Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nat Commun ; 14(1): 2575, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37142597

ABSTRACT

Noradrenergic and mesenchymal identities have been characterized in neuroblastoma cell lines according to their epigenetic landscapes and core regulatory circuitries. However, their relationship and relative contribution in patient tumors remain poorly defined. We now document spontaneous and reversible plasticity between the two identities, associated with epigenetic reprogramming, in several neuroblastoma models. Interestingly, xenografts with cells from each identity eventually harbor a noradrenergic phenotype suggesting that the microenvironment provides a powerful pressure towards this phenotype. Accordingly, such a noradrenergic cell identity is systematically observed in single-cell RNA-seq of 18 tumor biopsies and 15 PDX models. Yet, a subpopulation of these noradrenergic tumor cells presents with mesenchymal features that are shared with plasticity models, indicating that the plasticity described in these models has relevance in neuroblastoma patients. This work therefore emphasizes that intrinsic plasticity properties of neuroblastoma cells are dependent upon external cues of the environment to drive cell identity.


Subject(s)
Cell Plasticity , Neuroblastoma , Humans , Neuroblastoma/metabolism , Cell Line, Tumor , Tumor Microenvironment/genetics
2.
Clin Cancer Res ; 29(7): 1317-1331, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36602782

ABSTRACT

PURPOSE: ALK-activating mutations are identified in approximately 10% of newly diagnosed neuroblastomas and ALK amplifications in a further 1%-2% of cases. Lorlatinib, a third-generation anaplastic lymphoma kinase (ALK) inhibitor, will soon be given alongside induction chemotherapy for children with ALK-aberrant neuroblastoma. However, resistance to single-agent treatment has been reported and therapies that improve the response duration are urgently required. We studied the preclinical combination of lorlatinib with chemotherapy, or with the MDM2 inhibitor, idasanutlin, as recent data have suggested that ALK inhibitor resistance can be overcome through activation of the p53-MDM2 pathway. EXPERIMENTAL DESIGN: We compared different ALK inhibitors in preclinical models prior to evaluating lorlatinib in combination with chemotherapy or idasanutlin. We developed a triple chemotherapy (CAV: cyclophosphamide, doxorubicin, and vincristine) in vivo dosing schedule and applied this to both neuroblastoma genetically engineered mouse models (GEMM) and patient-derived xenografts (PDX). RESULTS: Lorlatinib in combination with chemotherapy was synergistic in immunocompetent neuroblastoma GEMM. Significant growth inhibition in response to lorlatinib was only observed in the ALK-amplified PDX model with high ALK expression. In this PDX, lorlatinib combined with idasanutlin resulted in complete tumor regression and significantly delayed tumor regrowth. CONCLUSIONS: In our preclinical neuroblastoma models, high ALK expression was associated with lorlatinib response alone or in combination with either chemotherapy or idasanutlin. The synergy between MDM2 and ALK inhibition warrants further evaluation of this combination as a potential clinical approach for children with neuroblastoma.


Subject(s)
Lung Neoplasms , Neuroblastoma , Mice , Animals , Humans , Anaplastic Lymphoma Kinase/genetics , Aminopyridines/therapeutic use , Lactams, Macrocyclic/pharmacology , Lactams, Macrocyclic/therapeutic use , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy
3.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-36054452

ABSTRACT

BACKGROUND: High-risk neuroblastoma is a pediatric cancer with still a dismal prognosis, despite multimodal and intensive therapies. Tumor microenvironment represents a key component of the tumor ecosystem the complexity of which has to be accurately understood to define selective targeting opportunities, including immune-based therapies. METHODS: We combined various approaches including single-cell transcriptomics to dissect the tumor microenvironment of both a transgenic mouse neuroblastoma model and a cohort of 10 biopsies from neuroblastoma patients, either at diagnosis or at relapse. Features of related cells were validated by multicolor flow cytometry and functional assays. RESULTS: We show that the immune microenvironment of MYCN-driven mouse neuroblastoma is characterized by a low content of T cells, several phenotypes of macrophages and a population of cells expressing signatures of myeloid-derived suppressor cells (MDSCs) that are molecularly distinct from the various macrophage subsets. We document two cancer-associated fibroblasts (CAFs) subsets, one of which corresponding to CAF-S1, known to have immunosuppressive functions. Our data unravel a complex content in myeloid cells in patient tumors and further document a striking correspondence of the microenvironment populations between both mouse and human tumors. We show that mouse intratumor T cells exhibit increased expression of inhibitory receptors at the protein level. Consistently, T cells from patients are characterized by features of exhaustion, expressing inhibitory receptors and showing low expression of effector cytokines. We further functionally demonstrate that MDSCs isolated from mouse neuroblastoma have immunosuppressive properties, impairing the proliferation of T lymphocytes. CONCLUSIONS: Our study demonstrates that neuroblastoma tumors have an immunocompromised microenvironment characterized by dysfunctional T cells and accumulation of immunosuppressive cells. Our work provides a new and precious data resource to better understand the neuroblastoma ecosystem and suggest novel therapeutic strategies, targeting both tumor cells and components of the microenvironment.


