Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Nature ; 620(7973): 402-408, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532929

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) regulates tumour initiation, progression, metastasis and resistance to anti-cancer therapy1-7. Although great progress has been made in understanding the role of EMT and its regulatory mechanisms in cancer, no therapeutic strategy to pharmacologically target EMT has been identified. Here we found that netrin-1 is upregulated in a primary mouse model of skin squamous cell carcinoma (SCC) exhibiting spontaneous EMT. Pharmacological inhibition of netrin-1 by administration of NP137, a netrin-1-blocking monoclonal antibody currently used in clinical trials in human cancer (ClinicalTrials.gov identifier NCT02977195 ), decreased the proportion of EMT tumour cells in skin SCC, decreased the number of metastases and increased the sensitivity of tumour cells to chemotherapy. Single-cell RNA sequencing revealed the presence of different EMT states, including epithelial, early and late hybrid EMT, and full EMT states, in control SCC. By contrast, administration of NP137 prevented the progression of cancer cells towards a late EMT state and sustained tumour epithelial states. Short hairpin RNA knockdown of netrin-1 and its receptor UNC5B in EPCAM+ tumour cells inhibited EMT in vitro in the absence of stromal cells and regulated a common gene signature that promotes tumour epithelial state and restricts EMT. To assess the relevance of these findings to human cancers, we treated mice transplanted with the A549 human cancer cell line-which undergoes EMT following TGFß1 administration8,9-with NP137. Netrin-1 inhibition decreased EMT in these transplanted A549 cells. Together, our results identify a pharmacological strategy for targeting EMT in cancer, opening up novel therapeutic interventions for anti-cancer therapy.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Squamous Cell , Epithelial-Mesenchymal Transition , Netrin-1 , Skin Neoplasms , Animals , Humans , Mice , A549 Cells , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Netrin Receptors/antagonists & inhibitors , Netrin Receptors/deficiency , Netrin Receptors/genetics , Netrin-1/antagonists & inhibitors , Netrin-1/deficiency , Netrin-1/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Disease Models, Animal , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Neoplasm Metastasis/drug therapy , Single-Cell Gene Expression Analysis , RNA-Seq , Epithelial Cell Adhesion Molecule/metabolism , Xenograft Model Antitumor Assays , Transforming Growth Factor beta1/pharmacology
2.
Nature ; 620(7973): 409-416, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532934

ABSTRACT

Netrin-1 is upregulated in cancers as a protumoural mechanism1. Here we describe netrin-1 upregulation in a majority of human endometrial carcinomas (ECs) and demonstrate that netrin-1 blockade, using an anti-netrin-1 antibody (NP137), is effective in reduction of tumour progression in an EC mouse model. We next examined the efficacy of NP137, as a first-in-class single agent, in a Phase I trial comprising 14 patients with advanced EC. As best response we observed 8 stable disease (8 out of 14, 57.1%) and 1 objective response as RECIST v.1.1 (partial response, 1 out of 14 (7.1%), 51.16% reduction in target lesions at 6 weeks and up to 54.65% reduction during the following 6 months). To evaluate the NP137 mechanism of action, mouse tumour gene profiling was performed, and we observed, in addition to cell death induction, that NP137 inhibited epithelial-to-mesenchymal transition (EMT). By performing bulk RNA sequencing (RNA-seq), spatial transcriptomics and single-cell RNA-seq on paired pre- and on-treatment biopsies from patients with EC from the NP137 trial, we noted a net reduction in tumour EMT. This was associated with changes in immune infiltrate and increased interactions between cancer cells and the tumour microenvironment. Given the importance of EMT in resistance to current standards of care2, we show in the EC mouse model that a combination of NP137 with carboplatin-paclitaxel outperformed carboplatin-paclitaxel alone. Our results identify netrin-1 blockade as a clinical strategy triggering both tumour debulking and EMT inhibition, thus potentially alleviating resistance to standard treatments.


