Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Med Oncol ; 39(1): 4, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34739635

ABSTRACT

Immunohistochemistry and recent molecular technologies progressively guided access to personalized anti-tumoral therapies. We explored the feasibility, efficacy, and the impact of molecular profiling in patients with advanced brain tumors. This multicentric prospective trial ProfiLER enrolled patients with primary brain tumors, who have been pre-treated with at least one line of anti-cancer treatment, and for whom molecular profiles had been achieved using next-generation sequencing and/or comparative genomic hybridization on fresh or archived samples from tumor, relapse, or biopsies. A molecular tumor board weekly analyzed results and proposed molecular-based recommended therapy (MBRT). From February 2013 to December 2015, we enrolled 141 patients with primary brain tumor and analyzed 105 patients for whom tumor genomic profiles had been achieved. Histology mainly identified glioblastoma (N = 46, 44%), low-grade glioma (N = 26, 25%), high-grade glioma (N = 12, 11%), and atypical and anaplastic meningioma (N = 8, 8%). Forty-three (41%) patients presented at least one actionable molecular alteration. Out of 61 alterations identified, the most frequent alterations occurred in CDKN2A (N = 18), EGFR (N = 12), PDGFRa (N = 8), PTEN (N = 8), CDK4 (N = 7), KIT (N = 6), PIK3CA (N = 5), and MDM2 (N = 3). Sixteen (15%) patients could not be proposed for a MBRT due to early death (N = 5), lack of available clinical trials (N = 9), or inappropriate results (N = 2). Only six (6%) of the 27 (26%) patients for whom a MBRT had been proposed finally initiated MBRT (everolimus (N = 3), erlotinib (N = 1), ruxolitinib (N = 1), and sorafenib (N = 1)), but discontinued treatment for toxicity (N = 4) or clinical progression (N = 2). High-throughput sequencing in patients with brain tumors may be routinely performed, especially when macroscopic surgery samples are available; nevertheless delays should be reduced. Criteria for clinical trial enrollment should be reconsidered in patients with brain tumors, and a panel of genes specifically dedicated to neurologic tumors should be developed to help decision-making in clinical practice.


Subject(s)
Brain Neoplasms , Clinical Decision-Making , Precision Medicine/methods , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Comparative Genomic Hybridization , Female , Genomics , Humans , Male , Middle Aged , Prospective Studies , Young Adult
2.
JCO Precis Oncol ; 4: 785-795, 2020 Nov.
Article in English | MEDLINE | ID: mdl-35050753

ABSTRACT

PURPOSE: The Program to Establish the Genetic and Immunologic Profile of Patient's Tumor for All Types of Advanced Cancer study (ClinicalTrials.gov identifier: NCT01774409) analyzed the genome of refractory cancers to identify a potential molecular-based recommended therapy (MBRT). The objectives of the pediatric substudy were to describe the incidence of genomic mutations, the MBRT, and the treatments undertaken with a molecular-targeted agent in a pediatric cohort. METHODS: The tumor genome was analyzed within a 69-gene next-generation sequencing panel and an array comparative genomic hybridization assay. The results were evaluated by a multidisciplinary molecular board, and the targeted therapies were provided in the setting of a clinical trial or through compassionate use programs, when indicated. RESULTS: Between November 2013 and June 2017, 50 patients younger than 19 years who were treated for a high-risk or relapsing tumor were included. Sarcomas (n = 24; 47%), CNS tumors (n = 14; 29%), and neuroblastomas (n = 5; 10%) were the most frequent tumor subtypes. Seven patients (14%) were excluded because no DNA could be recovered. Among the 43 remaining patients, 10 exhibited at least one targetable genomic alteration. Ultimately, four patients (8%) were treated with the recommended targeted therapy. CONCLUSION: The results of this study confirm treatment with a targeted therapy for pediatric patients with cancer is still limited at present, as also is reported for adults.

