Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Oncogene ; 37(5): 627-637, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28991230

ABSTRACT

Alu sequences are the most abundant short interspersed repeated elements in the human genome. Here we show that in a cell culture model of colorectal cancer (CRC) progression, we observe accumulation of Alu RNA that is associated with reduced DICER1 levels. Alu RNA induces epithelial-to-mesenchymal transition (EMT) by acting as a molecular sponge of miR-566. Moreover, Alu RNA accumulates as consequence of DICER1 deficit in colorectal, ovarian, renal and breast cancer cell lines. Interestingly, Alu RNA knockdown prevents DICER1 depletion-induced EMT despite global microRNA (miRNA) downregulation. Alu RNA expression is also induced by transforming growth factor-ß1, a major driver of EMT. Corroborating this data, we found that non-coding Alu RNA significantly correlates with tumor progression in human CRC patients. Together, these findings reveal an unexpected DICER1-dependent, miRNA-independent role of Alu RNA in cancer progression that could bring mobile element transcripts in the fields of cancer therapeutic and prognosis.


Subject(s)
Alu Elements/genetics , Colorectal Neoplasms/genetics , DEAD-box RNA Helicases/metabolism , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Ribonuclease III/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colon/pathology , Colorectal Neoplasms/pathology , DEAD-box RNA Helicases/genetics , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/secondary , MicroRNAs/metabolism , RNA/metabolism , Ribonuclease III/genetics , Transforming Growth Factor beta1/metabolism
2.
Oncogenesis ; 6(10): e390, 2017 Oct 23.
Article in English | MEDLINE | ID: mdl-29058695

ABSTRACT

Breast cancer-associated fibroblasts (CAFs) have a crucial role in tumor initiation, metastasis and therapeutic resistance by secreting various growth factors, cytokines, protease and extracellular matrix components. Soluble factors secreted by CAFs are involved in many pathways including inflammation, metabolism, proliferation and epigenetic modulation, suggesting that CAF-dependent reprograming of cancer cells affects a large set of genes. This paracrine signaling has an important role in tumor progression, thus deciphering some of these processes could lead to relevant discoveries with subsequent clinical implications. Here, we investigated the mechanisms underlying the changes in gene expression patterns associated with the cross-talk between breast cancer cells and the stroma. From RNAseq data obtained from breast cancer cell lines grown in presence of CAF-secreted factors, we identified 372 upregulated genes, exhibiting an expression level positively correlated with the stromal content of breast cancer specimens. Furthermore, we observed that gene expression changes were not mediated through significant DNA methylation changes. Nevertheless, CAF-secreted factors but also stromal content of the tumors remarkably activated specific genes characterized by a DNA methylation pattern: hypermethylation at transcription start site and shore regions. Experimental approaches (inhibition of DNA methylation, knockdown of methyl-CpG-binding domain protein 2 and chromatin immunoprecipitation assays) indicated that this set of genes was epigenetically controlled. These data elucidate the importance of epigenetics marks in the cancer cell reprogramming induced by stromal cell and indicated that the interpreters of the DNA methylation signal have a major role in the response of the cancer cells to the microenvironment.

