ERBB2 inhibitor, counteracts BCAR4-driven tumor cell growth, a clinical relevant observation.


Sujet(s)
Tumeurs du sein/génétique , Carcinome lobulaire/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Adulte , Sujet âgé , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Carcinome lobulaire/métabolisme , Carcinome lobulaire/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Lapatinib , Adulte d'âge moyen , Inhibiteurs de protéines kinases/pharmacologie , Quinazolines/pharmacologie , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Récepteur ErbB-3/génétique , Récepteur ErbB-3/métabolisme
16.
PLoS One ; 10(4): e0122146, 2015.
Article de Anglais | MEDLINE | ID: mdl-25859847

RÉSUMÉ

The cell of origin of the five subtypes (I-V) of germ cell tumors (GCTs) are assumed to be germ cells from different maturation stages. This is (potentially) reflected in their methylation status as fetal maturing primordial germ cells are globally demethylated during migration from the yolk sac to the gonad. Imprinted regions are erased in the gonad and later become uniparentally imprinted according to fetal sex. Here, 91 GCTs (type I-IV) and four cell lines were profiled (Illumina's HumanMethylation450BeadChip). Data was pre-processed controlling for cross hybridization, SNPs, detection rate, probe-type bias and batch effects. The annotation was extended, covering snRNAs/microRNAs, repeat elements and imprinted regions. A Hidden Markov Model-based genome segmentation was devised to identify differentially methylated genomic regions. Methylation profiles allowed for separation of clusters of non-seminomas (type II), seminomas/dysgerminomas (type II), spermatocytic seminomas (type III) and teratomas/dermoid cysts (type I/IV). The seminomas, dysgerminomas and spermatocytic seminomas were globally hypomethylated, in line with previous reports and their demethylated precursor. Differential methylation and imprinting status between subtypes reflected their presumed cell of origin. Ovarian type I teratomas and dermoid cysts showed (partial) sex specific uniparental maternal imprinting. The spermatocytic seminomas showed uniparental paternal imprinting while testicular teratomas exhibited partial imprinting erasure. Somatic imprinting in type II GCTs might indicate a cell of origin after global demethylation but before imprinting erasure. This is earlier than previously described, but agrees with the totipotent/embryonic stem cell like potential of type II GCTs and their rare extra-gonadal localization. The results support the common origin of the type I teratomas and show strong similarity between ovarian type I teratomas and dermoid cysts. In conclusion, we identified specific and global methylation differences between GCT subtypes, providing insight into their developmental timing and underlying developmental biology. Data and extended annotation are deposited at GEO (GSE58538 and GPL18809).


Sujet(s)
Méthylation de l'ADN , Tumeurs embryonnaires et germinales/génétique , Tumeurs embryonnaires et germinales/anatomopathologie , Lignée cellulaire tumorale , Femelle , Génome humain , Étude d'association pangénomique , Humains , Mâle , Séminome/génétique , Séminome/anatomopathologie , Tératome/génétique , Tératome/anatomopathologie
17.
PLoS One ; 9(6): e98330, 2014.
Article de Anglais | MEDLINE | ID: mdl-24887064

RÉSUMÉ

BACKGROUND: Originating from Primordial Germ Cells/gonocytes and developing via a precursor lesion called Carcinoma In Situ (CIS), Germ Cell Cancers (GCC) are the most common cancer in young men, subdivided in seminoma (SE) and non-seminoma (NS). During physiological germ cell formation/maturation, epigenetic processes guard homeostasis by regulating the accessibility of the DNA to facilitate transcription. Epigenetic deregulation through genetic and environmental parameters (i.e. genvironment) could disrupt embryonic germ cell development, resulting in delayed or blocked maturation. This potentially facilitates the formation of CIS and progression to invasive GCC. Therefore, determining the epigenetic and functional genomic landscape in GCC cell lines could provide insight into the pathophysiology and etiology of GCC and provide guidance for targeted functional experiments. RESULTS: This study aims at identifying epigenetic footprints in SE and EC cell lines in genome-wide profiles by studying the interaction between gene expression, DNA CpG methylation and histone modifications, and their function in the pathophysiology and etiology of GCC. Two well characterized GCC-derived cell lines were compared, one representative for SE (TCam-2) and the other for EC (NCCIT). Data were acquired using the Illumina HumanHT-12-v4 (gene expression) and HumanMethylation450 BeadChip (methylation) microarrays as well as ChIP-sequencing (activating histone modifications (H3K4me3, H3K27ac)). Results indicate known germ cell markers not only to be differentiating between SE and NS at the expression level, but also in the epigenetic landscape. CONCLUSION: The overall similarity between TCam-2/NCCIT support an erased embryonic germ cell arrested in early gonadal development as common cell of origin although the exact developmental stage from which the tumor cells are derived might differ. Indeed, subtle difference in the (integrated) epigenetic and expression profiles indicate TCam-2 to exhibit a more germ cell-like profile, whereas NCCIT shows a more pluripotent phenotype. The results provide insight into the functional genome in GCC cell lines.


Sujet(s)
Carcinome embryonnaire/génétique , Épigenèse génétique , Analyse de profil d'expression de gènes , Tumeurs embryonnaires et germinales/génétique , Séminome/génétique , Carcinome embryonnaire/anatomopathologie , Lignée cellulaire tumorale , Méthylation de l'ADN , Humains , Tumeurs embryonnaires et germinales/anatomopathologie , Séminome/anatomopathologie
18.
BMC Bioinformatics ; 15: 141, 2014 May 15.
Article de Anglais | MEDLINE | ID: mdl-24884391

RÉSUMÉ

BACKGROUND: Array based methylation profiling is a cost-effective solution to study the association between genome methylation and human disease & development. Available tools to analyze the Illumina Infinium HumanMethylation450 BeadChip focus on comparing methylation levels per locus. Other tools combine multiple probes into a range, identifying differential methylated regions (DMRs). These tools all require groups of samples to compare. However, comparison of unique, individual samples is essential in situations where larger sample sizes are not possible. RESULTS: DMRforPairs was designed to compare regional methylation status between unique samples. It identifies probe dense genomic regions and quantifies/tests their (difference in) methylation level between the samples. As a proof of concept, DMRforPairs is applied to public data from four human cell lines: two lymphoblastoid cell lines from healthy individuals and the cancer cell lines A431 and MCF7 (including 2 technical replicates each). DMRforPairs identified an increasing number of DMRs related to the sample phenotype when biological similarity of the samples decreased. DMRs identified by DMRforPairs were related to the biological origin of the cell lines. CONCLUSION: To our knowledge, DMRforPairs is the first tool to identify and visualize relevant and significant differentially methylated regions between unique samples.


Sujet(s)
Méthylation de l'ADN , Séquençage par oligonucléotides en batterie/méthodes , Logiciel , Lignée cellulaire , Lignée cellulaire tumorale , Femelle , Génomique , Humains , Cellules MCF-7 , Mâle
19.
PLoS One ; 9(1): e83585, 2014.
Article de Anglais | MEDLINE | ID: mdl-24404135

RÉSUMÉ

The transcription factor SOX2, associated with amongst others OCT3/4, is essential for maintenance of pluripotency and self-renewal of embryonic stem cells. SOX2 is highly expressed in embryonal carcinoma (EC), the stem cell component of malignant nonseminomatous germ cell tumors, referred to as germ cell cancer (GCC). In fact, OCT3/4 together with SOX2 is an informative diagnostic tool for EC in a clinical setting. Several studies support the hypothesis that SOX2 is a relevant oncogenic factor in various cancers and recently, SOX2 has been suggested as a putative therapeutic target for early stage EC. We demonstrate the presence of genomic amplification of SOX2 in an EC cell line, NCCIT, using array comparative genome hybridization and fluorescence in situ hybridization. Down-regulation of SOX2 by targeted siRNA provokes NCCIT cells towards apoptosis, while inhibition of OCT3/4 expression induced differentiation, with retained SOX2 levels. Mice pluripotent xenografts from NCCIT (N-NCCIT and N2-NCCIT) show a consistent SOX2 expression, in spite of loss of the expression of OCT3/4, and differentiation, with retained presence of genomic amplification. No SOX2 amplification has been identified in primary pure and mixed EC in vivo patient samples so far. The data presented in this study are based on a single EC cell line with a SOX2 amplification, with NT2 as control EC cell line, showing no profound induction of apoptosis upon SOX2 downregulation. The findings are of relevance to identify mechanisms involved in the pathogenesis of EC tumors, and support the model of SOX2-oncogene dependency of EC, which however, does not exclude induction of differentiation. This finding is likely related to the presence of wild type p53 in GCC, resulting in expression of downstream target genes, amongst others miR-34a, miR-145 and SOX2, associated to the unique sensitivity of GCC to DNA damaging agents.


Sujet(s)
Carcinome embryonnaire/génétique , Carcinome embryonnaire/métabolisme , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription SOX-B1/métabolisme , Animaux , Apoptose/génétique , Marqueurs biologiques tumoraux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Amplification de gène , Expression des gènes , Techniques de knock-down de gènes , Extinction de l'expression des gènes , Hétérogreffes , Humains , Souris , Modèles biologiques , Facteur de transcription Oct-3/génétique , Facteur de transcription Oct-3/métabolisme , Interférence par ARN
20.
Mol Oncol ; 7(6): 1083-92, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24012110

RÉSUMÉ

Germ cell cancers (GCC) are the most frequent malignancy in young Caucasian males. GCC can consist of seminomas (SE) and non-seminomas (malignant NS: embryonal carcinoma (EC), yolk sac tumor (YS), choriocarcinoma (CH) and teratoma (TE)). Current serum-markers used for diagnosis and follow-up (AFP, hCG) are predominantly related to YS and CH and marker positivity can vary during disease. Therefore, stable markers consistently identifying more GCC components, specifically the stem cell components SE and EC, are of interest. Expression of the embryonic stem cell miR-371-3 and miR-302/367 clusters in SE/EC/YS suggest possible application of these micro-RNAs as GCC tumor-markers. The TSmiR protocol constitutes a complete, quality-controlled pipeline for the detection of miRs in serum, based on magnetic bead-based purification and qPCR quantification. As a proof of principle, TSmiR was applied to five independent serum sample series including 80 GCCs, 47 controls, 11 matched pre/post orchidectomy samples and 12 no-GCC testicular masses. GCC serum samples showed a consistent, significant (p < 0.0064) increase of miR-371/372/373/367 levels. Analogous, serum levels returned to baseline after orchidectomy (stage-I disease). Moreover, there was a trend toward higher miR levels in patients with metastasis. These results imply suitability for diagnosis and follow-up. TSmiR showed an overall sensitivity of 98%, clearly outperforming the traditional serum markers AFP/hCG (36%/57%, sensitivity(AFP) = 3%/45%; sensitivity(hCG) = 62%/66%, SE/NS). TSmiR misclassified one tumor as a control. Serum AFP/hCG and TSmiR combined identified all T samples correctly. In conclusion, TSmiR constitutes a highly sensitive and reproducible serum test for GCC patients, suitable to be prospectively tested for diagnostic and follow-up purposes.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , microARN/métabolisme , Tumeurs embryonnaires et germinales/diagnostic , Tumeurs embryonnaires et germinales/métabolisme , ARN tumoral/métabolisme , Tumeurs du testicule/diagnostic , Tumeurs du testicule/métabolisme , Adolescent , Adulte , Marqueurs biologiques tumoraux/génétique , Enfant , Enfant d'âge préscolaire , Humains , Mâle , microARN/génétique , Tumeurs embryonnaires et germinales/génétique , Tumeurs embryonnaires et germinales/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , ARN tumoral/génétique , Tumeurs du testicule/génétique , Tumeurs du testicule/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...