Subject(s)
Endometrial Neoplasms , Epithelial-Mesenchymal Transition , Netrin-1 , Animals , Female , Humans , Mice , Biopsy , Carboplatin/administration & dosage , Carboplatin/pharmacology , Carboplatin/therapeutic use , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Profiling , Netrin-1/antagonists & inhibitors , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , RNA-Seq , Single-Cell Gene Expression Analysis , Tumor Microenvironment/drug effects
3.
Mol Cell ; 72(3): 413-425.e5, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30293784

ABSTRACT

c-Kit is a classic proto-oncogene either mutated or upregulated in cancer cells, and this leads to its constitutive kinase activation and, thus, to uncontrolled proliferation. Although the pro-oncogenic role of c-Kit is of no doubt, some observations do not fit well with c-Kit solely as a tumor-promoting moiety. We show here that c-Kit actively triggers cell death in various cancer cell lines unless engaged by its ligand stem cell factor (SCF). This pro-death activity is enhanced when the kinase activation of c-Kit is silenced and is due to c-Kit intracellular cleavage by caspase-like protease at D816. Moreover, in vivo, overexpression of a c-Kit kinase-dead mutant inhibits tumor growth, and this intrinsic c-Kit tumor-suppressive activity is dependent on the D816 cleavage. Thus, c-Kit acts both as a proto-oncogene via its kinase activity and as a tumor suppressor via its dependence receptor activity.


Subject(s)
Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/physiology , Animals , Apoptosis , Cell Division , Cell Line, Tumor , Female , Humans , Mice , Mice, SCID , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogenes , Stem Cell Factor/metabolism
4.
EMBO J ; 40(3): e105537, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33351190

ABSTRACT

The netrin-1/DCC ligand/receptor pair has key roles in central nervous system (CNS) development, mediating axonal, and neuronal navigation. Although expression of netrin-1 and DCC is maintained in the adult brain, little is known about their role in mature neurons. Notably, netrin-1 is highly expressed in the adult substantia nigra, leading us to investigate a role of the netrin-1/DCC pair in adult nigral neuron fate. Here, we show that silencing netrin-1 in the adult substantia nigra of mice induces DCC cleavage and a significant loss of dopamine neurons, resulting in motor deficits. Because loss of adult dopamine neurons and motor impairments are features of Parkinson's disease (PD), we studied the potential impact of netrin-1 in different animal models of PD. We demonstrate that both overexpression of netrin-1 and brain administration of recombinant netrin-1 are neuroprotective and neurorestorative in mouse and rat models of PD. Of interest, we observed that netrin-1 levels are significantly reduced in PD patient brain samples. These results highlight the key role of netrin-1 in adult dopamine neuron fate, and the therapeutic potential of targeting netrin-1 signaling in PD.


Subject(s)
DCC Receptor/metabolism , Netrin-1/genetics , Netrin-1/metabolism , Parkinson Disease/genetics , Substantia Nigra/cytology , Animals , Cell Death , Disease Models, Animal , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Down-Regulation , Female , Gene Silencing , Humans , Male , Mice , Parkinson Disease/etiology , Parkinson Disease/metabolism , Rats , Signal Transduction , Substantia Nigra/metabolism
5.
J Neurosci ; 43(29): 5414-5430, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37286351

ABSTRACT

Multiple myeloma (MM) is a neoplasia of B plasma cells that often induces bone pain. However, the mechanisms underlying myeloma-induced bone pain (MIBP) are mostly unknown. Using a syngeneic MM mouse model, we show that periosteal nerve sprouting of calcitonin gene-related peptide (CGRP+) and growth associated protein 43 (GAP43+) fibers occurs concurrent to the onset of nociception and its blockade provides transient pain relief. MM patient samples also showed increased periosteal innervation. Mechanistically, we investigated MM induced gene expression changes in the dorsal root ganglia (DRG) innervating the MM-bearing bone of male mice and found alterations in pathways associated with cell cycle, immune response and neuronal signaling. The MM transcriptional signature was consistent with metastatic MM infiltration to the DRG, a never-before described feature of the disease that we further demonstrated histologically. In the DRG, MM cells caused loss of vascularization and neuronal injury, which may contribute to late-stage MIBP. Interestingly, the transcriptional signature of a MM patient was consistent with MM cell infiltration to the DRG. Overall, our results suggest that MM induces a plethora of peripheral nervous system alterations that may contribute to the failure of current analgesics and suggest neuroprotective drugs as appropriate strategies to treat early onset MIBP.SIGNIFICANCE STATEMENT Multiple myeloma (MM) is a painful bone marrow cancer that significantly impairs the quality of life of the patients. Analgesic therapies for myeloma-induced bone pain (MIBP) are limited and often ineffective, and the mechanisms of MIBP remain unknown. In this manuscript, we describe cancer-induced periosteal nerve sprouting in a mouse model of MIBP, where we also encounter metastasis to the dorsal root ganglia (DRG), a never-before described feature of the disease. Concomitant to myeloma infiltration, the lumbar DRGs presented blood vessel damage and transcriptional alterations, which may mediate MIBP. Explorative studies on human tissue support our preclinical findings. Understanding the mechanisms of MIBP is crucial to develop targeted analgesic with better efficacy and fewer side effects for this patient population.