3.
Oncogene ; 36(48): 6712-6724, 2017 11 30.
Article in English | MEDLINE | ID: mdl-28783179

ABSTRACT

Hepatitis C virus (HCV) infection is a leading cause of hepatocellular carcinoma (HCC), mainly through cirrhosis induction, spurring research for a deeper understanding of HCV versus host interactions in cirrhosis. The present study investigated crosstalks between HCV infection and UNC5A, a netrin-1 dependence receptor that is inactivated in cancer. UNC5A and HCV parameters were monitored in patients samples (n=550) as well as in in vitro. In patients, UNC5A mRNA expression is significantly decreased in clinical HCV(+) specimens irrespective of the viral genotype, but not in (HBV)(+) liver biopsies, as compared to uninfected samples. UNC5A mRNA is downregulated in F2 (3-fold; P=0.009), in F3 (10-fold, P=0.0004) and more dramatically so in F4/cirrhosis (44-fold; P<0.0001) histological stages of HCV(+) hepatic lesions compared to histologically matched HCV(-) tissues. UNC5A transcript was found strongly downregulated in HCC samples (33-fold; P<0.0001) as compared with non-HCC samples. In vivo, association of UNC5A transcripts with polyribosomes is decreased by 50% in HCV(+) livers. Consistent results were obtained in vitro showing HCV-dependent depletion of UNC5A in HCV-infected hepatocyte-like cells and in primary human hepatocytes. Using luciferase reporter constructs, HCV cumulatively decreased UNC5A transcription from the UNC5 promoter and translation in a UNC5A 5'UTR-dependent manner. Proximity ligation assays, kinase assays, as well as knockdown and forced expression experiments identified UNC5A as capable of impeding autophagy and promoting HCV restriction through specific impact on virion infectivity, in a cell death-independent and DAPK-related manner. In conclusion, while the UNC5A dependence receptor counteracts HCV persistence through regulation of autophagy in a DAPK-dependent manner, it is dramatically decreased in all instances in HCC samples, and specifically by HCV in cirrhosis. Such data argue for the evaluation of the implication of UNC5A in liver carcinogenesis.


Subject(s)
Hepatitis C/metabolism , Receptors, Cell Surface/metabolism , Autophagy , Cell Line, Tumor , Gene Expression , Hepacivirus/physiology , Hepatocytes/physiology , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Netrin Receptors , Precancerous Conditions/metabolism , Receptors, Cell Surface/genetics , Virus Replication
4.
Cell Death Differ ; 23(3): 442-53, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26292756

ABSTRACT

While a great deal of progress has been made in understanding the molecular mechanisms that regulate retino-tectal mapping, the determinants that target retinal projections to specific layers of the optic tectum remain elusive. Here we show that two independent RGMa-peptides, C- and N-RGMa, activate two distinct intracellular pathways to regulate axonal growth. C-RGMa utilizes a Leukemia-associated RhoGEF (LARG)/Rho/Rock pathway to inhibit axonal growth. N-RGMa on the other hand relies on ϒ-secretase cleavage of the intracellular portion of Neogenin to generate an intracellular domain (NeICD) that uses LIM-only protein 4 (LMO4) to block growth. In the developing tectum (E18), overexpression of C-RGMa and dominant-negative LARG (LARG-PDZ) induced overshoots in the superficial tectal layer but not in deeper tectal layers. In younger embryos (E12), C-RGMa and LARG-PDZ prevented ectopic projections toward deeper tectal layers, indicating that C-RGMa may act as a barrier to descending axons. In contrast both N-RGMa and NeICD overexpression resulted in aberrant axonal-paths, all of which suggests that it is a repulsive guidance molecule. Thus, two RGMa fragments activate distinct pathways resulting in different axonal responses. These data reveal how retinal projections are targeted to the appropriate layer in their target tissue.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Nerve Tissue Proteins/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , Animals , Cell Enlargement , Chick Embryo , Organ Specificity , Retinal Ganglion Cells/physiology , Superior Colliculi/cytology , Superior Colliculi/enzymology , Tissue Culture Techniques
5.
Cell Death Dis ; 4: e871, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24136235

ABSTRACT

The receptor tyrosine kinase Met and its ligand, the hepatocyte growth factor, are essential to embryonic development, whereas the deregulation of Met signaling is associated with tumorigenesis. While ligand-activated Met promotes survival, caspase-dependent generation of the p40 Met fragment leads to apoptosis induction - hallmark of the dependence receptor. Although the survival signaling pathways induced by Met are well described, the pro-apoptotic signaling pathways are unknown. We show that, although p40 Met contains the entire kinase domain, it accelerates apoptosis independently of kinase activity. In cell cultures undergoing apoptosis, the fragment shows a mitochondrial localization, required for p40 Met-induced cell death. Fulminant hepatic failure induced in mice leads to the generation of p40 Met localized also in the mitochondria, demonstrating caspase cleavage of Met in vivo. According to its localization, the fragment induces mitochondrial permeabilization, which is inhibited by Bak silencing and Bcl-xL overexpression. Moreover, Met silencing delays mitochondrial permeabilization induced by an apoptotic treatment. Thus, the Met-dependence receptor in addition to its well-known role in survival signaling mediated by its kinase activity, also participates in the intrinsic apoptosis pathway through the generation of p40 Met - a caspase-dependent fragment of Met implicated in the mitochondrial permeabilization process.