3.
J Alzheimers Dis ; 52(1): 223-42, 2016 03 08.
Article in English | MEDLINE | ID: mdl-27060954

ABSTRACT

Recent studies have shown that inoculation of susceptible mice with amyloid-ß (Aß) peptides accelerates Aß deposition in the brain, supporting the idea that Aß may be self-amplifying; however, the exact mechanism is not understood. Here we provide evidence that Aß may self-amplify, in part, by inhibiting α-secretase ADAM10 (a disintegrin and metalloprotease) cleavage of full-length Aß precursor protein (FL AßPP) and therefore allow greater ß-secretase processing, and that Aß itself is a substrate for ADAM10. Exposure of primary neuronal cultures from PDAßPP mice to exogenous rat Aß1- 40 resulted in increased de novo human Aß1-42 production and exposure of cells to Aß decreased production of ADAM10 cleavage product soluble AßPPα (sAßPPα). In a cell-free assay, Aß decreased ADAM10 cleavage of the chimeric substrate MBP-AßPPC125 and Aß itself was apparently cleaved by the enzyme. The axonal guidance and trophic factor netrin-1, however, reduced the Aß1- 40-induced Aß1-42 increase, increased sAßPPα, and reversed the Aß-induced sAßPPα decrease in vitro. In vivo, induction of netrin-1 expression in PDAßPPSwe/Ind transgenic mice resulted in reductions in both Aß1-42 and Aß1- 40, and ICV delivery of netrin-1 to PDAßPPSwe/Ind mice increased sAßPPα, decreased Aß, and improved working memory. Finally, to support further study of netrin-1's potential as a therapeutic for Alzheimer's disease, pilot gene therapy studies were performed and a netrin mimetic peptide synthesized and tested that, like netrin, can increase sAßPPα and decrease Aß1-42in vitro. Taken together, these data provide mechanistic insights into Aß self-amplification and the ability of netrin-1 to disrupt it.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/metabolism , Genetic Therapy/methods , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , ADAM10 Protein/metabolism , Alzheimer Disease/psychology , Animals , Biomimetics , Cell Line, Tumor , Cognition/physiology , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Humans , Memory, Short-Term/physiology , Mice, Transgenic , Netrin-1 , Pilot Projects , Rats , Recognition, Psychology/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
PLoS One ; 8(8): e70545, 2013.
Article in English | MEDLINE | ID: mdl-23950959

ABSTRACT

Several human small heat shock proteins (sHsps) are phosphorylated oligomeric chaperones that enhance stress resistance. They are characterized by their ability to interact and form polydispersed hetero-oligomeric complexes. We have analyzed the cellular consequences of the stable expression of either wild type HspB5 or its cataracts and myopathies inducing R120G mutant in growing and oxidative stress treated HeLa cells that originally express only HspB1. Here, we describe that wild type and mutant HspB5 induce drastic and opposite effects on cell morphology and oxidative stress resistance. The cellular distribution and phosphorylation of these polypeptides as well as the oligomerization profile of the resulting hetero-oligomeric complexes formed by HspB1 with the two types of exogenous polypeptides revealed the dominant effects induced by HspB5 polypeptides towards HspB1. The R120G mutation enhanced the native size and salt resistance of HspB1-HspB5 complex. However, in oxidative conditions the interaction between HspB1 and mutant HspB5 was drastically modified resulting in the aggregation of both partners. The mutation also induced the redistribution of HspB1 phosphorylated at serine 15, originally observed at the level of the small oligomers that do not interact with wild type HspB5, to the large oligomeric complex formed with mutant HspB5. This phosphorylation stabilized the interaction of HspB1 with mutant HspB5. A dominant negative effect towards HspB1 appears therefore as an important event in the cellular sensitivity to oxidative stress mediated by mutated HspB5 expression. These observations provide novel data that describe how a mutated sHsp can alter the protective activity of another member of this family of chaperones.


Subject(s)
HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Mutation , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism , Gene Expression , HSP27 Heat-Shock Proteins/chemistry , HeLa Cells , Humans , Oxidative Stress/genetics , Phosphorylation , Protein Binding , Protein Multimerization , Protein Transport , alpha-Crystallin B Chain/chemistry
5.
J Biol Chem ; 287(35): 30014-23, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22782894