Subject(s)
Bone Diseases , Multiple Myeloma , Nerve Tissue , Humans , Mice , Male , Animals , Multiple Myeloma/complications , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Quality of Life , Pain/metabolism , Nerve Tissue/metabolism , Nerve Tissue/pathology , Ganglia, Spinal/metabolism
6.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34470826

ABSTRACT

Netrin-1, a secreted protein recently characterized as a relevant cancer therapeutic target, is the antiapoptotic ligand of the dependence receptors deleted in colorectal carcinoma and members of the UNC5H family. Netrin-1 is overexpressed in several aggressive cancers where it promotes cancer progression by inhibiting cell death induced by its receptors. Interference of its binding to its receptors has been shown, through the development of a monoclonal neutralizing antinetrin-1 antibody (currently in phase II of clinical trial), to actively induce apoptosis and tumor growth inhibition. The transcription factor p53 was shown to positively regulate netrin-1 gene expression. We show here that netrin-1 could be a target gene of the N-terminal p53 isoform Δ40p53, independent of full-length p53 activity. Using stable cell lines, harboring wild-type or null-p53, in which Δ40p53 expression could be finely tuned, we prove that Δ40p53 binds to and activates the netrin-1 promoter. In addition, we show that forcing immortalized human skeletal myoblasts to produce the Δ40p53 isoform, instead of full-length p53, leads to the up-regulation of netrin-1 and its receptor UNC5B and promotes cell survival. Indeed, we demonstrate that netrin-1 interference, in the presence of Δ40p53, triggers apoptosis in cancer and primary cells, leading to tumor growth inhibition in preclinical in vivo models. Finally, we show a positive correlation between netrin-1 and Δ40p53 gene expression in human melanoma and colorectal cancer biopsies. Hence, we propose that inhibition of netrin-1 binding to its receptors should be a promising therapeutic strategy in human tumors expressing high levels of Δ40p53.


Subject(s)
Carcinogenesis , Netrin Receptors/physiology , Netrin-1/physiology , Protein Isoforms/physiology , Tumor Suppressor Protein p53/physiology , Up-Regulation/physiology , Apoptosis/physiology , Cell Line, Tumor , Gene Silencing , Humans , Netrin-1/genetics , Promoter Regions, Genetic , Protein Binding
7.
Hepatology ; 76(5): 1345-1359, 2022 11.
Article in English | MEDLINE | ID: mdl-35253915