Subject(s)
Apoptosis , Caspases/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Animals , Cell Survival , Cytochromes c/metabolism , Dogs , Epithelial Cells/enzymology , Gene Silencing , Humans , Ligands , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Permeability , Protein Transport , Subcellular Fractions/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
6.
Br J Cancer ; 107(1): 63-70, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22627320

ABSTRACT

BACKGROUND: The small stress heat shock protein 27 (Hsp27) has recently turned as a promising target for cancer treatment. Hsp27 upregulation is associated with tumour growth and resistance to chemo- and radio-therapeutic treatments, and several ongoing drugs inhibiting Hsp27 expression are under clinical trial. Hsp27 is now well described to counteract apoptosis and its elevated expression is associated with increased aggressiveness of several primary tumours. However, its role in the later stage of tumour progression and, more specifically, in the later and most deadly stage of tumour metastasis is still unclear. METHODS/RESULTS: In the present study, we showed by qRT-PCR that Hsp27 gene is overexpressed in a large fraction of the metastatic breast cancer area in 53 patients. We further analysed the role of this protein in mice during bone metastasis invasion and establishment by using Hsp27 genetically depleted MDA-MB231/B02 human breast cancer cell line as a model. We demonstrate that Hsp27 silencing led to reduced cell migration and invasion in vitro and that in vivo it correlated with a decreased ability of breast cancer cells to metastasise and grow in the skeleton. CONCLUSION: Altogether, these data characterised Hsp27 as a potent therapeutic target in breast cancer bone metastasis and skeletal tumour growth.


Subject(s)
Bone Neoplasms/prevention & control , Breast Neoplasms/genetics , HSP27 Heat-Shock Proteins/genetics , Animals , Apoptosis/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Targeting , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Small Interfering/pharmacology , Transfection , Transplantation, Heterologous
7.
Cell Death Differ ; 19(1): 107-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21760595

ABSTRACT

In 2009, the Nomenclature Committee on Cell Death (NCCD) proposed a set of recommendations for the definition of distinct cell death morphologies and for the appropriate use of cell death-related terminology, including 'apoptosis', 'necrosis' and 'mitotic catastrophe'. In view of the substantial progress in the biochemical and genetic exploration of cell death, time has come to switch from morphological to molecular definitions of cell death modalities. Here we propose a functional classification of cell death subroutines that applies to both in vitro and in vivo settings and includes extrinsic apoptosis, caspase-dependent or -independent intrinsic apoptosis, regulated necrosis, autophagic cell death and mitotic catastrophe. Moreover, we discuss the utility of expressions indicating additional cell death modalities. On the basis of the new, revised NCCD classification, cell death subroutines are defined by a series of precise, measurable biochemical features.


Subject(s)
Apoptosis , Autophagy , Cells/metabolism , Cells/pathology , Necrosis , Terminology as Topic , Animals , Caspases/metabolism , Humans , Mitosis
8.
Oncogene ; 29(13): 1865-82, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20173780

ABSTRACT

Dependence receptors (DRs) now form a family of more than a dozen membrane receptors that are not linked by their structure, but by common functional traits. The most notable is their ability to trigger two opposite signaling pathways: in the presence of ligand, these receptors activate classic signaling pathways implicated in cell survival, migration and differentiation. In the absence of ligand, they do not stay inactive, rather they elicit an apoptotic signal. Thus, cells expressing this kind of receptor are dependent on the presence of ligand in the extracellular environment to survive. This review will recapitulate the increasing data regarding the molecular mechanisms associated with DRs, their potential implication during development, as well as their deregulation during tumorigenesis and, finally, their emergence as new possible therapeutic targets for cancer treatment.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Cell Survival/physiology , Neoplasms/pathology , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Drug Delivery Systems , ErbB Receptors/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Ligands , Neoplasms/therapy , Phosphatidylinositol 3-Kinases/physiology , TNF-Related Apoptosis-Inducing Ligand/physiology
9.
Cell Death Differ ; 16(10): 1344-51, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19543238

ABSTRACT

Netrin-1 was recently proposed to control tumorigenesis by inhibiting apoptosis induced by the dependence receptors DCC (Deleted in colorectal cancer) and UNC5H. Although the loss of these dependence receptors' expression has been described as a selective advantage for tumor growth and progression in numerous cancers, recent observations have shown that some tumors may use an alternative strategy to block dependence receptor-induced programmed cell death: the autocrine expression of netrin-1. This alternative strategy has been observed in a large fraction of aggressive breast cancers, neuroblastoma, pancreatic adenocarcinoma, and lung cancer. This observation is of potential interest regarding future targeted therapy, as in such cases interfering with the ability of netrin-1 to inhibit DCC or UNC5H-induced cell death is associated with apoptosis of netrin-1-expressing tumor cells in vitro, and with inhibition of tumor growth or metastasis in different animal tumor models. The understanding of the mechanism by which netrin-1 inhibits cell death is therefore of interest. Here, we show that netrin-1 triggers the multimerization of both DCC and UNC5H2 receptors, and that multimerization of the intracellular domain of DCC and UNC5H2 is the critical step to inhibit the proapoptotic effects of both of these receptors. Taking advantage of this property, we utilized a recombinant specific domain of DCC that (i) interacts with netrin-1 and (ii) inhibits netrin-1-induced multimerization, to trigger apoptosis in netrin-dependent tumor cells.


Subject(s)
Apoptosis , Neoplasms/metabolism , Nerve Growth Factors/pharmacology , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/pharmacology , Animals , Cell Line , Chickens , DCC Receptor , Disease Models, Animal , Humans , Netrin Receptors , Netrin-1 , Protein Multimerization/drug effects , Recombinant Proteins/pharmacology , Tumor Suppressor Proteins/metabolism
10.
Cell Death Differ ; 16(8): 1093-107, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19373242

ABSTRACT

Cell death is essential for a plethora of physiological processes, and its deregulation characterizes numerous human diseases. Thus, the in-depth investigation of cell death and its mechanisms constitutes a formidable challenge for fundamental and applied biomedical research, and has tremendous implications for the development of novel therapeutic strategies. It is, therefore, of utmost importance to standardize the experimental procedures that identify dying and dead cells in cell cultures and/or in tissues, from model organisms and/or humans, in healthy and/or pathological scenarios. Thus far, dozens of methods have been proposed to quantify cell death-related parameters. However, no guidelines exist regarding their use and interpretation, and nobody has thoroughly annotated the experimental settings for which each of these techniques is most appropriate. Here, we provide a nonexhaustive comparison of methods to detect cell death with apoptotic or nonapoptotic morphologies, their advantages and pitfalls. These guidelines are intended for investigators who study cell death, as well as for reviewers who need to constructively critique scientific reports that deal with cellular demise. Given the difficulties in determining the exact number of cells that have passed the point-of-no-return of the signaling cascades leading to cell death, we emphasize the importance of performing multiple, methodologically unrelated assays to quantify dying and dead cells.


Subject(s)
Cell Death , Apoptosis , Eukaryotic Cells/cytology , Flow Cytometry , Guidelines as Topic , Humans , Immunoblotting , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Spectrometry, Fluorescence
11.
Cell Death Differ ; 16(5): 655-63, 2009 May.
Article in English | MEDLINE | ID: mdl-19148186

ABSTRACT

The beta-amyloid precursor protein (APP) is an orphan transmembrane receptor whose physiological role is largely unknown. APP is cleaved by proteases generating amyloid-beta (Abeta) peptide, the main component of the amyloid plaques that are associated with Alzheimer's disease. Here, we show that APP binds netrin-1, a multifunctional guidance and trophic factor. Netrin-1 binding modulates APP signaling triggering APP intracellular domain (AICD)-dependent gene transcription. Furthermore, netrin-1 binding suppresses Abeta peptide production in brain slices from Alzheimer model transgenic mice. In this mouse model, decreased netrin-1 expression is associated with increased Abeta concentration, thus supporting netrin-1 as a key regulator of Abeta production. Finally, we show that netrin-1 brain administration in Alzheimer model transgenic mice may be associated with an amelioration of the Alzheimer's phenotype.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Brain/pathology , Cell Line , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mice, Transgenic , Nerve Growth Factors/administration & dosage , Netrin-1 , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Transcription, Genetic , Transfection , Tumor Suppressor Proteins/administration & dosage
12.
Cell Mol Life Sci ; 62(22): 2599-616, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16158190

ABSTRACT

Netrin-1 has been shown to play a crucial role in neuronal navigation during nervous system development mainly through its interaction with its receptors DCC and UNC5H. However, initially the DCC (deleted in colorectal cancer) gene was proposed as a putative tumor suppressor gene. It was then difficult to reconcile the two activities of DCC until the observation that DCC belongs to an emerging family of receptors named dependence receptors. Such receptors share the property of inducing apoptosis in the absence of ligand, hence creating a cellular state of dependence on the ligand. Thus, netrin-1 may not only be a chemotropic factor for neurons but also a survival factor. We will review here the identification of netrin-1 and its receptors, the signaling pathways initiated in the presence or absence of netrin-1. We will suggest some possible roles of netrin-1 in nervous system development, neovascularisation, adhesion and tumorigenesis.


Subject(s)
Chemotactic Factors/physiology , Neoplasms/etiology , Neoplasms/metabolism , Nerve Growth Factors/physiology , Neurons/physiology , Tumor Suppressor Proteins/physiology , Animals , Humans , Neoplasms/pathology , Netrin-1
14.
Cell Death Differ ; 12(8): 1031-43, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16015380

ABSTRACT

Cells depend for their survival on stimulation by trophic factors and other prosurvival signals, the withdrawal of which induces apoptosis, both via the loss of antiapoptotic signaling and the activation of proapoptotic signaling via specific receptors. These receptors, dubbed dependence receptors, activate apoptotic pathways following the withdrawal of trophic factors and other supportive stimuli. Such receptors may feature in developmental cell death, carcinogenesis (including metastasis), neurodegeneration, and possibly subapoptotic events such as neurite retraction and somal atrophy. Mechanistic studies of dependence receptors suggest that these receptors form ligand-dependent complexes that include specific caspases. Complex formation in the absence of ligand leads to caspase activation by a mechanism that is typically dependent on caspase cleavage of the receptor itself, releasing proapoptotic peptides. Cellular dependence receptors, considered in the aggregate, may thus form a system of molecular integration, analogous to the electrical integration system provided by dendritic arbors in the nervous system.


Subject(s)
Apoptosis/physiology , Receptors, Cell Surface/physiology , Animals , Caspases/metabolism , Cell Adhesion Molecules/physiology , DCC Receptor , Enzyme Activation , Humans , Netrin Receptors , Tumor Suppressor Proteins/physiology
15.
Pathol Biol (Paris) ; 53(6): 328-33, 2005 Jul.
Article in French | MEDLINE | ID: mdl-16004944

ABSTRACT

Currently, an increasing number of receptors appear to belong to the dependence receptors family. These proteins have the capacity to induce a program of apoptosis in settings of absence of their ligand. A cell that expresses one of these receptors is thus dependent on the presence of the ligand to survive. The observation that these receptors are lost in many cancers is then suggesting that this loss is a selective advantage for tumor development because it leads tumor cells not to be dependent for survival on the presence of the ligand. We propose to focus this review on the role of some of these receptors that have been intensively studied: the dependence receptors that bind the netrin-1. After having pointed out their role in the development of the nervous system and in cell death induction, we will discuss their putative role in the pathological context of tumorigenesis and more particularly in the control of colorectal cancers.


Subject(s)
Colorectal Neoplasms , Nerve Growth Factors/physiology , Receptors, Cell Surface/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Humans , Neoplasms , Nervous System/growth & development , Netrin Receptors , Netrin-1
16.
Br J Cancer ; 93(1): 1-6, 2005 Jul 11.
Article in English | MEDLINE | ID: mdl-15956977

ABSTRACT

Although cancer is a multifaceted disease, all cancer types share identical molecular and cellular mechanisms. These mechanisms involve a collection of alterations critical to the normal physiological functioning of cells, such as alterations of growth factor signalling pathways, angiogenesis, cell adhesion signals, DNA replication and apoptotic cell death. Many genes involved in the processes enumerated above are functionally inactive in tumour cells, designating them as putative 'tumour suppressor genes'. Back in the early 1990s, Vogelstein and colleagues suggested that a gene called DCC (for Deleted in Colorectal Cancer) could be a tumour suppressor gene because it was found to be deleted in more than 70% of colorectal cancers, as well as in many other cancers. During the last 15 years, controversial data have failed to firmly establish whether DCC is indeed a tumour suppressor gene. However, the recent observations that DCC triggers cell death and is a receptor for netrin-1, a molecule recently implicated in colorectal tumorigenesis, have prompted a renewal of interest in the role of DCC in tumorigenesis and suggest that the netrin-1/receptor pairs act as novel negative regulators of tumour development.


Subject(s)
Colorectal Neoplasms/physiopathology , Nerve Growth Factors/physiology , Receptors, Cell Surface/physiology , Tumor Suppressor Proteins/physiology , Colorectal Neoplasms/genetics , Genes, DCC , Genes, Tumor Suppressor , Humans , Netrin Receptors , Netrin-1
17.
Cell Mol Life Sci ; 61(15): 1854-66, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15289929

ABSTRACT

The recently described family of dependence receptors is a new family of functionally related receptors. These proteins have little sequence similarity but display the common feature of inducing two completely opposite intracellular signals depending on ligand availability: in the presence of ligand, these receptors transduce a positive signal leading to survival, differentiation or migration, while in the absence of ligand, the receptors initiate or amplify a negative signal for apoptosis. Thus, cells that express these proteins manifest a state of dependence on their respective ligands. The mechanisms that trigger cell death induction in the absence of ligand are in large part unknown, but typically require cleavage by specific caspases. In this review we will present the proposed mechanisms for cell death induction by these receptors and their potential function in nervous system development and regulation of tumorigenesis.


Subject(s)
Apoptosis/physiology , Receptors, Nerve Growth Factor/physiology , Animals , Apoptosis/genetics , Caspases/physiology , Cell Transformation, Neoplastic/pathology , Chick Embryo , Multigene Family , Nervous System/embryology , Nervous System/pathology , Nervous System Physiological Phenomena , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/genetics
18.
Apoptosis ; 9(1): 37-49, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14739597

ABSTRACT

A new family of functionally-related receptors has recently been proposed, dubbed dependence receptors. These proteins, only some of which share sequence similarities, display the common property that they transduce two different intracellular signals: in the presence of ligand, these receptors transduce a positive signal leading to survival, differentiation or migration; conversely, in the absence of ligand, the receptors initiate or amplify a signal for programmed cell death. Thus cells that express these proteins at sufficient concentrations manifest a state of dependence on their respective ligands. The signaling that mediates cell death induction upon ligand withdrawal is in large part uncharacterized, but typically includes a required interaction with, and cleavage by, specific caspases. Here, we review the current knowledge concerning dependence receptors, including the shared mechanisms for cell death induction and their potential relevance in nervous system development and regulation of tumorigenesis.


Subject(s)
Apoptosis , Animals , Axons , Caspases/metabolism , Cell Movement , Enzyme Activation , Humans , Integrins/metabolism , Ligands , Models, Biological , Neoplasms/metabolism , Nerve Growth Factors/metabolism , Nervous System/metabolism , Netrin Receptors , Netrin-1 , Receptor, Nerve Growth Factor , Receptors, Cell Surface/metabolism , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins
19.
Cell Death Differ ; 9(12): 1343-51, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12478471

ABSTRACT

The olfactory epithelium of adult mouse, where primary sensory neurons are massively committed to apoptosis by removal of their synaptic target, was used as a model to determine in vivo mechanisms for neuronal cell death induction. A macro-array assay revealed that the death of olfactory neurons is accompanied with over-expression of the serine protease inhibitor Spi2. This over-expression is associated with decreased serine protease activity in the olfactory mucosa. Moreover, in vitro or in vivo inhibition of serine proteases induced apoptotic death of olfactory neuronal cells. Interestingly, Spi2 over-expression is not occurring in olfactory neurons but in cells of the lamina propria, suggesting that Spi2 may act extracellularly as a cell death inducer. In that sense, we present evidence that in vitro Spi2 overexpression generates a secreted signal for olfactory neuron death. Hence, taken together these results document a possible novel mechanism for apoptosis induction that might occur in response to neurodegenerative insults.