ABSTRACT

The multifunctional protein netrin-1 was initially discovered as the main attractive cue for commissural axon guidance by acting through its receptor DCC. Recently, we have shown that netrin-1 also interacts with the orphan transmembrane receptor amyloid precursor protein (APP). APP is cleaved by proteases, generating amyloid-ß peptide, the main component of the amyloid plaques that are associated with Alzheimer disease. Our previous work demonstrated that via its interaction with APP, netrin-1 is a negative regulator of amyloid-ß production in adult brain, but the biological relevance of APP/netrin-1 interaction under non-pathological conditions was unknown. We show here that during commissural axon navigation, APP, expressed at the growth cone, is part of the DCC receptor complex mediating netrin-1-dependent axon guidance. APP interacts with DCC in the presence of netrin-1 and enhances netrin-1-mediated DCC intracellular signaling, such as MAPK activation. Inactivation of APP in mice is associated with reduced commissural axon outgrowth. Thus, APP functionally acts as a co-receptor for DCC to mediate axon guidance.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Growth Cones/metabolism , Nerve Growth Factors/metabolism , Proteolysis , Tumor Suppressor Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , DCC Receptor , Growth Cones/pathology , Mice , Mice, Mutant Strains , Nerve Growth Factors/genetics , Netrin-1 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/genetics
6.
Nat Cell Biol ; 11(6): 739-46, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19465923

ABSTRACT

Sonic hedgehog (Shh) and its main receptor, Patched (Ptc), are implicated in both neural development and tumorigenesis. Besides its classic morphogenic activity, Shh is also a survival factor. Along this line, Ptc has been shown to function as a dependence receptor; it induces apoptosis in the absence of Shh, whereas its pro-apoptotic activity is blocked in the presence of Shh. Here we show that, in the absence of its ligand, Ptc interacts with the adaptor protein DRAL (downregulated in rhabdomyosarcoma LIM-domain protein; also known as FHL2). DRAL is required for the pro-apoptotic activity of Ptc both in immortalized cells and during neural tube development in chick embryos. We demonstrate that, in the absence of Shh, Ptc recruits a protein complex that includes DRAL, one of the caspase recruitment (CARD)-domain containing proteins TUCAN (family member, 8) or NALP1 (NLR family, pyrin domain containing 1) and apical caspase-9. Ptc triggers caspase-9 activation and enhances cell death through a caspase-9-dependent mechanism. Thus, we propose that in the absence of its ligand Shh the dependence receptor Ptc serves as the anchor for a caspase-activating complex that includes DRAL, and caspase-9.


Subject(s)
Apoptosis/physiology , Caspase 9/metabolism , Hedgehog Proteins/metabolism , Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Cell Line , Chick Embryo , Hedgehog Proteins/genetics , Homeodomain Proteins/genetics , Humans , LIM-Homeodomain Proteins , Multiprotein Complexes/metabolism , Muscle Proteins/genetics , NLR Proteins , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Patched Receptors , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Two-Hybrid System Techniques
7.
Proc Natl Acad Sci U S A ; 105(38): 14465-70, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18796601

ABSTRACT

DCC (Deleted in Colorectal Cancer) is a putative tumor suppressor whose expression is lost in numerous cancers and whose tumor suppressor activity appears to be dependent on its ability to trigger apoptosis when disengaged by its ligand netrin-1. In this sense, netrin-1 is a survival factor that controls tumorigenesis. However, netrin-1 is also the prototypical axon guidance cue and has been shown to orient many neurons or axons, especially commissural axons, during spinal cord development. Here we show that netrin-1 is not only an attractive cue for developing commissural axons but also promotes their survival. In primary neuronal culture, in mice or in chick embryos, netrin-1 inhibits the proapoptotic activity of DCC in developing commissural neurons. Thus, adequate commissural neurons navigation requires both the attractive activity of netrin-1 and the anti-apoptotic function of this cue.


Subject(s)
Cell Survival/genetics , Nerve Growth Factors/metabolism , Neurons/physiology , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/physiology , Cell Line , Cells, Cultured , Chick Embryo , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Genes, DCC/genetics , Humans , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Netrin-1 , Neural Tube/embryology , Rats , Spinal Cord/embryology , Tumor Suppressor Proteins/genetics
8.
Proc Natl Acad Sci U S A ; 103(11): 4128-33, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16537496

ABSTRACT

DCC (deleted in colorectal cancer) is a putative tumor suppressor gene whose expression is lost in numerous cancers. DCC also encodes the main receptor for the neuronal navigation cue netrin-1. It has been shown that DCC belongs to the so-called family of dependence receptors. Such receptors induce apoptosis when their ligand is absent, thus conferring a state of cellular dependence on ligand availability. We recently proposed that DCC is a tumor suppressor because it induces the death of tumor cells that grow in settings of ligand unavailability. Moreover, it seems that the DCC/netrin-1 pair may also regulate neuron survival during nervous system development. However, the mechanisms by which DCC triggers cell death are still unknown. We show here that the localization of DCC to lipid rafts is a prerequisite for its proapoptotic activity, both in immortalized cells and in primary neurons. The presence of DCC in lipid rafts probably allows the formation of an adequate submembrane complex, because the interaction of caspase-9 with DCC is inhibited by the disorganization of lipid rafts. Thus, dependence receptors may require lipid raft localization for cell death signaling.