ABSTRACT

BACKGROUND AND AIMS: Netrin-1 displays protumoral properties, though the pathological contexts and processes involved in its induction remain understudied. The liver is a major model of inflammation-associated cancer development, leading to HCC. APPROACH AND RESULTS: A panel of cell biology and biochemistry approaches (reverse transcription quantitative polymerase chain reaction, reporter assays, run-on, polysome fractionation, cross linking immunoprecipitation, filter binding assay, subcellular fractionation, western blotting, immunoprecipitation, stable isotope labeling by amino acids in cell culture) on in vitro-grown primary hepatocytes, human liver cell lines, mouse samples and clinical samples was used. We identify netrin-1 as a hepatic inflammation-inducible factor and decipher its mode of activation through an exhaustive eliminative approach. We show that netrin-1 up-regulation relies on a hitherto unknown mode of induction, namely its exclusive translational activation. This process includes the transfer of NTN1 (netrin-1) mRNA to the endoplasmic reticulum and the direct interaction between the Staufen-1 protein and this transcript as well as netrin-1 mobilization from its cell-bound form. Finally, we explore the impact of a phase 2 clinical trial-tested humanized anti-netrin-1 antibody (NP137) in two distinct, toll-like receptor (TLR) 2/TLR3/TLR6-dependent, hepatic inflammatory mouse settings. We observe a clear anti-inflammatory activity indicating the proinflammatory impact of netrin-1 on several chemokines and Ly6C+ macrophages. CONCLUSIONS: These results identify netrin-1 as an inflammation-inducible factor in the liver through an atypical mechanism as well as its contribution to hepatic inflammation.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Humans , Animals , Toll-Like Receptor 2 , Nerve Growth Factors/metabolism , Toll-Like Receptor 3 , Toll-Like Receptor 6 , Tumor Suppressor Proteins/metabolism , Inflammation/metabolism , Anti-Inflammatory Agents , RNA, Messenger , Amino Acids , Netrin Receptors
8.
Nature ; 545(7654): 350-354, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28445456

ABSTRACT

Netrin-1 is an evolutionarily conserved, secreted extracellular matrix protein involved in axon guidance at the central nervous system midline. Netrin-1 is expressed by cells localized at the central nervous system midline, such as those of the floor plate in vertebrate embryos. Growth cone turning assays and three-dimensional gel diffusion assays have shown that netrin-1 can attract commissural axons. Loss-of-function experiments further demonstrated that commissural axon extension to the midline is severely impaired in the absence of netrin-1 (refs 3, 7, 8, 9). Together, these data have long supported a model in which commissural axons are attracted by a netrin-1 gradient diffusing from the midline. Here we selectively ablate netrin-1 expression in floor-plate cells using a Ntn1 conditional knockout mouse line. We find that hindbrain and spinal cord commissural axons develop normally in the absence of floor-plate-derived netrin-1. Furthermore, we show that netrin-1 is highly expressed by cells in the ventricular zone, which can release netrin-1 at the pial surface where it binds to commissural axons. Notably, Ntn1 deletion from the ventricular zone phenocopies commissural axon guidance defects previously described in Ntn1-knockout mice. These results show that the classical view that attraction of commissural axons is mediated by a gradient of floor-plate-derived netrin-1 is inaccurate and that netrin-1 primarily acts locally by promoting growth cone adhesion.


Subject(s)
Axon Guidance , Growth Cones/metabolism , Nerve Growth Factors/metabolism , Spinal Cord/cytology , Tumor Suppressor Proteins/metabolism , Animals , Cell Adhesion , Female , Male , Mice , Mice, Knockout , Nerve Growth Factors/deficiency , Netrin-1 , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Tumor Suppressor Proteins/deficiency
9.
Development ; 145(2)2018 01 17.
Article in English | MEDLINE | ID: mdl-29343638

ABSTRACT

During the development of the central nervous system (CNS), only motor axons project into peripheral nerves. Little is known about the cellular and molecular mechanisms that control the development of a boundary at the CNS surface and prevent CNS neuron emigration from the neural tube. It has previously been shown that a subset of spinal cord commissural axons abnormally invades sensory nerves in Ntn1 hypomorphic embryos and Dcc knockouts. However, whether netrin 1 also plays a similar role in the brain is unknown. In the hindbrain, precerebellar neurons migrate tangentially under the pial surface, and their ventral migration is guided by netrin 1. Here, we show that pontine neurons and inferior olivary neurons, two types of precerebellar neurons, are not confined to the CNS in Ntn1 and Dcc mutant mice, but that they invade the trigeminal, auditory and vagus nerves. Using a Ntn1 conditional knockout, we show that netrin 1, which is released at the pial surface by ventricular zone progenitors is responsible for the CNS confinement of precerebellar neurons. We propose, that netrin 1 distribution sculpts the CNS boundary by keeping CNS neurons in netrin 1-rich domains.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Netrin-1/metabolism , Peripheral Nervous System/embryology , Peripheral Nervous System/metabolism , Animals , Cell Movement/genetics , Cell Movement/physiology , Central Nervous System/cytology , DCC Receptor/deficiency , DCC Receptor/genetics , DCC Receptor/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Netrin-1/deficiency , Netrin-1/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Peripheral Nervous System/cytology , Pregnancy
10.
PLoS Biol ; 16(5): e2002912, 2018 05.
Article in English | MEDLINE | ID: mdl-29750782