Subject(s)
Apoptosis/genetics , Nuclear Proteins/metabolism , Olfactory Bulb/injuries , Olfactory Receptor Neurons/metabolism , Serine Endopeptidases/metabolism , Serpins , Animals , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Models, Biological , Nuclear Proteins/genetics , Olfactory Bulb/surgery , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Olfactory Receptor Neurons/cytology , RNA, Messenger/metabolism , Signal Transduction/physiology
20.
Neuroscience ; 109(4): 643-56, 2002.
Article in English | MEDLINE | ID: mdl-11927147

ABSTRACT

Netrin-1 is a bifunctional secreted protein that directs axon extension in various groups of developing axonal tracts. The transmembrane DCC (deleted in colorectal cancer) receptor is described as netrin-1 receptor and is involved in the attractive effects of netrin-1. In this study, we examined the spatio-temporal expression patterns of both netrin-1 and DCC in the rat olfactory system at different stages of development and during axonal regeneration following unilateral bulbectomy. High DCC expression was detected on the pioneer olfactory axons as they are extending toward the telencephalon. This expression was transient since from embryonic day 16 onwards, DCC was no longer detected along the olfactory nerve path. From embryonic day 14 until birth, DCC was also expressed within the mesenchyme surrounding the olfactory epithelium. During the same period, netrin-1 protein was detected along the trajectory of olfactory axons up to the olfactory bulb and its expression pattern in the nasal mesenchyme largely overlapped that of DCC. Moreover, netrin-1 continued to be present during the two first post-natal weeks, and a weak protein expression still persisted in the dorso-medial region of the olfactory epithelium in adult rats. While unilateral bulbectomy induced a transient up-regulation of netrin-1 in the lamina propria, particularly in the dorso-medial region of the neuroepithelium, no DCC expression was detected on the regenerating olfactory axons. In the developing olfactory bulb, the extension of mitral cell axons was associated with DCC presence while netrin-1 was absent along this axonal path. DCC was also highly expressed in the newly formed glomeruli after birth, and a weak DCC expression was still detected in the glomerular layer in adult rats. Taken together, these data support the notion that netrin-1, via DCC expressed on axons, may play a role in promoting outgrowth and/or guidance of pioneering olfactory axons toward the olfactory bulb primordium. Moreover, association of netrin-1 with mesenchymal DCC may provide a permissive environment to the growth of both pioneer and later-growing axons. The maintenance of netrin-1 expression in the nasal mesenchyme of adult rats as well as its regional up-regulation following unilateral bulbectomy infer that netrin-1, even in the absence of DCC, may be involved in the process of axonal growth of newly differentiated olfactory receptor neurons probably through the use of other receptors.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Differentiation/physiology , Growth Cones/metabolism , Nerve Growth Factors/metabolism , Nerve Regeneration/physiology , Olfactory Nerve/embryology , Olfactory Nerve/growth & development , Tumor Suppressor Proteins/metabolism , Animals , Animals, Newborn , Dendrites/metabolism , Dendrites/ultrastructure , Denervation , Female , Fetus , Gene Expression Regulation, Developmental/physiology , Growth Cones/ultrastructure , Male , Mesoderm/cytology , Mesoderm/metabolism , Nasal Cavity/cytology , Nasal Cavity/embryology , Nasal Cavity/growth & development , Netrin-1 , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Olfactory Mucosa/growth & development , Olfactory Nerve/cytology , Pregnancy , Rats , Rats, Wistar , Vomeronasal Organ/cytology , Vomeronasal Organ/embryology , Vomeronasal Organ/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...