Subject(s)
Neuroblastoma , Transcriptome , Animals , Child , Ecosystem , Humans , Mice , Neoplasm Recurrence, Local , Neuroblastoma/pathology , Tumor Microenvironment/genetics
4.
Cancers (Basel) ; 14(11)2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35681734

ABSTRACT

Neuroblastoma arising from the adrenal differ from ganglionic neuroblastoma both genetically and clinically, with adrenal tumors being associated with a more severe prognosis. The different tumor properties may be linked to specific tumor founder cells in adrenal and sympathetic ganglia. To address this question, we first set up cultures of mouse sympathetic neuroblasts and adrenal chromaffin cells. These cultures were then treated with various proliferation inhibitors to identify lineage-specific responses. We show that neuroblast and chromaffin cell proliferation was affected by WNT, ALK, IGF1, and PRC2/EZH2 signaling inhibitors to a similar extent. However, differential effects were observed in response to bromodomain and extraterminal (BET) protein inhibitors (JQ1, GSK1324726A) and to the CDK-7 inhibitor THZ1, with BET inhibitors preferentially affecting chromaffin cells, and THZ1 preferentially affecting neuroblasts. The differential dependence of chromaffin cells and neuroblasts on BET and CDK signaling may indicate different mechanisms during tumor initiation in sympathetic ganglia and adrenal.

5.
Cancer Res ; 81(19): 4994-5006, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34341072

ABSTRACT

Ewing sarcoma is characterized by pathognomonic translocations, most frequently fusing EWSR1 with FLI1. An estimated 30% of Ewing sarcoma tumors also display genetic alterations in STAG2, TP53, or CDKN2A (SPC). Numerous attempts to develop relevant Ewing sarcoma models from primary human cells have been unsuccessful in faithfully recapitulating the phenotypic, transcriptomic, and epigenetic features of Ewing sarcoma. In this study, by engineering the t(11;22)(q24;q12) translocation together with a combination of SPC mutations, we generated a wide collection of immortalized cells (EWIma cells) tolerating EWSR1-FLI1 expression from primary mesenchymal stem cells (MSC) derived from a patient with Ewing sarcoma. Within this model, SPC alterations strongly favored Ewing sarcoma oncogenicity. Xenograft experiments with independent EWIma cells induced tumors and metastases in mice, which displayed bona fide features of Ewing sarcoma. EWIma cells presented balanced but also more complex translocation profiles mimicking chromoplexy, which is frequently observed in Ewing sarcoma and other cancers. Collectively, these results demonstrate that bone marrow-derived MSCs are a source of origin for Ewing sarcoma and also provide original experimental models to investigate Ewing sarcomagenesis. SIGNIFICANCE: These findings demonstrate that Ewing sarcoma can originate from human bone-marrow-derived mesenchymal stem cells and that recurrent mutations support EWSR1-FLI1 translocation-mediated transformation.


Subject(s)
Cell Transformation, Neoplastic , Disease Susceptibility , Mesenchymal Stem Cells/metabolism , Sarcoma, Ewing/etiology , Sarcoma, Ewing/metabolism , Animals , Biomarkers , CRISPR-Cas Systems , Cells, Cultured , Computational Biology/methods , Disease Models, Animal , Gene Editing , Gene Expression Profiling , Gene Rearrangement , Gene Targeting , Heterografts , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Mesenchymal Stem Cells/pathology , Mice , Mutation , Sarcoma, Ewing/pathology , Translocation, Genetic
6.
Oncotarget ; 10(48): 4937-4950, 2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31452835