ABSTRACT

The neurotrophin-3 (NT-3) receptor tropomyosin receptor kinase C (TrkC/NTRK3) has been described as a dependence receptor and, as such, triggers apoptosis in the absence of its ligand NT-3. This proapoptotic activity has been proposed to confer a tumor suppressor activity to this classic tyrosine kinase receptor (RTK). By investigating interacting partners that might facilitate TrkC-induced cell death, we have identified the basic helix-loop-helix (bHLH) transcription factor Hey1 and importin-α3 (karyopherin alpha 4 [KPNA4]) as direct interactors of TrkC intracellular domain, and we show that Hey1 is required for TrkC-induced apoptosis. We propose here that the cleaved proapoptotic portion of TrkC intracellular domain (called TrkC killer-fragment [TrkC-KF]) is translocated to the nucleus by importins and interacts there with Hey1. We also demonstrate that Hey1 and TrkC-KF transcriptionally silence mouse double minute 2 homolog (MDM2), thus contributing to p53 stabilization. p53 transcriptionally regulates the expression of TrkC-KF cytoplasmic and mitochondrial interactors cofactor of breast cancer 1 (COBRA1) and B cell lymphoma 2-associated X (BAX), which will subsequently trigger the intrinsic pathway of apoptosis. Of interest, TrkC was proposed to constrain tumor progression in neuroblastoma (NB), and we demonstrate in an avian model that TrkC tumor suppressor activity requires Hey1 and p53.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Receptor, trkC/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Chick Embryo , Chromatin/metabolism , Gene Expression Regulation , HCT116 Cells , HEK293 Cells , Humans , Karyopherins/metabolism , Mice
11.
Mol Cell ; 51(5): 632-46, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034695

ABSTRACT

The neurotrophin receptor TrkC was recently identified as a dependence receptor, and, as such, it triggers apoptosis in the absence of its ligand, NT-3. The molecular mechanism for apoptotic engagement involves the double cleavage of the receptor's intracellular domain, leading to the formation of a proapoptotic "killer" fragment (TrkC KF). Here, we show that TrkC KF interacts with Cobra1, a putative cofactor of BRCA1, and that Cobra1 is required for TrkC-induced apoptosis. We also show that, in the developing chick neural tube, NT-3 silencing is associated with neuroepithelial cell death that is rescued by Cobra1 silencing. Cobra1 shuttles TrkC KF to the mitochondria, where it promotes Bax activation, cytochrome c release, and apoptosome-dependent apoptosis. Thus, we propose that, in the absence of NT-3, the proteolytic cleavage of TrkC leads to the release of a killer fragment that triggers mitochondria-dependent apoptosis via the recruitment of Cobra1.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Nuclear Proteins/metabolism , Receptor, trkC/metabolism , Animals , Chick Embryo/metabolism , Cytochromes c/metabolism , Cytosol/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Neurons/metabolism , Neurotrophin 3/metabolism , Neurotrophin 3/pharmacology , Nuclear Proteins/genetics , Peptide Fragments/metabolism , RNA-Binding Proteins , Receptor, trkC/genetics , bcl-2-Associated X Protein/metabolism
12.
PLoS Genet ; 13(9): e1007024, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28945745

ABSTRACT

The importance of regulated necrosis in pathologies such as cerebral stroke and myocardial infarction is now fully recognized. However, the physiological relevance of regulated necrosis remains unclear. Here, we report a conserved role for p53 in regulating necrosis in Drosophila and mammalian spermatogenesis. We found that Drosophila p53 is required for the programmed necrosis that occurs spontaneously in mitotic germ cells during spermatogenesis. This form of necrosis involved an atypical function of the initiator caspase Dronc/Caspase 9, independent of its catalytic activity. Prevention of p53-dependent necrosis resulted in testicular hyperplasia, which was reversed by restoring necrosis in spermatogonia. In mouse testes, p53 was required for heat-induced germ cell necrosis, indicating that regulation of necrosis is a primordial function of p53 conserved from invertebrates to vertebrates. Drosophila and mouse spermatogenesis will thus be useful models to identify inducers of necrosis to treat cancers that are refractory to apoptosis.