Subject(s)
Apoptosis/physiology , Membrane Microdomains/metabolism , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Caspase 9 , Caspases/metabolism , Cell Line , Cells, Cultured , DCC Receptor , Humans , Nerve Growth Factors/metabolism , Netrin-1 , Neurons/cytology , Neurons/metabolism , Rats , Receptors, Cell Surface/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Tumor Suppressor Proteins/genetics
9.
J Cell Sci ; 118(Pt 8): 1687-92, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15811950

ABSTRACT

During development, axons migrate long distances in responses to attractive or repulsive signals that are detected by their growth cones. One of these signals is mediated by netrin-1, a diffusible laminin-related molecule that both attracts and repels growth cones via interaction with its receptor DCC (deleted in colorectal cancer). Here we show that DCC in both commissural neurons and immortalized cells, is partially associated with cholesterol- and sphingolipid-enriched membrane domains named lipid rafts. This localization of DCC in lipid rafts is mediated by the palmitoylation within its transmembrane region. Moreover, this raft localization of DCC is required for netrin-1-induced DCC-dependent ERK activation, and netrin-1-mediated axon outgrowth requires lipid raft integrity. Thus, the presence of axon guidance-related receptors in lipid rafts appears to be a crucial pre-requisite for growth cone response to chemo-attractive or repulsive cues.


Subject(s)
Cell Adhesion Molecules/metabolism , Chemotaxis/physiology , Growth Cones/metabolism , Membrane Microdomains/metabolism , Nerve Growth Factors/metabolism , Spinal Cord/embryology , Tumor Suppressor Proteins/metabolism , Animals , Cell Communication/physiology , Cell Line , Cells, Cultured , Cues , DCC Receptor , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Membrane Lipids/metabolism , Netrin-1 , Palmitic Acid/metabolism , Rats , Receptors, Cell Surface , Signal Transduction/physiology
10.
Nature ; 417(6887): 443-7, 2002 May 23.
Article in English | MEDLINE | ID: mdl-11986622

ABSTRACT

Neuronal growth cones are guided to their targets by attractive and repulsive guidance cues. In mammals, netrin-1 is a bifunctional cue, attracting some axons and repelling others. Deleted in colorectal cancer (Dcc) is a receptor for netrin-1 that mediates its chemoattractive effect on commissural axons, but the signalling mechanisms that transduce this effect are poorly understood. Here we show that Dcc activates mitogen-activated protein kinase (MAPK) signalling, by means of extracellular signal-regulated kinase (ERK)-1 and -2, on netrin-1 binding in both transfected cells and commissural neurons. This activation is associated with recruitment of ERK-1/2 to a Dcc receptor complex. Inhibition of ERK-1/2 antagonizes netrin-dependent axon outgrowth and orientation. Thus, activation of MAPK signalling through Dcc contributes to netrin signalling in axon growth and guidance.


Subject(s)
Cell Adhesion Molecules/metabolism , DNA-Binding Proteins , Growth Cones/drug effects , Growth Cones/enzymology , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/pharmacology , Transcription Factors , Tumor Suppressor Proteins/metabolism , 3T3 Cells , Animals , Cell Adhesion Molecules/genetics , Cell Line , Cell Size/drug effects , Culture Techniques , DCC Receptor , Enzyme Activation/drug effects , Genes, Reporter , Growth Cones/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Nerve Growth Factors/metabolism , Netrin-1 , Precipitin Tests , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface , Signal Transduction/drug effects , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/growth & development , Tumor Suppressor Proteins/genetics , Two-Hybrid System Techniques , Xenopus , ets-Domain Protein Elk-1
SELECTION OF CITATIONS
SEARCH DETAIL
...