ABSTRACT

The ALK gene is a major oncogene of neuroblastoma cases exhibiting ALK activating mutations. Here, we characterized two neuroblastoma cell lines established from a stage 4 patient at diagnosis either from the primary tumor (PT) or from the bone marrow (BM). Both cell lines exhibited similar genomic profiles. All cells in the BM-derived cell line exhibited an ALK F1174L mutation, whereas this mutation was present in only 5% of the cells in the earliest passages of the PT-derived cell line. The BM-derived cell line presented with a higher proliferation rate in vitro and injections in Nude mice resulted in tumor formation only for the BM-derived cell line. Next, we observed that the F1174L mutation frequency in the PT-derived cell line increased with successive passages. Further Whole Exome Sequencing revealed a second ALK mutation, L1196M, in this cell line. Digital droplet PCR documented that the allele fractions of both mutations changed upon passages, and that the F1174L mutation reached 50% in late passages, indicating clonal evolution. In vitro treatment of the PT-derived cell line exhibiting the F1174L and L1196M mutations with the alectinib inhibitor resulted in an enrichment of the L1196M mutation. Using xenografts, we documented a better efficacy of alectinib compared to crizotinib on tumor growth and an enrichment of the L1196M mutation at the end of both treatments. Finally, single-cell RNA-seq analysis was consistent with both mutations resulting in ALK activation. Altogether, this study provides novel insights into ALK mutation dynamics in a neuroblastoma model harbouring two ALK mutations.

7.
Oncogene ; 37(11): 1417-1429, 2018 03.
Article in English | MEDLINE | ID: mdl-29321660

ABSTRACT

Activating mutations of the ALK receptor occur in a subset of neuroblastoma tumors. We previously demonstrated that Alk mutations cooperate with MYCN overexpression to induce neuroblastoma in mice and identified Ret as being strongly upregulated in MYCN/Alkmut tumors. By a genetic approach in vivo, we now document an oncogenic cooperation between activated Ret and MYCN overexpression in neuroblastoma formation. We show that MYCN/RetM919T tumors exhibit histological features and expression profiles close to MYCN/Alkmut tumors. We show that RET transcript levels decrease precedes RET protein levels decrease upon ALK inhibition in neuroblastoma cell lines. Etv5 was identified as a candidate transcription factor regulating Ret expression from murine MYCN/Alkmut tumor transcriptomic data. We demonstrate that ETV5 is regulated both at the protein and mRNA levels upon ALK activation or inhibition in neuroblastoma cell lines and that this regulation precedes RET modulation. We document that ALK activation induces ETV5 protein upregulation through stabilization in a MEK/ERK-dependent manner. We show that RNAi-mediated inhibition of ETV5 decreases RET expression. Reporter assays indicate that ETV5 is able to drive RET gene transcription. ChIP-seq analysis confirmed ETV5 binding on the RET promoter and identified an enhancer upstream of the promoter. Finally, we demonstrate that combining RET and ALK inhibitors reduces tumor growth more efficiently than each single agent in MYCN and AlkF1178L-driven murine neuroblastoma. Altogether, these results define the ERK-ETV5-RET pathway as a critical axis driving neuroblastoma oncogenesis downstream of activated ALK.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Carcinogenesis/genetics , Gain of Function Mutation , Neuroblastoma/genetics , Anaplastic Lymphoma Kinase/metabolism , Animals , Carcinogenesis/pathology , Cells, Cultured , DNA-Binding Proteins/physiology , Female , Gain of Function Mutation/physiology , HEK293 Cells , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neuroblastoma/pathology , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction/genetics , Transcription Factors/physiology , Xenograft Model Antitumor Assays
8.
Sci Rep ; 7(1): 16901, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203791

ABSTRACT

Metabolic diseases are characterized by a decreased action of insulin. During the course of the disease, usual treatments frequently fail and patients are finally submitted to insulinotherapy. There is thus a need for innovative therapeutic strategies to improve insulin action. Growth factor receptor-bound protein 14 (Grb14) is a molecular adapter that specifically binds to the activated insulin receptor (IR) and inhibits its tyrosine kinase activity. Molecules disrupting Grb14-IR binding are therefore potential insulin-sensitizing agents. We used Structure-Based Virtual Ligand Screening to generate a list of 1000 molecules predicted to hinder Grb14-IR binding. Using an acellular bioluminescence resonance energy transfer (BRET) assay, we identified, out of these 1000 molecules, 3 compounds that inhibited Grb14-IR interaction. Their inhibitory effect on insulin-induced Grb14-IR interaction was confirmed in co-immunoprecipitation experiments. The more efficient molecule (C8) was further characterized. C8 increased downstream Ras-Raf and PI3-kinase insulin signaling, as shown by BRET experiments in living cells. Moreover, C8 regulated the expression of insulin target genes in mouse primary hepatocytes. These results indicate that C8, by reducing Grb14-IR interaction, increases insulin signalling. The use of C8 as a lead compound should allow for the development of new molecules of potential therapeutic interest for the treatment of diabetes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Receptor, Insulin/metabolism , Sulfanilamides/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Binding Sites , Cell Survival/drug effects , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Insulin/metabolism , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Receptor, Insulin/chemistry , Signal Transduction/drug effects , Sulfanilamides/metabolism , Sulfanilamides/pharmacology
9.
Nat Genet ; 49(9): 1408-1413, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28740262

ABSTRACT

Neuroblastoma is a tumor of the peripheral sympathetic nervous system, derived from multipotent neural crest cells (NCCs). To define core regulatory circuitries (CRCs) controlling the gene expression program of neuroblastoma, we established and analyzed the neuroblastoma super-enhancer landscape. We discovered three types of identity in neuroblastoma cell lines: a sympathetic noradrenergic identity, defined by a CRC module including the PHOX2B, HAND2 and GATA3 transcription factors (TFs); an NCC-like identity, driven by a CRC module containing AP-1 TFs; and a mixed type, further deconvoluted at the single-cell level. Treatment of the mixed type with chemotherapeutic agents resulted in enrichment of NCC-like cells. The noradrenergic module was validated by ChIP-seq. Functional studies demonstrated dependency of neuroblastoma with noradrenergic identity on PHOX2B, evocative of lineage addiction. Most neuroblastoma primary tumors express TFs from the noradrenergic and NCC-like modules. Our data demonstrate a previously unknown aspect of tumor heterogeneity relevant for neuroblastoma treatment strategies.


Subject(s)
Cell Lineage/genetics , Gene Expression Regulation, Neoplastic/genetics , Neuroblastoma/genetics , Transcription Factors/genetics , Animals , Blotting, Western , Cell Line, Tumor/classification , Cell Lineage/drug effects , Doxycycline/pharmacology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Genetic Heterogeneity , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , RNA Interference , RNAi Therapeutics , Reverse Transcriptase Polymerase Chain Reaction , Single-Cell Analysis , Transcription Factors/metabolism , Xenograft Model Antitumor Assays/methods
10.
Oncotarget ; 7(12): 14898-911, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26910909

ABSTRACT

CD95, a member of the death receptor family initiates a caspase-dependent apoptosis, when activated by its ligand CD95L, thought to negatively regulate erythrocyte production in the bone marrow. We have previously shown that CD95 is overexpressed in two thirds of patients with a lower risk myelodysplastic syndrome (MDS) and that resistance to erythropoiesis-stimulating agents (ESA) is linked to poor residual erythropoiesis. In the present study, we show that CD95 overexpression and previous transfusion are independent predictive factors of ESA resistance. To investigate an alternative therapeutic strategy of anemia in ESA-resistant patients, we have conducted a preclinical study of the effects of APG101, a fusion protein consisting of the extracellular domain of human CD95 and the Fc region of human IgG1 on MDS erythropoiesis in vitro. APG101 increases the number of burst-forming unit-erythroid (BFU-E) progenitors derived from CD34+ progenitors in liquid culture and improves overall proliferation rate of erythroid precursors by inhibiting apoptosis. APG101 rescues BFU-E growth in MDS patients presenting with attrition of erythroid progenitors at baseline, independently of CD95 or CD95L expression level. Our data show that overexpression of CD95 at diagnosis is a hallmark of ESA resistance and that severe impairment of erythropoiesis is predictive of erythroid response to APG101 in vitro. These data provide a rationale for further clinical investigation of APG101 in an attempt to treat anemia in lower risk MDS patients.


Subject(s)
Erythropoiesis , Hematopoiesis , Immunoglobulin G/metabolism , Myelodysplastic Syndromes/prevention & control , Recombinant Fusion Proteins/metabolism , fas Receptor/metabolism , Aged , Aged, 80 and over , Apoptosis , Case-Control Studies , Cell Proliferation , Cells, Cultured , Female , Follow-Up Studies , Humans , Immunoglobulin G/genetics , Male , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Prognosis , Recombinant Fusion Proteins/genetics , Survival Rate , fas Receptor/genetics
11.
Arch Physiol Biochem ; 122(2): 54-60, 2016.
Article in English | MEDLINE | ID: mdl-26707268

ABSTRACT

CONTEXT: Insulin analogues are largely used for the treatment of diabetic patients, but concerns have been raised about their mitogenic/anti-apoptotic potential. It is therefore important to evaluate these analogues in different cell systems. OBJECTIVE: The aim of this work was to establish the pharmacological profiles of insulin analogues towards PI-3 kinase/Akt pathway in INS-1 ß-pancreatic cells. METHODS: Bioluminescence Resonance Energy Transfer (BRET), in cell western and caspase 3/7 assays, was used to study the effects of ligands. RESULTS: Among the five analogues evaluated, only glargine stimulated PI-3 kinase/Akt pathway with higher efficiency than insulin, whereas glargine's metabolite M1 was less efficient. However, glargine did not show higher anti-apoptotic efficiency than insulin. CONCLUSION: Glargine was more efficient than insulin for the activation of PI-3 kinase/Akt pathway, but not for the inhibition of caspase 3/7 activity. Moreover, glargine's metabolite M1 displayed lower efficiency than insulin towards PI-3 kinase/Akt activation and caspase 3/7 inhibition.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin/analogs & derivatives , Insulin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Cell Line , Insulin Glargine/metabolism , Insulin-Secreting Cells/drug effects , Phosphatidylinositol Phosphates/biosynthesis , Rats
12.
J Neurosci ; 35(50): 16531-44, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26674877

ABSTRACT

The RNA binding protein Lin28B is expressed in developing tissues and sustains stem and progenitor cell identity as a negative regulator of the Let-7 family of microRNAs, which induces differentiation. Lin28B is activated in neuroblastoma (NB), a childhood tumor in sympathetic ganglia and adrenal medulla. Forced expression of Lin28B in embryonic mouse sympathoadrenal neuroblasts elicits postnatal NB formation. However, the normal function of Lin28B in the development of sympathetic neurons and chromaffin cells and the mechanisms involved in Lin28B-induced tumor formation are unclear. Here, we demonstrate a mirror-image expression of Lin28B and Let-7a in developing chick sympathetic ganglia. Lin28B expression is not restricted to undifferentiated progenitor cells but, is observed in proliferating noradrenergic neuroblasts. Lin28 knockdown in cultured sympathetic neuroblasts decreases proliferation, whereas Let-7 inhibition increases the proportion of neuroblasts in the cell cycle. Lin28B overexpression enhances proliferation, but only during a short developmental period, and it does not reduce Let-7a. Effects of in vivo Lin28B overexpression were analyzed in the LSL-Lin28B(DBHiCre) mouse line. Sympathetic ganglion and adrenal medulla volume and the expression level of Let-7a were not altered, although Lin28B expression increased by 12- to 17-fold. In contrast, Let-7a expression was strongly reduced in LSL-Lin28B(DbhiCre) NB tumor tissue. These data demonstrate essential functions for endogenous Lin28 and Let-7 in neuroblast proliferation. However, Lin28B overexpression neither sustains neuroblast proliferation nor affects let-7 expression. Thus, in contrast to other pediatric tumors, Lin28B-induced NB is not due to expansion of proliferating embryonic neuroblasts, and Let-7-independent functions are implicated during initial NB development. SIGNIFICANCE STATEMENT: Lin28A/B proteins are highly expressed in early development and maintain progenitor cells by blocking the biogenesis and differentiation function of Let-7 microRNAs. Lin28B is aberrantly upregulated in the childhood tumor neuroblastoma (NB). NB develops in sympathetic ganglia and adrenal medulla and is elicited by forced Lin28B expression. We demonstrate that Lin28A/B and Let-7 are essential for sympathetic neuroblast proliferation during normal development. Unexpectedly, Lin28B upregulation in a mouse model does not affect neuroblast proliferation, ganglion size, and Let-7 expression during early postnatal development. Lin28B-induced NB, in contrast to other pediatric cancers, does not evolve from neuroblasts that continue to divide and involves Let-7-independent functions during initial development.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA-Binding Proteins/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Sympathetic Nervous System/growth & development , Adrenal Glands/metabolism , Animals , Cell Proliferation , Chick Embryo , DNA-Binding Proteins/physiology , Ganglia, Sympathetic/pathology , Mice , Mice, 129 Strain , MicroRNAs/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , RNA-Binding Proteins , Stem Cells/metabolism , Sympathetic Nervous System/physiology
13.
Oncotarget ; 5(9): 2703-13, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24811761

ABSTRACT

The ALK (Anaplastic Lymphoma Kinase) gene encodes a tyrosine kinase receptor preferentially expressed in the central and peripheral nervous systems. A syndromic presentation associating congenital neuroblastoma with severe encephalopathy and an abnormal shape of the brainstem has been described in patients harbouring de novo germline F1174V and F1245V ALK mutations. Here, we investigated the phenotype of knock-in (KI) mice bearing the AlkF1178L mutation (F1174L in human). Although heterozygous KI mice did not reproduce the severe breathing and feeding difficulties observed in human patients, behavioral tests documented a reduced activity during dark phases and an increased anxiety of mutated mice. Matings of heterozygotes yielded the expected proportions of wild-type, heterozygotes and homozygotes at birth but a high neonatal lethality was noticed for homozygotes. We documented Alk expression in several motor nuclei of the brainstem involved in the control of sucking and swallowing. Evaluation of basic physiological functions 12 hours after birth revealed slightly more apneas but a dramatic reduced milk intake for homozygotes compared to control littermates. Overall, our data demonstrate that Alk activation above a critical threshold is not compatible with survival in mice, in agreement with the extremely severe phenotype of patients carrying aggressive de novo ALK germline mutations.


Subject(s)
Behavior, Animal/physiology , Eating , Mutation/genetics , Neuroblastoma/genetics , Receptor Protein-Tyrosine Kinases/physiology , Respiration , Anaplastic Lymphoma Kinase , Animals , Animals, Newborn , Genes, Lethal , Humans , Immunoenzyme Techniques , Male , Mice , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phenotype
14.
Oncotarget ; 5(9): 2688-702, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24811913

ABSTRACT

Activating mutations of the ALK (Anaplastic lymphoma Kinase) gene have been identified in sporadic and familial cases of neuroblastoma, a cancer of early childhood arising from the sympathetic nervous system (SNS). To decipher ALK function in neuroblastoma predisposition and oncogenesis, we have characterized knock-in (KI) mice bearing the two most frequent mutations observed in neuroblastoma patients. A dramatic enlargement of sympathetic ganglia is observed in AlkF1178L mice from embryonic to adult stages associated with an increased proliferation of sympathetic neuroblasts from E14.5 to birth. In a MYCN transgenic context, the F1178L mutation displays a higher oncogenic potential than the R1279Q mutation as evident from a shorter latency of tumor onset. We show that tumors expressing the R1279Q mutation are sensitive to ALK inhibition upon crizotinib treatment. Furthermore, our data provide evidence that activated ALK triggers RET upregulation in mouse sympathetic ganglia at birth as well as in murine and human neuroblastoma. Using vandetanib, we show that RET inhibition strongly impairs tumor growth in vivo in both MYCN/KI AlkR1279Q and MYCN/KI AlkF1178L mice. Altogether, our findings demonstrate the critical role of activated ALK in SNS development and pathogenesis and identify RET as a therapeutic target in ALK mutated neuroblastoma.


Subject(s)
Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Mutation/genetics , Neuroblastoma/genetics , Neurogenesis , Proto-Oncogene Proteins c-ret/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Animals , Base Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Southern , Blotting, Western , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Integrases , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , N-Myc Proto-Oncogene Protein , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ret/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcriptional Activation , Tumor Cells, Cultured
15.
PLoS One ; 9(3): e92737, 2014.
Article in English | MEDLINE | ID: mdl-24647478

ABSTRACT

Stimulation of tyrosine kinase receptors initiates a signaling cascade that activates PI3K. Activated PI3K uses PIP2 to generate PIP3, which recruit Akt to the plasma membrane through its pleckstrin homology (PH) domain, permitting its activation by PDKs. Activated Akt controls important biological functions, including cell metabolism, proliferation and survival. The PI3K pathway is therefore an attractive target for drug discovery. However, current assays for measurement of PIP3 production are technically demanding and not amenable to high-throughput screening. We have established a MCF-7-derived breast cancer cell line, that stably co-expresses the PH domain of Akt fused to Renilla luciferase and YFP fused to a membrane localization signal. This BRET biosensor pair permits to monitor, in real time, in living cells, PIP3 production at the plasma membrane upon stimulation by different ligands, including insulin, the insulin analogue glargine, IGF1, IGF2 and EGF. Moreover, several known inhibitors that target different steps of the PI3K/Akt pathway caused inhibition of ligand-induced BRET. Cetuximab, a humanized anti-EGF receptor monoclonal antibody used for the treatment of cancer, completely inhibited EGF-induced BRET, and the tyrosine kinase inhibitor tyrphostine AG1024 inhibited insulin effect on PIP3 production. Moreover, the effects of insulin and IGF1 were inhibited by molecules that inhibit PI3K catalytic activity or the interaction between PIP3 and the PH domain of Akt. Finally, we showed that human serum induced a dose-dependent increase in BRET signal, suggesting that this stable clone may be used as a prognostic tool to evaluate the PI3K stimulatory activity present in serum of human patients. We have thus established a cell line, suitable for the screening and/or the study of molecules with stimulatory or inhibitory activities on the PI3K/Akt pathway that will constitute a new tool for translational research in diabetes and cancer.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Biosensing Techniques/methods , Phosphatidylinositol Phosphates/analysis , Breast Neoplasms/metabolism , Cell Line, Tumor , Humans
16.
FASEB J ; 28(2): 1010-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24174424

ABSTRACT

O-GlcNAcylation on serine/threonine is a post-translational modification that controls the activity of nucleocytoplasmic proteins according to glucose availability. We previously showed that O-GlcNAcylation of FoxO1 in liver cells increases its transcriptional activity. In the present study, we evaluated the potential involvement of FoxO1 O-GlcNAcylation in the context of pancreatic ß-cell glucotoxicity. FoxO1 was O-GlcNAcylated in INS-1 832/13 ß cells and isolated rat pancreatic islets. O-GlcNAcylation of FoxO1 resulted in a 2-fold increase in its transcriptional activity toward a FoxO1 reporter gene and a 3-fold increase in the expression of the insulin-like growth factor-binding protein 1 (Igfbp1) gene at the mRNA level, resulting in IGFBP1 protein oversecretion by the cells. Of note, increased IGFBP1 in the culture medium inhibited the activity of the insulin-like growth factor 1 receptor (IGF1R)/phosphatidyl inositol 3 kinase (PI3K)/Akt pathway. We reveal in this report a novel mechanism by which O-GlcNAcylation inhibits Akt activity through an autocrine mechanism. However, although inhibition of IGFBP1 expression using siRNA restored the PI3 kinase/Akt pathway, it did not rescue INS-1 832/13 cells from high-glucose- or O-glcNAcylation-induced cell death. In contrast, FoxO1 down-regulation by siRNA led to 30 to 60% protection of INS-1 832/13 cells from death mediated by glucotoxic conditions. Therefore, whereas FoxO1 O-GlcNAcylation inhibits Akt through an IGFBP1-mediated autocrine pathway, the deleterious effects of FoxO1 O-GlcNAcylation on cell survival appeared to be independent of this pathway.


Subject(s)
Forkhead Transcription Factors/metabolism , Insulin-Like Growth Factor Binding Protein 1/metabolism , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/genetics , Glucose/pharmacology , Immunoprecipitation , Insulin-Like Growth Factor Binding Protein 1/genetics , Rats
17.
PLoS One ; 8(7): e69150, 2013.
Article in English | MEDLINE | ID: mdl-23935944

ABSTRACT

O-GlcNAcylation (addition of N-acetyl-glucosamine on serine or threonine residues) is a post-translational modification that regulates stability, activity or localization of cytosolic and nuclear proteins. O-linked N-acetylgluocosmaine transferase (OGT) uses UDP-GlcNAc, produced in the hexosamine biosynthetic pathway to O-GlcNacylate proteins. Removal of O-GlcNAc from proteins is catalyzed by the ß-N-Acetylglucosaminidase (OGA). Recent evidences suggest that O-GlcNAcylation may affect the growth of cancer cells. However, the consequences of O-GlcNAcylation on anti-cancer therapy have not been evaluated. In this work, we studied the effects of O-GlcNAcylation on tamoxifen-induced cell death in the breast cancer-derived MCF-7 cells. Treatments that increase O-GlcNAcylation (PUGNAc and/or glucosoamine) protected MCF-7 cells from death induced by tamoxifen. In contrast, inhibition of OGT expression by siRNA potentiated the effect of tamoxifen on cell death. Since the PI-3 kinase/Akt pathway is a major regulator of cell survival, we used BRET to evaluate the effect of PUGNAc+glucosamine on PIP3 production. We observed that these treatments stimulated PIP3 production in MCF-7 cells. This effect was associated with an increase in Akt phosphorylation. However, the PI-3 kinase inhibitor LY294002, which abolished the effect of PUGNAc+glucosamine on Akt phosphorylation, did not impair the protective effects of PUGNAc+glucosamine against tamoxifen-induced cell death. These results suggest that the protective effects of O-GlcNAcylation are independent of the PI-3 kinase/Akt pathway. As tamoxifen sensitivity depends on the estrogen receptor (ERα) expression level, we evaluated the effect of PUGNAc+glucosamine on the expression of this receptor. We observed that O-GlcNAcylation-inducing treatment significantly reduced the expression of ERα mRNA and protein, suggesting a potential mechanism for the decreased tamoxifen sensitivity induced by these treatments. Therefore, our results suggest that inhibition of O-GlcNAcylation may constitute an interesting approach to improve the sensitivity of breast cancer to anti-estrogen therapy.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Protein Processing, Post-Translational , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Biosynthetic Pathways , Breast Neoplasms/metabolism , Cell Death/drug effects , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Hexosamines/biosynthesis , Humans , Insulin-Like Growth Factor I/pharmacology , MCF-7 Cells , Oximes/pharmacology , Phenylcarbamates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tamoxifen/analogs & derivatives
18.
PLoS One ; 8(8): e72182, 2013.
Article in English | MEDLINE | ID: mdl-23991058