Subject(s)
Necrosis/genetics , Spermatogenesis/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/genetics , Caspase 9/genetics , Caspases/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Germ Cells/growth & development , Germ Cells/pathology , Homeostasis/genetics , Humans , Hyperplasia/genetics , Hyperplasia/pathology , Male , Mice , Necrosis/pathology , Testis/growth & development , Testis/metabolism
13.
Dev Biol ; 442(2): 249-261, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30071216

ABSTRACT

The development of the sensory nervous system is the result of fine-tuned waves of neurogenesis and apoptosis which control the appropriate number of precursors and newly generated neurons and orient them toward a specific lineage. Neurotrophins and their tyrosine-kinase receptors (RTK) orchestrate this process. They have long been in the scope of the neurotrophic theory which established that a neuron is committed to die unless a trophic factor generated by its target provides it with a survival signal. The neural death has thus always been described as a "default" program, survival being the major player to control the number of cells. New insights have been brought by the gain of function studies which recently demonstrated that TrkC (NTRK3) is a "dependence receptor" able to actively trigger apoptosis in absence of its ligand NT-3. In order to address the role of TrkC pro-apoptotic activity in the control of sensory neurons number, we generated a TrkC gene-trap mutant mice. We found out that this new murine model recapitulates the sensory phenotype of TrkC constitutive mutants, with reduced DRG size and reduced number of DRG neurons. We engineered these mice strain with a lacZ reporter in order to follow the fate of neurons committed to a TrkC lineage and observed that they are specifically protected from NT-3 mediated apoptosis in NT-3/TrkC double knock-out embryos. Finally, using a chicken model we demonstrated that silencing NT-3 emanating from the ventral neural tube induced apoptosis in the DRG anlage. This apoptosis was inhibited by silencing TrkC. This work thus demonstrates that, during in vivo DRG development, TrkC behaves as a two-sided receptor transducing positive signals of neuronal survival in response to NT-3, but actively inducing neuronal cell death when unbound. This functional duality sets adequate number of neurons committed to a TrkC identity in the forming DRG.


Subject(s)
Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Receptor, trkC/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Animals , Apoptosis/physiology , Cell Line , Cell Survival/physiology , Chick Embryo , Female , Ganglia, Spinal/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism
14.
PLoS Biol ; 14(3): e1002421, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27031829

ABSTRACT

Hepatitis C virus (HCV) is an oncogenic virus associated with the onset of hepatocellular carcinoma (HCC). The present study investigated the possible link between HCV infection and Netrin-1, a ligand for dependence receptors that sustains tumorigenesis, in particular in inflammation-associated tumors. We show that Netrin-1 expression is significantly elevated in HCV+ liver biopsies compared to hepatitis B virus (HBV+) and uninfected samples. Furthermore, Netrin-1 was upregulated in all histological stages of HCV+ hepatic lesions, from minimal liver fibrosis to cirrhosis and HCC, compared to histologically matched HCV- tissues. Both cirrhosis and HCV contributed to the induction of Netrin-1 expression, whereas anti-HCV treatment resulted in a reduction of Netrin-1 expression. In vitro, HCV increased the level and translation of Netrin-1 in a NS5A-La-related protein 1 (LARP1)-dependent fashion. Knockdown and forced expression experiments identified the receptor uncoordinated receptor-5 (UNC5A) as an antagonist of the Netrin-1 signal, though it did not affect the death of HCV-infected cells. Netrin-1 enhanced infectivity of HCV particles and promoted viral entry by increasing the activation and decreasing the recycling of the epidermal growth factor receptor (EGFR), a protein that is dysregulated in HCC. Netrin-1 and HCV are, therefore, reciprocal inducers in vitro and in patients, as seen from the increase in viral morphogenesis and viral entry, both phenomena converging toward an increase in the level of infectivity of HCV virions. This functional association involving a cancer-related virus and Netrin-1 argues for evaluating the implication of UNC5 receptor ligands in other oncogenic microbial species.


