Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
EBioMedicine ; 62: 103131, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33254023

ABSTRACT

BACKGROUND: Undifferentiated pleomorphic sarcoma (UPS) is the most frequent, aggressive and less-characterized sarcoma subtype. This study aims to assess UPS molecular characteristics and identify specific therapeutic targets. METHODS: High-throughput technologies encompassing immunohistochemistry, RNA-sequencing, whole exome-sequencing, mass spectrometry, as well as radiomics were used to characterize three independent cohorts of 110, 25 and 41 UPS selected after histological review performed by an expert pathologist. Correlations were made with clinical outcome. Cell lines and xenografts were derived from human samples for functional experiments. FINDINGS: CD8 positive cell density was independently associated with metastatic behavior and prognosis. RNA-sequencing identified two main groups: the group A, enriched in genes involved in development and stemness, including FGFR2, and the group B, strongly enriched in genes involved in immunity. Immune infiltrate patterns on tumor samples were highly predictive of gene expression classification, leading to call the group B 'immune-high' and the group A 'immune-low'. This molecular classification and its prognostic impact were confirmed on an independent cohort of UPS from TCGA. Copy numbers alterations were significantly more frequent in immune-low UPS. Proteomic analysis identified two main proteomic groups that highly correlated with the two main transcriptomic groups. A set of nine radiomic features from conventional MRI sequences provided the basis for a radiomics signature that could select immune-high UPS on their pre-therapeutic imaging. Finally, in vitro and in vivo anti-tumor activity of FGFR inhibitor JNJ-42756493 was selectively shown in cell lines and patient-derived xenograft models derived from immune-low UPS. INTERPRETATION: Two main disease entities of UPS, with distinct immune phenotypes, prognosis, molecular features and MRI textures, as well as differential sensitivity to specific anticancer agents were identified. Immune-high UPS may be the best candidates for immune checkpoint inhibitors, whereas this study provides rational for assessing FGFR inhibition in immune-low UPS. FUNDING: This work was partly founded by a grant from La Ligue.


Subject(s)
Biomarkers, Tumor , Gene Expression Profiling , Sarcoma/etiology , Sarcoma/metabolism , Transcriptome , Animals , Cell Cycle/genetics , Computational Biology/methods , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Prognosis , Proteomics , Sarcoma/diagnosis , Sarcoma/therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Exome Sequencing
2.
Cancers (Basel) ; 12(10)2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33092134

ABSTRACT

We aimed to evaluate the therapeutic potential of the pan-FGFR inhibitor erdafitinib to treat dedifferentiated liposarcoma (DDLPS). FGFR expression and their prognostic value were assessed in a series of 694 samples of well-differentiated/dedifferentiated liposarcoma (WDLPS/DDLPS). The effect of erdafitinib-alone or in combination with other antagonists-on tumorigenicity was evaluated in vitro and in vivo. We detected overexpression of FGFR1 and/or FGFR4 in a subset of WDLPS and DDLPS and demonstrated correlation of this expression with poor prognosis. Erdafitinib treatment reduced cell viability, inducing apoptosis and strong inhibition of the ERK1/2 pathway. Combining erdafitinib with the MDM2 antagonist RG7388 exerted a synergistic effect on viability, apoptosis, and clonogenicity in one WDLPS and two DDLPS cell lines. Efficacy of this combination was confirmed in vivo on a DDLPS xenograft. Importantly, we report the efficacy of erdafitinib in one patient with refractory DDLPS showing disease stabilization for 12 weeks. We provide evidence that the FGFR pathway has therapeutic potential for a subset of DDLPS and that an FGFR1/FGFR4 expression might constitute a powerful biomarker to select patients for FGFR inhibitor clinical trials. In addition, we show that combining erdafitinib with RG7388 is a promising strategy for patients with DDLPS that deserves further investigation in the clinical setting.

3.
Cancer Res ; 80(16): 3236-3250, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32366479

ABSTRACT

There is great interest in understanding how the cancer stem cell population may be maintained in solid tumors. Here, we show that tumor cells exhibiting stem-like properties and expression of pluripotency markers NANOG and OCT4 can arise from original differentiated tumor cells freshly isolated from human glioblastomas (GBM) and that have never known any serum culture conditions. Induction of EGR1 by EGFR/ERK signaling promoted cell conversion from a less aggressive, more differentiated cellular state to a self-renewing and strongly tumorigenic state, expressing NANOG and OCT4. Expression of these pluripotency markers occurred before the cells re-entered the cell cycle, demonstrating their capacity to change and dedifferentiate without any cell divisions. In differentiated GBM cells, ERK-mediated repression of miR-199a-3p induced EGR1 protein expression and triggered dedifferentiation. Overall, this signaling pathway constitutes an ERK-mediated "toggle switch" that promotes pluripotency marker expression and stem-like features in GBM cells. SIGNIFICANCE: This study defines an ERK-mediated molecular mechanism of dedifferentiation of GBM cells into a stem-like state, expressing markers of pluripotency.See related commentary by Koncar and Agnihotri, p. 3195.


Subject(s)
Glioblastoma , MicroRNAs , Cell Dedifferentiation , Cell Differentiation , Early Growth Response Protein 1 , Glioblastoma/genetics , Humans , MicroRNAs/genetics , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells
4.
Genes Chromosomes Cancer ; 58(9): 673-679, 2019 09.
Article in English | MEDLINE | ID: mdl-30887579

ABSTRACT

Adipocytic tumors are rare in children and are mostly benign. Less than 25 cases of pediatric well-differentiated liposarcoma (WDLPS), atypical lipomatous tumors (ALT), and dedifferentiated liposarcoma (DDLPS) have been reported. Among them, only three cases were genetically analyzed. We describe the genetic features of a rapidly growing adipose tumor that occurred in the thigh of a 7-year-old girl. Histologically, it was composed of mature adipocytic cells with a few atypia. Molecular analysis showed high-level amplification of the 12q13-21 region including MDM2 among 64 amplified genes. MDM2 amplification is a diagnostic hallmark of ALT/WDLPS/DDLPS. In adult cases, it is typically located in ring or giant marker chromosomes. In the present case, extra-copies of MDM2 were located on double minute chromosomes (dmin). This raised the hypothesis of dmin being precursors of adult's rings and giant markers and may provide indications for a better understanding of the mechanisms of adipose tumor oncogenesis.


Subject(s)
Gene Amplification , Liposarcoma/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Soft Tissue Neoplasms/genetics , Adipocytes/metabolism , Adipocytes/pathology , Child , Chromosomes, Human, Pair 12/genetics , Female , Humans , Liposarcoma/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Soft Tissue Neoplasms/pathology
5.
Genetics ; 208(3): 951-961, 2018 03.
Article in English | MEDLINE | ID: mdl-29279323

ABSTRACT

Genome amplification in the form of rings or giant rod-shaped marker chromosomes (RGMs) is a common genetic alteration in soft tissue tumors. The mitotic stability of these structures is often rescued by perfectly functioning analphoid neocentromeres, which therefore significantly contribute to cancer progression. Here, we disentangled the genomic architecture of many neocentromeres stabilizing marker chromosomes in well-differentiated liposarcoma and lung sarcomatoid carcinoma samples. In cells carrying heavily rearranged RGMs, these structures were assembled as patchworks of multiple short amplified sequences, disclosing an extremely high level of complexity and definitely ruling out the existence of regions prone to neocentromere seeding. Moreover, by studying two well-differentiated liposarcoma samples derived from the onset and the recurrence of the same tumor, we documented an expansion of the neocentromeric domain that occurred during tumor progression, which reflects a strong selective pressure acting toward the improvement of the neocentromeric functionality in cancer. In lung sarcomatoid carcinoma cells we documented, extensive "centromere sliding" phenomena giving rise to multiple, closely mapping neocentromeric epialleles on separate coexisting markers occur, likely due to the instability of neocentromeres arising in cancer cells. Finally, by investigating the transcriptional activity of neocentromeres, we came across a burst of chimeric transcripts, both by extremely complex genomic rearrangements, and cis/trans-splicing events. Post-transcriptional editing events have been reported to expand and variegate the genetic repertoire of higher eukaryotes, so they might have a determining role in cancer. The increased incidence of fusion transcripts, might act as a driving force for the genomic amplification process, together with the increased transcription of oncogenes.


Subject(s)
Chromosomes, Human , Genetic Markers , Genomics , Neoplasms/genetics , Transcriptome , Cell Line, Tumor , Centromere , Genomics/methods , Humans , In Situ Hybridization, Fluorescence , Polymorphism, Single Nucleotide , Transcription, Genetic , Whole Genome Sequencing
6.
Mol Cell Biol ; 36(7): 1180-93, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26830228

ABSTRACT

White adipose tissue (WAT) is essential for maintaining metabolic function, especially during obesity. The intronic microRNAs miR-33a and miR-33b, located within the genes encoding sterol regulatory element-binding protein 2 (SREBP-2) and SREBP-1, respectively, are transcribed in concert with their host genes and function alongside them to regulate cholesterol, fatty acid, and glucose metabolism. SREBP-1 is highly expressed in mature WAT and plays a critical role in promoting in vitro adipocyte differentiation. It is unknown whether miR-33b is induced during or involved in adipogenesis. This is in part due to loss of miR-33b in rodents, precluding in vivo assessment of the impact of miR-33b using standard mouse models. This work demonstrates that miR-33b is highly induced upon differentiation of human preadipocytes, along with SREBP-1. We further report that miR-33b is an important regulator of adipogenesis, as inhibition of miR-33b enhanced lipid droplet accumulation. Conversely, overexpression of miR-33b impaired preadipocyte proliferation and reduced lipid droplet formation and the induction of peroxisome proliferator-activated receptor γ (PPARγ) target genes during differentiation. These effects may be mediated by targeting of HMGA2, cyclin-dependent kinase 6 (CDK6), and other predicted miR-33b targets. Together, these findings demonstrate a novel role of miR-33b in the regulation of adipocyte differentiation, with important implications for the development of obesity and metabolic disease.


Subject(s)
Adipogenesis/genetics , Adipose Tissue, White/cytology , MicroRNAs/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Genetic Loci , Humans , MicroRNAs/biosynthesis , MicroRNAs/physiology , Stem Cells/cytology
7.
Mod Pathol ; 28(11): 1404-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26336885

ABSTRACT

HMGA2, CDK4, and JUN genes have been described as frequently coamplified with MDM2 in atypical lipomatous tumor, well-differentiated liposarcoma, and dedifferentiated liposarcoma. We studied the frequency of amplification of these genes in a series of 48 dedifferentiated liposarcomas and 68 atypical lipomatous tumors/well-differentiated liposarcomas. We correlated their amplification status with clinicopathological features and outcomes. Histologically, both CDK4 (P=0.007) and JUN (P=0.005) amplifications were associated with dedifferentiated liposarcoma, whereas amplification of the proximal parts of HMGA2 (5'-untranslated region (UTR) and exons 1-3) was associated with atypical lipomatous tumor/well-differentiated liposarcoma (P=0.01). CDK4 amplification was associated with axial tumors. Amplification of 5'-UTR and exons 1-3 of HMGA2 was associated with primary status and grade 1. Shorter overall survival was correlated with: age >64 years (P=0.03), chemotherapy used in first intent (P<0.001), no surgery (P=0.003), grade 3 (P<0.001), distant metastasis (P<0.001), node involvement (P=0.006), and CDK4 amplification (P=0.07). In multivariate analysis, distant metastasis (HR=8.8) and grade 3 (HR=18.2) were associated with shorter overall survival. A shorter recurrence-free survival was associated with dedifferentiated liposarcoma (P<0.001), grade 3 (P<0.001), node involvement (P<0.001), distant metastasis (P=0.02), recurrent status (P=0.009), axial location (P=0.001), and with molecular features such as CDK4 (P=0.05) and JUN amplification (P=0.07). Amplification of 5'-UTR and exons 1-3 (P=0.08) and 3'-UTR (P=0.01) of HMGA2 were associated with longer recurrence-free survival. Distant metastasis was associated with shorter recurrence-free survival (HR=5.8) in multivariate analysis. Dedifferentiated liposarcoma type was associated with axial location, grade 3 and recurrent status. In conclusion, we showed that the amplification of HMGA2 was associated with the atypical lipomatous tumor/well-differentiated liposarcoma histological type and a good prognosis, whereas CDK4 and JUN amplifications were associated with dedifferentiated liposarcoma histology and a bad prognosis. In addition, we also provided the first description of the molecular evolution of a well-differentiated liposarcoma into four successive dedifferentiated liposarcoma relapses, which was consistent with our general observations.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Gene Amplification , Genes, jun/genetics , HMGA2 Protein/genetics , Liposarcoma/genetics , Liposarcoma/pathology , Soft Tissue Neoplasms/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Comparative Genomic Hybridization , Disease-Free Survival , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Liposarcoma/mortality , Male , Middle Aged , Oncogene Protein p65(gag-jun)/genetics , Prognosis , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/pathology
8.
Cancer Genet ; 208(10): 469-81, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26319758

ABSTRACT

Both epidemiologic and cytogenetic data on pediatric adipose tissue tumors are scarce. Pediatric adipose tumors are mainly represented by lipomas, though only 28 cytogenetic descriptions of pediatric lipoma have been reported to date. Similar to adult cases, most of these pediatric lipomas harbored rearrangements of the chromosomal regions 12q14-q15 and 6p21, involving the HMGA2 and HMGA1 genes. Further cytogenetic studies of pediatric lipoma would be useful to determinate whether some partner genes of HMGA2, such as NFIB, may have a specific role in the early onset of these tumors. Cytogenetically, the best documented pediatric adipose tumor is lipoblastoma, which is the second most frequent adipose tumor in children. Chromosomal alterations in lipoblastoma, observed in 61% of cases studied by conventional cytogenetics, typically involve the 8q11-q12 region. The target gene of this rearrangement is PLAG1. Anomalies of PLAG1 have been observed in 70% of cases of pediatric adipose tumors studied by molecular cytogenetics methods, such as fluorescence in situ hybridization (FISH) or comparative genomic hybridization on array (array-CGH). The rare described cases of malignant pediatric adipose tumors in children are mostly myxoid liposarcomas. In the 27 cases explored at the genetic level, all pediatric myxoid liposarcomas showed the classical rearrangement of the DDIT3 gene at 12q13. In conclusion, the epidemiology and the prevalence of histological types of adipose tissue tumors differ in the pediatric population compared with adults, whereas chromosomal and genic rearrangements are similar to those of adult cases in each histological type.


Subject(s)
Lipoblastoma/epidemiology , Lipoblastoma/genetics , Liposarcoma/epidemiology , Liposarcoma/genetics , Child , Child, Preschool , Chromosome Aberrations , Cytogenetic Analysis , Humans , Infant , Lipoblastoma/pathology , Liposarcoma/pathology , Prevalence , Recombinant Fusion Proteins/genetics
9.
Carcinogenesis ; 36(1): 32-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25344834

ABSTRACT

Syndecan-1 (SDC1/CD138) is one of the main cell surface proteoglycans and is involved in crucial biological processes. Only a few studies have analyzed the role of SDC1 in mesenchymal tumor pathogenesis. In particular, its involvement in adipose tissue tumors has never been investigated. Dedifferentiated liposarcoma, one of the most frequent types of malignant adipose tumors, has a high potential of recurrence and metastastic evolution. Classical chemotherapy is inefficient in metastatic dedifferentiated liposarcoma and novel biological markers are needed for improving its treatment. In this study, we have analyzed the expression of SDC1 in well-differentiated/dedifferentiated liposarcomas and showed that SDC1 is highly overexpressed in dedifferentiated liposarcoma compared with normal adipose tissue and lipomas. Silencing of SDC1 in liposarcoma cells impaired cell viability and proliferation. Using the human multipotent adipose-derived stem cell model of human adipogenesis, we showed that SDC1 promotes proliferation of undifferentiated adipocyte progenitors and inhibits their adipogenic differentiation. Altogether, our results support the hypothesis that SDC1 might be involved in liposarcomagenesis. It might play a prominent role in the dedifferentiation process occurring when well-differentiated liposarcoma progress to dedifferentiated liposarcoma. Targeting SDC1 in these tumors might provide a novel therapeutic strategy.


Subject(s)
Adipogenesis , Adipose Tissue/pathology , Cell Differentiation , Cell Transformation, Neoplastic/pathology , Liposarcoma/pathology , Syndecan-1/metabolism , Adipose Tissue/metabolism , Blotting, Western , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Flow Cytometry , Humans , Immunoenzyme Techniques , Liposarcoma/genetics , Liposarcoma/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology , Syndecan-1/antagonists & inhibitors , Syndecan-1/genetics
10.
Genes Chromosomes Cancer ; 52(6): 580-90, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23508853

ABSTRACT

Most lipomas are characterized by translocations involving the HMGA2 gene in 12q14.3. These rearrangements lead to the fusion of HMGA2 with an ectopic sequence from the translocation chromosome partner. Only five fusion partners of HMGA2 have been identified in lipomas so far. The identification of novel fusion partners of HMGA2 is important not only for diagnosis in soft tissue tumors but also because these genes might have an oncogenic role in other tumors. We observed that t(1;12)(p32;q14) was the second most frequent translocation in our series of lipomas after t(3;12)(q28;q14.3). We detected overexpression of HMGA2 mRNA and protein in all t(1;12)(p32;q14) lipomas. We used a fluorescence in situ hybridization-based positional cloning strategy to characterize the 1p32 breakpoint. In 11 cases, we identified PPAP2B, a member of the lipid phosphate phosphatases family as the 1p32 target gene. Reverse transcription-polymerase chain reaction analysis followed by nucleotide sequencing of the fusion transcript indicated that HMGA2 3' untranslated region (3'UTR) fused with exon 6 of PPAP2B in one case. In other t(1;12) cases, the breakpoint was extragenic, located in the 3'region flanking PPAP2B 3'UTR. Moreover, in one case showing a t(1;6)(p32;p21) we observed a rearrangement of PPAP2B and HMGA1, which suggests that HMGA1 might also be a fusion partner for PPAP2B. Our results also revealed that adipocytic differentiation of human mesenchymal stem cells derived from adipose tissue was associated with a significant decrease in PPAP2B mRNA expression suggesting that PPAP2B might play a role in adipogenesis.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 1/genetics , HMGA2 Protein/genetics , Lipoma/genetics , Phosphatidate Phosphatase/genetics , Translocation, Genetic/genetics , 3' Untranslated Regions/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adolescent , Adult , Blotting, Western , Female , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Karyotyping , Male , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Young Adult
11.
Virchows Arch ; 461(1): 67-78, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22678079

ABSTRACT

While surgery is the usual treatment for localized well-differentiated and dedifferentiated liposarcomas (WDLPS/DDLPS), the therapeutic options for patients with advanced disease are limited. The classical antimitotic treatments are most often inefficient. The establishment of genetically characterized cell lines is therefore crucial for providing in vitro models for novel targeted therapies. We have used spectral karyotyping, fluorescence in situ hybridization with whole chromosome painting and locus-specific probes, and array-comparative genomic hybridization to identify the chromosomal and molecular alterations of a novel cell line established from a recurring sclerosing WDLPS. The karyotype was hypertriploid and showed multiple structural anomalies. All cells retained the presence of a giant marker chromosome that had been previously identified in the primary cell cultures. This giant chromosome contained high-level amplification of chromosomal regions 12q13-21 and lacked the alpha-satellite centromeric sequences associated with WDLPS/DDLPS. The 12q amplicon was large, containing 370 amplified genes. The DNA copy number ranged from 3 to 57. The highest levels of amplification were observed at 12q14.3 for GNS, WIF1, and HMGA2. We analyzed the mRNA expression status by real-time reverse transcription polymerase chain reaction for six genes from this amplicon: MDM2, HMGA2, CDK4, TSPAN31, WIF1, and YEATS4. mRNA overexpression was correlated with genomic amplification. A second amplicon originating from 10p11-14 was also present in the giant marker chromosome. The 10p amplicon contained 62 genes, including oncogenes such as MLLT10, previously described in chimeric fusion with MLL in leukemias, NEBL, and BMI1.


Subject(s)
Cell Line, Tumor , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 12/genetics , Liposarcoma/genetics , Primary Cell Culture , Adult , Cell Differentiation , Comparative Genomic Hybridization , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Liposarcoma/pathology , Male , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
13.
J Pathol ; 224(4): 575-88, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21598253

ABSTRACT

Aberrations of specialized metabolic pathways might be implicated in the development of neoplasias. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors with important functions in metabolism. PPARß/δ and PPARγ act in the proliferation and differentiation of adipose tissue progenitor cells. Thus, a potential use of PPARγ agonists for the treatment of liposarcoma had been suggested, but clinical trials failed to detect beneficial effects. We show here that PPARδ is highly expressed in liposarcoma compared to lipoma and correlates with proliferation. Stimulation of liposarcoma cell lines with a specific PPARδ agonist increases proliferation, which is abolished by a PPARδ-siRNA or a specific PPARδ antagonist. Expression of the adipose tissue secretory factor leptin is lower in liposarcoma compared to lipoma and leptin reduces proliferation of liposarcoma cell lines. PPARδ activation stimulates cell migration whereas leptin diminishes it. We demonstrate that PPARδ directly represses leptin as: (a) leptin becomes down-regulated upon PPARδ activation; (b) PPARδ represses leptin promoter activity in different sarcoma cell lines; (c) deletion of a PPAR/RxR binding element in the leptin promoter abolishes repression by PPARδ; and (d) in chromatin immunoprecipitation we confirm in vivo binding of PPARδ to the leptin promoter. Our data suggest inhibition of PPARδ as a potential novel strategy to reduce liposarcoma cell proliferation.


Subject(s)
Liposarcoma/metabolism , PPAR delta/metabolism , PPAR-beta/metabolism , Adult , Aged , Aged, 80 and over , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation , Female , Humans , Leptin/antagonists & inhibitors , Leptin/biosynthesis , Leptin/pharmacology , Lipoma/metabolism , Lipoma/pathology , Liposarcoma/pathology , Male , Middle Aged , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , PPAR delta/pharmacology , PPAR delta/physiology , PPAR-beta/physiology , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Cells, Cultured
14.
Genes Chromosomes Cancer ; 50(6): 442-55, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21412931

ABSTRACT

The aim of our study was first to assess the role of HMGA2 expression in the pathogenesis of adipocytic tumors (AT) and, second, to seek a potential correlation between overexpression of HMGA2 and let-7 expression inhibition by analyzing a series of 56 benign and malignant AT with molecular cytogenetic data. We measured the levels of expression of HMGA2 mRNA and of eight members of the let-7 microRNA family using quantitative RT-PCR and expression of HMGA2 protein using immunohistochemistry. HMGA2 was highly overexpressed in 100% of well-differentiated/dedifferentiated liposarcomas (WDLPS/DDLPS), all with HMGA2 amplification, and 100% of lipomas with HMGA2 rearrangement. Overexpression of HMGA2 mRNA was detected in 76% of lipomas without HMGA2 rearrangement. HMGA2 protein expression was detected in 100% of lipomas with HMGA2 rearrangement and 48% of lipomas without HMGA2 rearrangement. We detected decreased expression levels of some let-7 members in a significant proportion of AT. Notably, let-7b and let-7g were inhibited in 61% of WDLPS/DDLPS. In lipomas, each type of let-7 was inhibited in approximately one-third of the cases. Although overexpression of both HMGA2 mRNA and protein in a majority of ordinary lipomas without HMGA2 structural rearrangement may have suggested a potential role for let-7 microRNAs, we did not observe a significant link with let-7 inhibition in such cases. Our results indicate that inhibition of let-7 microRNA expression may participate in the deregulation of HMGA2 in AT but that this inhibition is neither a prominent stimulator for HMGA2 overexpression nor a surrogate to genomic HMGA2 rearrangements.


Subject(s)
Adipocytes/metabolism , HMGA2 Protein/genetics , Lipoma/genetics , Liposarcoma/genetics , MicroRNAs/genetics , Adipocytes/pathology , Adult , Aged , Aged, 80 and over , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Lipoma/pathology , Liposarcoma/pathology , Male , Middle Aged
15.
Clin Cancer Res ; 15(18): 5696-703, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19737942

ABSTRACT

PURPOSE: The MDM2 and HMGA2 genes are consistently amplified in well-differentiated/dedifferentiated liposarcomas (WDLPS/DDLPS) whereas CDK4 is frequently but not always amplified in these tumors. Our goal was to determine whether the absence of CDK4 amplification was (a) correlated to a specific clinico-histopathologic profile; and (b) compensated by another genomic anomaly involving the CCND1/CDK4/P16INK4a/RB1/E2F pathway. EXPERIMENTAL DESIGN: We compared the clinical characteristics of a series of 143 WDLPS/DDLPS with amplification of both MDM2 and CDK4 (MDM2+/CDK4+) to a series of 45 WDLPS/DDLPS with MDM2 amplification and no CDK4 amplification (MDM2+/CDK4-). We used fluorescence in situ hybridization, real time quantitative reverse transcription PCR, and immunohistochemistry to explore the status of CCND1, P16INK4a, P14ARF, and RB1. RESULTS: We found that MDM2+/CDK4- WDLPS/DDLPS represent a distinct clinical subgroup with favorable prognostic features, including low-grade lipoma-like histology, peripheral location, and lower rate of recurrence. By using fluorescence in situ hybridization, we found that genomic aberrations expected to be alternative mechanisms for compensating the lack of CDK4 amplification, such as RB1 and CDKN2A deletions or CCND1 amplification, were very uncommon. In contrast, by using real time quantitative reverse transcription PCR and immunohistochemistry, we observed that overexpression of P16INK4a (and P14ARF) and CCND1 and reduced expression of RB1 were very frequent, independently of the CDK4 status. CONCLUSIONS: Our results underscore the complex coordinated regulation of the RB and p53 growth-control pathways in WDLPS/DDLPS. Because the absence of CDK4 amplification is not specifically counterbalanced by a genomic alteration of the CCND1/CDK4/P16INK4a/RB1/E2F pathway, CDK4 amplification may only represent a "MDM2-HMGA2-helper" in WDLPS/DDLPS tumorigenesis.


Subject(s)
Cell Dedifferentiation , Cell Differentiation , Cyclin-Dependent Kinase 4/genetics , Gene Amplification/genetics , Liposarcoma/genetics , Liposarcoma/pathology , Adult , Aged , Aged, 80 and over , Cyclin-Dependent Kinase 4/metabolism , Female , Humans , Liposarcoma/diagnosis , Male , Middle Aged , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Young Adult
16.
Virchows Arch ; 452(6): 689-96, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18253748

ABSTRACT

Dermatofibrosarcoma protuberans (DFSP) is a rare, dermal neoplasm of intermediate malignancy. It is made of spindle-shaped tumor cells in a storiform pattern positive for CD34. Cytogenetically, DFSP cells are characterized by either supernumerary ring chromosomes composed of sequences derived from chromosomes 17 and 22 or more rarely of translocations t(17;22). These chromosomal rearrangements lead to the formation of a specific chimeric gene fusing COL1A1 to PDGFB. So far, the COL1A1-PDGFB fusion gene remains the sole fusion gene identified in DFSP. However, some observations suggest that genes, other than COL1A1 and PDGFB, might be involved in some DFSP cases. We report in this paper a DFSP case presenting as a unique chromosomal abnormality a complex translocation between chromosomes 5 and 8. This is the first report of a DFSP case where the lack of chromosomes 17 and 22 rearrangement and the absence of COL1A1-PDGFB fusion gene have been demonstrated. Using fluorescence in situ hybridization analysis, we showed that the CSPG2 gene at 5q14.3 and the PTK2B gene at 8p21.2 were disrupted by this rearrangement. Although rare, the existence of cases of DFSP negative for the COL1A1-PDGFB fusion has to be taken in consideration when performing molecular diagnosis for a tumor suspected to be a DFSP.


Subject(s)
Chromosomes, Human, Pair 8/genetics , Dermatofibrosarcoma/genetics , Focal Adhesion Kinase 2/genetics , Skin Neoplasms/genetics , Translocation, Genetic , Versicans/genetics , Aged , Chromosomes, Human, Pair 5/genetics , Gene Rearrangement , Humans , Male , Oncogene Proteins, Fusion/genetics
17.
Int J Cancer ; 122(10): 2233-41, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18214854

ABSTRACT

Data concerning the fine structure of the 12q13-15 amplicon which contains MDM2 and CDK4 in well-differentiated and dedifferentiated liposarcomas (WDLPS/DDLPS) are scarce. We investigated a series of 38 WDLPS/DDLPS using fluorescence in situ hybridization analysis with 17 probes encompassing the 12q13-15 region. In addition, using quantitative RT-PCR we studied the expression of MDM2, CDK4, DDIT3 (CHOP/GADD153), DYRK2, HMGA2, TSPAN31 and YEATS4 (GAS41) in 11 cases. We showed that CDK4 (12q14.1) belonged to a distinct amplicon than MDM2 (12q15). There was no continuity in the amplified sequences between MDM2 and CDK4. Moreover, while MDM2 was amplified and overexpressed in all cases, CDK4 was not amplified or overexpressed in 13% of cases. The centromeric border of the CDK4 amplicon was located immediately downstream the 5' end of DDIT3, a gene known for being involved in myxoid liposarcoma translocations. DDIT3 was amplified in 3 cases and overexpressed in 9 cases. The overexpression of DDIT3 was correlated to the CDK4 amplification and not to its own amplification status. This suggested that the CDK4 amplicon, as well as the overexpression of DDIT3, might be generated by the disruption of a fragile region in 5' DDIT3. HMGA2 was always amplified and rearranged indicating that it plays a central role in WDLPS/DDLPS. HMGA2 rearrangement frequently resulted in a loss of the 3' end region that is a binding site for let-7. We also found a frequent amplification and overexpression of YEATS4, an oncogene that inactivates P53, suggesting that YEATS4 might play an important role together with MDM2 in WDLPS/DDLPS oncogenesis.


Subject(s)
Cell Differentiation , Chromosomes, Human, Pair 12/genetics , Cyclin-Dependent Kinase 4/genetics , HMGA2 Protein/genetics , Liposarcoma/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Adult , Aged , Aged, 80 and over , Female , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Lipoma/genetics , Lipoma/pathology , Liposarcoma/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor CHOP/genetics , Transcription Factors/genetics
18.
Bull Cancer ; 94(2): 179-89, 2007 Feb.
Article in French | MEDLINE | ID: mdl-17337387

ABSTRACT

Dermatofibrosarcoma protuberans (DP) is a rare, slow growing dermal neoplasm of intermediate malignancy. It is made of spindle-shaped tumor cells in a storiform pattern often positive for CD34. The preferred treatment for DP is a surgical wide excision with pathologically sane margins of 3 cm. At the cytogenetic level, DP cells are characterized by either supernumerary ring chromosomes composed of sequences derived from chromosomes 17 and 22 or more rarely of translocations t(17;22). Rings have been mainly observed in adults whereas translocations have been reported in all pediatric cases. These chromosomal rearrangements lead to the formation of a specific fusion gene : COL1A1-PDGFB detected in rings as well as in translocations. DP is therefore a unique example of tumor in which the same molecular event occurs either on rings or linear translocation derivatives and the chromosomal abnormalities display an age-related pattern. So far, the COL1A1-PDGFB fusion gene remains the only fusion gene identified in this tumor. It is also present in variant forms of DP such as giant cell fibroblastoma, Bednar tumor, adult superficial fibrosarcoma and the granular cell variant of DP demonstrating that these tumors are not distinct entities but morphological variants of DP. The breakpoint localization in PDGFB was found to be remarkably constant, placing exon 2 of PDGFB under the control of the COL1A1 promoter. In contrast, the COL1A1 breakpoint was found to be variably located within the exons of the alpha-helical coding region (exons 7-47). No correlation between the breakpoint location in COL1A1 and the age of the patient or any clinical or histological particularity has been established. Moreover, no preferential breakpoint appears to be more particularly linked to one or another variant of DP. The COL1A1-PDGFB fusion gene is detectable either by multiplex RT-PCR with a combination of forward primers designed from a variety of COL1A1 exons and one reverse primer for PDGFB exon 2, or by in situ fluorescence hybridization (FISH) on interphase nuclei from frozen or fixed paraffin-embbeded sections. The COL1A1-PDGFB fusion gene is not found in approximately 8% of DP cases, suggesting that genes other than COL1A1 or PDGFB might be involved in a small subset of cases. It has been proposed that PDGFB acts as a mitogen in DP cells by autocrine stimulation of the PDGF receptor. The PDGF receptor tyrosine kinase antagonist imatinib mesylate has recently been used in clinical trials; its efficiency has been demonstrated in several cases, which allows it to be considered as a novel treatment strategy for metastatic or locally advanced DP.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 22/genetics , Dermatofibrosarcoma/genetics , Oncogene Proteins, Fusion/genetics , Ring Chromosomes , Skin Neoplasms/genetics , Adult , Age Factors , Antineoplastic Agents/therapeutic use , Benzamides , Child , Dermatofibrosarcoma/pathology , Dermatofibrosarcoma/therapy , Humans , Imatinib Mesylate , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Translocation, Genetic/genetics
20.
Science ; 310(5748): 667-70, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16179433

ABSTRACT

All animals coordinate growth and maturation to reach their final size and shape. In insects, insulin family molecules control growth and metabolism, whereas pulses of the steroid 20-hydroxyecdysone (20E) initiate major developmental transitions. We show that 20E signaling also negatively controls animal growth rates by impeding general insulin signaling involving localization of the transcription factor dFOXO and transcription of the translation inhibitor 4E-BP. We also demonstrate that the larval fat body, equivalent to the vertebrate liver, is a key relay element for ecdysone-dependent growth inhibition. Hence, ecdysone counteracts the growth-promoting action of insulins, thus forming a humoral regulatory loop that determines organismal size.


Subject(s)
Drosophila melanogaster/growth & development , Ecdysterone/physiology , Insulin Antagonists , Insulin/physiology , Animals , Body Size , Crosses, Genetic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila melanogaster/metabolism , Fat Body/physiology , Insect Proteins/physiology , Larva/growth & development , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...