ABSTRACT

Neuroblastoma is a pediatric cancer of the peripheral nervous system in which structural chromosome aberrations are emblematic of aggressive tumors. In this study, we performed an in-depth analysis of somatic rearrangements in two neuroblastoma cell lines and two primary tumors using paired-end sequencing of mate-pair libraries and RNA-seq. The cell lines presented with typical genetic alterations of neuroblastoma and the two tumors belong to the group of neuroblastoma exhibiting a profile of chromothripsis. Inter and intra-chromosomal rearrangements were identified in the four samples, allowing in particular characterization of unbalanced translocations at high resolution. Using complementary experiments, we further characterized 51 rearrangements at the base pair resolution that revealed 59 DNA junctions. In a subset of cases, complex rearrangements were observed with templated insertion of fragments of nearby sequences. Although we did not identify known particular motifs in the local environment of the breakpoints, we documented frequent microhomologies at the junctions in both chromothripsis and non-chromothripsis associated breakpoints. RNA-seq experiments confirmed expression of several predicted chimeric genes and genes with disrupted exon structure including ALK, NBAS, FHIT, PTPRD and ODZ4. Our study therefore indicates that both non-homologous end joining-mediated repair and replicative processes may account for genomic rearrangements in neuroblastoma. RNA-seq analysis allows the identification of the subset of abnormal transcripts expressed from genomic rearrangements that may be involved in neuroblastoma oncogenesis.


Subject(s)
Chromosome Aberrations , Chromosome Breakpoints , Gene Rearrangement/genetics , Neuroblastoma/genetics , Translocation, Genetic/genetics , Acid Anhydride Hydrolases/genetics , Anaplastic Lymphoma Kinase , Base Sequence , Cell Line, Tumor , Child, Preschool , DNA Copy Number Variations , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Neuroblastoma/pathology , Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Transcription, Genetic
19.
PLoS One ; 7(7): e41992, 2012.
Article in English | MEDLINE | ID: mdl-22848683

ABSTRACT

BACKGROUND: In diabetic patients, the pharmacokinetics of injected human insulin does not permit optimal control of glycemia. Fast and slow acting insulin analogues have been developed, but they may have adverse properties, such as increased mitogenic or anti-apoptotic signaling. Insulin/IGF1 hybrid receptors (IR/IGF1R), present in most tissues, have been proposed to transmit biological effects close to those of IGF1R. However, the study of hybrid receptors is difficult because of the presence of IR and IGF1R homodimers. Our objective was to perform the first study on the pharmacological properties of the five marketed insulin analogues towards IR/IGF1R hybrids. METHODOLOGY: To study the effect of insulin analogues on IR/IGF1R hybrids, we used our previously developed Bioluminescence Resonance Energy Transfer (BRET) assay that permits specific analysis of the pharmacological properties of hybrid receptors. Moreover, we have developed a new, highly sensitive BRET-based assay to monitor phophatidylinositol-3 phosphate (PIP(3)) production in living cells. Using this assay, we performed a detailed pharmacological analysis of PIP(3) production induced by IGF1, insulin and insulin analogues in living breast cancer-derived MCF-7 and MDA-MB231 cells. RESULTS: Among the five insulin analogues tested, only glargine stimulated IR/IGF1R hybrids with an EC50 that was significantly lower than insulin and close to that of IGF1. Glargine more efficiently stimulated PIP(3) production in MCF-7 cells but not in MDA-MB231 cells as compared to insulin. In contrast, glargine metabolites M1 and M2 showed lower potency for hybrid receptors stimulation, PIP(3) production, Akt and Erk1/2 phosphorylation and DNA synthesis in MCF-7 cells, compared to insulin. CONCLUSION: Glargine, possibly acting through IR/IGF1R hybrids, displays higher potency, whereas its metabolites M1 and M2 display lower potency than insulin for the stimulation of proliferative/anti-apoptotic pathways in MCF-7 cells.


Subject(s)
Insulin, Long-Acting/metabolism , Insulin, Long-Acting/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/metabolism , HEK293 Cells , Humans , Insulin Glargine , MCF-7 Cells , Phosphatidylinositol Phosphates/biosynthesis , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...