Subject(s)
ErbB Receptors/metabolism , Hepatitis C/metabolism , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Autoantigens/metabolism , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Transformation, Neoplastic , Hepatitis C/complications , Hepatitis C/virology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/virology , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Netrin-1 , Ribonucleoproteins/metabolism , Up-Regulation , Viral Nonstructural Proteins/metabolism , Virus Internalization , SS-B Antigen
15.
Biochem Biophys Res Commun ; 495(4): 2425-2431, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29277614

ABSTRACT

In the liver, HBV and HCV infections, exposure to toxics, genetic and metabolic disorders may induce endoplasmic reticulum (ER) stress and the unfolding protein response (UPR). The UPR allows cells to reach ER homeostasis after lumen overload, but also fosters survival of damaged cells and therefore HCC onset. Dependence receptors such as UNC5A trigger apoptosis when unbound to their ligands. We have previously shown that the main dependence receptor ligand, netrin-1, could protect cells against UPR-induced apoptosis through sustained translation. In this study, we show that UNC5A is cumulatively downregulated by the UPR at the transcriptional level in vitro and at the translational level both in vitro and in vivo. We have found that the 5'-untranslated region of the UNC5A mRNA shares a certain homology degree with that of netrin-1, suggesting linked translational regulatory mechanisms, at least during the initial stages of the UPR. RNAi and forced expression studies identified UNC5A as a modulator of cell death in the context of the UPR. UNC5A decrease of association with polysomes and expression oriented cells towards UPR-associated hepatocytic survival. Such data indicate that cooperation between the UPR and UNC5A depletion as previously observed by ourselves in HCC patients samples may foster liver cancer development and growth.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Netrin-1/genetics , Receptors, Cell Surface/genetics , Unfolded Protein Response/genetics , Apoptosis/genetics , Carcinogenesis , Cell Line, Tumor , Epigenetic Repression/genetics , Humans , Netrin Receptors
16.
Mol Cell ; 40(6): 863-76, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21172653

ABSTRACT

The UNC5H dependence receptors promote apoptosis in the absence of their ligand, netrin-1, and this is important for neuronal and vascular development and for limitation of cancer progression. UNC5H2 (also called UNC5B) triggers cell death through the activation of the serine-threonine protein kinase DAPk. While performing a siRNA screen to identify genes implicated in UNC5H-induced apoptosis, we identified the structural subunit PR65ß of the holoenzyme protein phosphatase 2A (PP2A). We show that UNC5H2/B recruits a protein complex that includes PR65ß and DAPk and retains PP2A activity. PP2A activity is required for UNC5H2/B-induced apoptosis, since it activates DAPk by triggering its dephosphorylation. Moreover, netrin-1 binding to UNC5H2/B prevents this effect through interaction of the PP2A inhibitor CIP2A to UNC5H2/B. Thus we show here that, in the absence of netrin-1, recruitment of PP2A to UNC5H2/B allows the activation of DAPk via a PP2A-mediated dephosphorylation and that this mechanism is involved in angiogenesis regulation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , Receptors, Cell Surface/metabolism , Death-Associated Protein Kinases , Humans , Netrin Receptors , Phosphorylation , Tumor Cells, Cultured
17.
Biochem Biophys Res Commun ; 488(3): 466-470, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28501620

ABSTRACT

Alzheimer's disease is a growing concern in the context of the increasing lifespan of the populations. The work presented here is part of the fight against this threat. It supports a therapeutic approach to reduce the incidence of Alzheimer's disease, taking advantage of the specific binding of several domains of Netrin-1 to the ß-amyloid precursor protein. This basic knowledge shall then be used to predict, design or characterize lead compounds that may in turn inhibit/delay Alzheimer's disease's progression, extending the therapeutic offer of the other leads already being investigated in this line. The present work is focused on the interaction of the various portions of APP with the three domains of Netrin-1, the so-called LamNT, EGF-like and NTR domains respectively. It reveals in detail which portions of APP and Netrin-1 are specifically involved in these interactions, using ELISA technique in combination with protein-protein binding simulations. So far unsuspected interaction sites located in Netrin-1 EGF-like and NTR domains open possibilities for new therapeutic approaches in which these sites will be specifically targeted.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Nerve Growth Factors/metabolism , Plaque, Amyloid/metabolism , Tumor Suppressor Proteins/metabolism , Amyloid beta-Protein Precursor/chemistry , Humans , Models, Molecular , Nerve Growth Factors/chemistry , Netrin-1 , Protein Binding , Tumor Suppressor Proteins/chemistry
18.
Nat Mater ; 20(6): 722-723, 2021 06.
Article in English | MEDLINE | ID: mdl-34045700
19.
Nature ; 482(7386): 534-7, 2011 Dec 11.
Article in English | MEDLINE | ID: mdl-22158121

ABSTRACT

The role of deleted in colorectal carcinoma (DCC) as a tumour suppressor has been a matter of debate for the past 15 years. DCC gene expression is lost or markedly reduced in the majority of advanced colorectal cancers and, by functioning as a dependence receptor, DCC has been shown to induce apoptosis unless engaged by its ligand, netrin-1 (ref. 2). However, so far no animal model has supported the view that the DCC loss-of-function is causally implicated as predisposing to aggressive cancer development. To investigate the role of DCC-induced apoptosis in the control of tumour progression, here we created a mouse model in which the pro-apoptotic activity of DCC is genetically silenced. Although the loss of DCC-induced apoptosis in this mouse model is not associated with a major disorganization of the intestines, it leads to spontaneous intestinal neoplasia at a relatively low frequency. Loss of DCC-induced apoptosis is also associated with an increase in the number and aggressiveness of intestinal tumours in a predisposing APC mutant context, resulting in the development of highly invasive adenocarcinomas. These results demonstrate that DCC functions as a tumour suppressor via its ability to trigger tumour cell apoptosis.


Subject(s)
Disease Progression , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis/genetics , Caspases/metabolism , Cells, Cultured , DCC Receptor , Disease Models, Animal , Fibroblasts , Gene Silencing , Genes, APC , HEK293 Cells , Humans , Intestinal Neoplasms/metabolism , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Netrin-1 , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
20.
Nucleic Acids Res ; 43(12): 5838-54, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26007656

ABSTRACT

DNA methylation is thought to induce transcriptional silencing through the combination of two mechanisms: the repulsion of transcriptional activators unable to bind their target sites when methylated, and the recruitment of transcriptional repressors with specific affinity for methylated DNA. The Methyl CpG Binding Domain proteins MeCP2, MBD1 and MBD2 belong to the latter category. Here, we present MBD2 ChIPseq data obtained from the endogenous MBD2 in an isogenic cellular model of oncogenic transformation of human mammary cells. In immortalized (HMEC-hTERT) or transformed (HMLER) cells, MBD2 was found in a large proportion of methylated regions and associated with transcriptional silencing. A redistribution of MBD2 on methylated DNA occurred during oncogenic transformation, frequently independently of local DNA methylation changes. Genes downregulated during HMEC-hTERT transformation preferentially gained MBD2 on their promoter. Furthermore, depletion of MBD2 induced an upregulation of MBD2-bound genes methylated at their promoter regions, in HMLER cells. Among the 3,160 genes downregulated in transformed cells, 380 genes were methylated at their promoter regions in both cell lines, specifically associated by MBD2 in HMLER cells, and upregulated upon MBD2 depletion in HMLER. The transcriptional MBD2-dependent downregulation occurring during oncogenic transformation was also observed in two additional models of mammary cell transformation. Thus, the dynamics of MBD2 deposition across methylated DNA regions was associated with the oncogenic transformation of human mammary cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Methylation , DNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Binding Sites , Breast/cytology , Cell Line , Cell Line, Transformed , Down-Regulation , Epithelial Cells/metabolism , Female , Homeodomain Proteins/metabolism , Humans , Phenotype , Telomerase/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL