Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(25): 3515-3527, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28166194

ABSTRACT

Wild-type p53 (wtp53) is described as a tumour suppressor gene; mutations in this gene occur in many human cancers and promote oncogenic capacity. Here, we establish that the oncogenic activity of mutant p53 (mtp53) is driven by the WASP-interacting protein (WIP). WIP knockdown from mtp53-expressing glioblastoma and breast cancer cells (BCC) greatly reduced proliferation and growth capacity of cancer stem cell (CSC)-like cells and decreased CSC-like markers (CD133, CD44 or YAP/TAZ). mtp53 overexpression in human astrocytes enhanced their proliferative capacity in suspension culture and increased expression of CSC markers and WIP. WIP knockdown compromised tumour glioblastoma and BCC growth capacity in vivo. We show that WIP is phosphorylated by AKT2 and is regulated by mtp53/p63 through enhancement of PI3K/AKT2-mediated integrin/receptor recycling pathways. WIP regulates this oncogenic pathway by controlling YAP/TAZ stability. We thus establish a new CSC signalling pathway downstream of mtp53 in which AKT2 regulates WIP and controls YAP/TAZ stability.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , Glioblastoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mutation , Neoplastic Stem Cells/metabolism , Phosphoproteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cytoskeletal Proteins/genetics , Female , Glioblastoma/genetics , Glioblastoma/pathology , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , MCF-7 Cells , Male , Neoplastic Stem Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphorylation/genetics , Protein Stability , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
2.
Mol Neurobiol ; 51(3): 1206-20, 2015.
Article in English | MEDLINE | ID: mdl-24965601

ABSTRACT

Alzheimer's disease (AD) is characterized by the presence of amyloid plaques mainly consisting of hydrophobic ß-amyloid peptide (Aß) aggregates and neurofibrillary tangles (NFTs) composed principally of hyperphosphorylated tau. Aß oligomers have been described as the earliest effectors to negatively affect synaptic structure and plasticity in the affected brains, and cellular prion protein (PrP(C)) has been proposed as receptor for these oligomers. The most widely accepted theory holds that the toxic effects of Aß are upstream of change in tau, a neuronal microtubule-associated protein that promotes the polymerization and stabilization of microtubules. However, tau is considered decisive for the progression of neurodegeneration, and, indeed, tau pathology correlates well with clinical symptoms such as dementia. Different pathways can lead to abnormal phosphorylation, and, as a consequence, tau aggregates into paired helical filaments (PHF) and later on into NFTs. Reported data suggest a regulatory tendency of PrP(C) expression in the development of AD, and a putative relationship between PrP(C) and tau processing is emerging. However, the role of tau/PrP(C) interaction in AD is poorly understood. In this study, we show increased susceptibility to Aß-derived diffusible ligands (ADDLs) in neuronal primary cultures from PrP(C) knockout mice, compared to wild-type, which correlates with increased tau expression. Moreover, we found increased PrP(C) expression that paralleled with tau at early ages in an AD murine model and in early Braak stages of AD in affected individuals. Taken together, these results suggest a protective role for PrP(C) in AD by downregulating tau expression, and they point to this protein as being crucial in the molecular events that lead to neurodegeneration in AD.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Phosphorylation , Plaque, Amyloid/metabolism , Prions/metabolism
3.
Biochim Biophys Acta ; 1843(2): 245-52, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24286867

ABSTRACT

Chaperones are critical for the folding and regulation of a wide array of cellular proteins. Heat Shock Proteins (Hsps) are the most representative group of chaperones. Hsp90 represents up to 1-2% of soluble protein. Although the Hsp90 role is being studied in neurodegenerative diseases, its role in neuronal differentiation remains mostly unknown. Since neuronal polarity mechanisms depend on local stability and degradation, we asked whether Hsp90 could be a regulator of axonal polarity and growth. Thus, we studied the role of Hsp90 activity in a well established model of cultured hippocampal neurons using an Hsp90 specific inhibitor, 17-AAG. Our present data shows that Hsp90 inhibition at different developmental stages disturbs neuronal polarity formation or axonal elongation. Hsp90 inhibition during the first 3h in culture promotes multiple axon morphology, while this inhibition after 3h slows down axonal elongation. Hsp90 inhibition was accompanied by decreased Akt and GSK3 expression, as well as, a reduced Akt activity. In parallel, we detected an alteration of kinesin-1 subcellular distribution. Moreover, these effects were seconded by changes in Hsp70/Hsc70 subcellular localization that seem to compensate the lack of Hsp90 activity. In conclusion, our data strongly suggests that Hsp90 activity is necessary to control the expression, activity or location of specific kinases and motor proteins during the axon specification and axon elongation processes. Even more, our data demonstrate the existence of a "time-window" for axon specification in this model of cultured neurons after which the inhibition of Hsp90 only affects axonal elongation mechanisms.


Subject(s)
Cell Polarity , HSP90 Heat-Shock Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Axons/drug effects , Axons/metabolism , Benzoquinones/pharmacology , Cell Polarity/drug effects , Glycogen Synthase Kinase 3/metabolism , Growth Cones/drug effects , Growth Cones/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hippocampus/cytology , Kinesins/metabolism , Lactams, Macrocyclic/pharmacology , Mice , Neurons/drug effects , Neurons/enzymology , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
4.
Neuropathol Appl Neurobiol ; 40(3): 311-26, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23741998

ABSTRACT

AIMS: Transmissible spongiform encephalopathies, also called prion diseases, are characterized by the cerebral accumulation of misfolded prion protein (PrP(SC) ) and subsequent neurodegeneration. However, despite considerable research effort, the molecular mechanisms underlying prion-induced neurodegeneration are poorly understood. Here, we explore the hypothesis that prions induce dysfunction of the PI3K/Akt/GSK-3 signalling pathway. METHODS: We employed two parallel approaches. Using cell cultures derived from mouse primary neurones and from a human neuronal cell line, we identified common elements that were modified by the neurotoxic fragment of PrP(106-126) . These studies were then complemented by comparative analyses in a mouse model of prion infection. RESULTS: The presence of a polymerized fragment of the prion protein (PrP(106-126) ) or of a prion strain altered PI3K-mediated signalling, as evidenced by Akt inhibition and GSK-3 activation. PI3K activation by the addition of insulin or the expression of a constitutively active Akt mutant restored normal levels of Akt and GSK-3 activity. These changes were correlated with a reduction in caspase activity and an increase in neuronal survival. Moreover, we found that activation of caspase 3, Erk and GSK-3 are common features of PrP(106-126) -mediated neurotoxicity in cellular systems and prion infection in the mouse cerebellum, while activation of caspase 12 and JNK was observed in cellular models. CONCLUSIONS: Our findings in cell culture and in vivo models of prion disease demonstrate marked alterations to the PI3K/Akt/GSK-3 pathway and suggest that two additional pathways contribute to PrP-induced neurotoxicity as responsible of JNK and caspase 12 activation.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Prion Diseases/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Humans , Mice , Peptide Fragments/metabolism , Prions/metabolism
5.
Biochim Biophys Acta ; 1833(5): 1052-64, 2013 May.
Article in English | MEDLINE | ID: mdl-23295457

ABSTRACT

The oestradiol plays an important role in normal brain development and exerts neuroprotective actions. Oestradiol is mainly produced in the ovary and in addition is locally synthesised in the brain. Most of the oestradiol functions have been associated with its capacity to directly bind and dimerize "classical oestrogen receptors" (ERs), alpha and beta. The ERs' actions have been classified as "genomic" and "non-genomic" depending on whether protein synthesis occurs through ER driven transcription or not. Indeed, recent evidence suggests that oestrogen may also act as a more general "trophic factor". Hence, we have studied the capacity of oestradiol to activate the PI3K/Akt pathway and its implication in axonal growth and neuronal morphogenesis. Our data show that when oestrogen receptors are blocked the axonal and dendritic lengths are reduced in mouse primary neurons. We found that Akt/Rheb/mTORC1 responds to ER activation in neurons and that some elements of this pathway are able to restore a normal neuronal morphology even in the presence of oestrogen receptor antagonist. All these data demonstrate a new mechanism regulated by oestradiol, at least in neuronal morphogenesis.


Subject(s)
Estradiol , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Brain/growth & development , Brain/metabolism , Cell Line , Estradiol/administration & dosage , Estradiol/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , Mechanistic Target of Rapamycin Complex 1 , Mice , Neuroblastoma/metabolism , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction
6.
J Neuroendocrinol ; 24(1): 191-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21722217

ABSTRACT

Oestradiol acts in the brain by multiple mechanisms, including the regulation of transcriptional activity through classical oestrogen receptors, α and ß, and by the activation of membrane/cytoplasm-initiated signalling cascades. In neuroblastoma cells, primary neurones in culture and in the brain in vivo, oestradiol activates the phosphoinositide 3-kinase/Akt/glycogen synthase kinase 3 signalling pathway by a mechanism involving oestrogen receptor α. Through this pathway, oestradiol regulates the stability of ß-catenin, induces the translocation of ß-catenin to the cell nucleus and regulates ß-catenin-mediated transcription through the T cell factor/DNA complex. Genomic analyses in neuroblastoma cells have revealed that the set of genes regulated by oestradiol through ß-catenin is not identical to that regulated by the Wnt signalling pathway, revealing a new mechanism for oestradiol signalling in neurones.


Subject(s)
Estradiol/pharmacology , Neurons/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , beta Catenin/metabolism , Animals , Neurons/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
7.
Cereb Cortex ; 22(5): 1191-202, 2012 May.
Article in English | MEDLINE | ID: mdl-21810783

ABSTRACT

Wiskott-Aldrich syndrome protein (WASP) -interacting protein (WIP) is an actin-binding protein involved in the regulation of actin polymerization in cells, such as fibroblasts and lymphocytes. Despite its recognized function in non-neuronal cells, the role of WIP in the central nervous system has not been examined previously. We used WIP-deficient mice to examine WIP function both in vivo and in vitro. We report here that WIP(-)(/-) hippocampal neurons exhibit enlargement of somas as well as overgrowth of neuritic and dendritic branches that are more evident in early developmental stages. Dendritic arborization and synaptogenesis, which includes generation of postsynaptic dendritic spines, are actin-dependent processes that occur in parallel at later stages. WIP deficiency also increases the amplitude and frequency of miniature excitatory postsynaptic currents, suggesting that WIP(-)(/-) neurons have more mature synapses than wild-type neurons. These findings reveal WIP as a previously unreported regulator of neuronal maturation and synaptic activity.


Subject(s)
Carrier Proteins/metabolism , Hippocampus/growth & development , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , Synapses/metabolism , Animals , Blotting, Western , Cytoskeletal Proteins , Excitatory Postsynaptic Potentials/physiology , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Microscopy, Confocal
8.
CNS Neurol Disord Drug Targets ; 10(2): 208-14, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21222634

ABSTRACT

Few targets for neuroprotection have been defined in Alzheimer's disease (AD). Recent data from the role of Wnt, insulin-like growth factor-1 and estradiol pathways in AD suggest some therapeutic targets for disease treatment, and have led us to evaluate the "common factors" in these pathways as further candidate targets. These data have led us to propose that glycogen synthase kinase-3 (GSK-3) inhibition appears to be a common feature of these pathways. Besides, considering that GSK-3 activation seems to be correlated with neurodegeneration, its selection as a relevant target appears obvious. The capacity of different GSK-3 inhibitors to prevent amyloid ß-peptide neurotoxicity and tau phosphorylation has been evaluated in order to develop novel clinical and therapeutic approaches. Different approaches could be used to search for new neuroprotective compounds. The most classical of these is to first define the target and then design a specific in vitro screening assay for it. Alternatively, a cell model of cell culture could be used as a "primary screen". Following this rationale, we have used a combined approach in which we first used an in vitro system to select compounds able to inhibit recombinant GSK3ß. Subsequently, we subjected the candidate compounds to three consecutive cell-based complementary screening assays. First, cell viability was assessed using a neuroblastoma cell line before assaying the capacity of the compounds to reduce tau phosphorylation. Finally, we designed a neuronal cell model of apoptosis using the phosphatidylinositol kinase-3 inhibitor LY294002. Finally, we summarize several new compounds with "neuroprotective" properties.


Subject(s)
Apoptosis/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Glycogen Synthase Kinase 3/metabolism , Humans , Molecular Targeted Therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation
9.
J Neurosci Res ; 86(3): 668-74, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17893926

ABSTRACT

Recent evidence suggests that intramolecular autophosphorylation is responsible for the tyrosine phosphorylation (pY) of residues 279 or 216 of glycogen synthase kinase-3 (GSK-3alpha or beta), an event that appears to play an important role in regulating this kinase. This provocative hypothesis was based on the capacity of certain nonselective GSK-3 inhibitors to alter both the activity of GSK-3 and its pY. Inhibitors of GSK-3 are not always capable of preventing this tyrosine phosphorylation, which may require an extended period of time. For example, although lithium chloride inhibits GSK-3 activity, this inhibition does not alter its pY content. Furthermore, even when GSK-3 activity is impaired, GSK-3 pY can still be modified by physiological or pharmacological agents. Taken together, these data indicate that GSK-3 kinase activity is not necessarily correlated with the extent of GSK-3 pY. We hypothesized that some as-yet-unidentified tyrosine kinases and phosphatases may also regulate this kinase.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Protein Kinase Inhibitors/pharmacology , Tyrosine/metabolism , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/chemistry , Lysophospholipids/pharmacology , Mice , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Vanadates/pharmacology
10.
Apoptosis ; 12(2): 395-409, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17191124

ABSTRACT

The isolation of new molecules from marine sources opens the door to their possible therapeutic use against tumors and other pathological conditions. Indeed, we recently defined the cytotoxicity of ES 285, obtained from the clam Mactromeris polynima, and its affects on the cells microfilament but not the microtubule network. Considering the analogy between ES 285 and sphingosine-related lipids, we wondered whether ES 285 might affect the activity of PKC at the intracellular level. While we anticipated that ES 285 might inhibit PKC, it turns out that in contrast it serves to activate PKC at the cellular level. Indeed, like other sphingosine-related lipids, ES 285 induces the phosphorylation of MARCKS. Additionally, we further examined the cytotoxicity of ES 285 to elucidate the molecular mechanisms through which this compound triggers apoptosis. When the influence of ES 285 on "cell death markers" was assessed, it became clear that ES285 activates caspase 3 and 12, and that it modified the phosphorylation of p53. In contrast, ES 285 does not affect other pathways widely implicated in regulating cell survival/apoptosis, such as JNK, Erks or Akt. Thus, these data suggest that ES 285-triggers an atypical cell death program when compared to other sphingosine-dependent apoptosis pathways.


Subject(s)
Alkanes/pharmacology , Lipids/pharmacology , Sphingolipids/pharmacology , Alkanes/chemistry , Animals , Caspase Inhibitors , Cell Death/drug effects , Cell Extracts , Cytochromes c/metabolism , Diglycerides/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flow Cytometry , Lethal Dose 50 , Lipids/chemistry , Mice , Mitochondria/drug effects , NIH 3T3 Cells , Protein Kinase C/antagonists & inhibitors , Protein Transport/drug effects , Sphingolipids/chemistry , Subcellular Fractions/drug effects , Tumor Suppressor Protein p53/metabolism
11.
Neuroscience ; 129(3): 583-92, 2004.
Article in English | MEDLINE | ID: mdl-15541880

ABSTRACT

The pathological significance of intracellular Abeta accumulation in vivo is not yet fully understood. To address this, we have studied transgenic rats expressing Alzheimer's-related transgenes that accumulate Abeta intraneuronally in the cerebral and hippocampal cortices but do not develop extracellular amyloid plaques. In these rats, the presence of intraneuronal Abeta is sufficient to provoke up-regulation of the phosphorylated form of extracellular-regulated kinase (ERK) 2 and its enzymatic activity in the hippocampus while no changes were observed in the activity or phosphorylation status of other putative tau kinases such as p38, glycogen synthase kinase 3, and cycline-dependent kinase 5. The increase in active phospho-ERK2 was accompanied by increased levels of tau phosphorylation at S396 and S404 ERK2 sites and a decrease in the phosphorylation of the CREB kinase p90RSK. In a water maze paradigm, male transgenic rats displayed a mild spatial learning deficit relative to control littermates. Our results suggest that in the absence of plaques, intraneuronal accumulation of Abeta peptide correlates with the initial steps in the tau-phosphorylation cascade, alterations in ERK2 signaling and impairment of higher CNS functions in male rats.


Subject(s)
Amyloid beta-Peptides/metabolism , Memory Disorders/physiopathology , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/genetics , Analysis of Variance , Animals , Animals, Genetically Modified , Animals, Newborn , Behavior, Animal , Blotting, Western/methods , Brain/cytology , Humans , Immunohistochemistry/methods , Maze Learning/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory Disorders/genetics , Phosphorylation , Presenilin-1 , Rats , Rats, Wistar , Reaction Time/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/genetics
12.
Mol Cell Neurosci ; 25(3): 363-73, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15033165

ABSTRACT

Estrogens regulate a wide set of neuronal functions such as gene expression, survival and differentiation in a manner not very different from that exerted by neurotrophins or by growth factors. The best-studied hormonal action is the transcriptional activation mediated by estrogen receptors. However, the direct effects of estrogen on growth factor signaling have not been well clarified. The present data show that estradiol, in vivo, induces a transient activation of GSK3 in the adult female rat hippocampus, followed by a more sustained inhibition, as inferred from phosphorylation levels of Tau. Similar data was obtained from cultured hippocampal neurons when treated with the hormone. The transient activation was confirmed by direct measure of GSK3 kinase activity. In addition, our results show a novel complex of estrogen receptor alpha, GSK3, and beta-catenin. The presence of the hormone removes beta-catenin from this complex. There is a second complex, also affected by estradiol, in which Tau is associated with GSK3, beta-catenin, and elements of the PI3 kinase complex. Considering the role of GSK3 in neurodegeneration, our data suggest that part of the neuroprotective effects of estrogen may be due to the control of GSK3.


Subject(s)
Cytoskeletal Proteins/metabolism , Estradiol/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hippocampus/drug effects , Hippocampus/enzymology , Receptors, Estrogen/metabolism , Trans-Activators/metabolism , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Female , Glycogen Synthase Kinase 3/metabolism , Hippocampus/metabolism , Mice , Rats , Rats, Wistar , beta Catenin
13.
Neurochem Int ; 44(4): 231-42, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14602086

ABSTRACT

Neuronal differentiation is a complex process in which many different signalling pathways may be involved. An increase in the intracellular levels of cyclic AMP (cAMP) has been shown to induce neuronal differentiation and also to cooperate with NGF to induce PC12 neurite outgrowth in a Ras-dependent manner. However, the neuritogenic activities associated with cAMP are still not well understood. The purpose of this study was to investigate the potential neuritogenic activities mediated by cAMP. For this purpose, we used the human neuroblastoma cell line SH-SY5Y. These neuroblastoma cells respond to cAMP by forming neurite-like extensions. We tried to identify some essential pathways involved in the cAMP-induced neurite elongation of these cells. Our results indicated that PKA is transiently activated in this elongation model. When we blocked PKA activity, elongation did not take place. Similarly, PI3K also plays an essential role because when we blocked this kinase activity, there was no neurite elongation. Indeed, over-expression of the p110-catalytic subunit or an activating form of the p85-regulatory subunit (p65) is able to induce some degree of neurite extension. Moreover, our results showed that when elongation is initiated, PI3K is still essential for maintenance of the neuronal morphology, whereas PKA or MAPK (ERKs or p38) activation does not appear to be necessary during this process.


Subject(s)
Cell Differentiation/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/metabolism , Neurons/cytology , Phosphatidylinositol 3-Kinases/physiology , Cell Line, Tumor , Fluorescent Antibody Technique, Indirect , Humans
14.
J Neurochem ; 87(4): 879-90, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14622119

ABSTRACT

Semaphorins (sema) constitute a family of molecules sharing a common extracellular domain (semaphorin domain). This family includes several types of secreted and membrane-associated molecules that are grouped into eight subclasses (subclasses 1-7 and viral semaphorins). Subclass 3 semaphorins are secreted molecules involved in axonal guidance, mainly through repulsive gradients and induction of growth cone collapse. More recently sema 3 molecules have been identified as positive factors in dependence of the type of neurons. Besides their axonal guidance function, some semaphorins have been implicated in apoptosis and survival. We investigated the effect of sema3C on survival and neurite outgrowth of rat cerebellar granule neurons (CGNs) in culture. 3T3 cells were stably transfected with sema3C. Several clonal lines were established and tested for their neuritogenic activity and one, S3C-8, was selected for the bulk of experiments. S3C-8 was co-cultured with CGNs. Sema3C enhanced CGN viability as assessed in co-cultures of CGNs with monolayers of S3C-8 in comparison with co-cultures of CGNs with control mock-transfected 3T3 cells. Moreover sema3C induced neuritogenesis of cultured CGNs, which express neuropilin-1 and -2. S3C-8 cells, overexpressing sema3C, were significantly more neuritogenic for CGN than poly l-lysine (PLL), a positive substrate for CGNs, as assessed by the measurement of the length of neurites and confirmed by Tau expression along the time of culture. CGNs co-cultured with S3C-8, showed up-regulation of the expression of axonal microtubule-associated proteins (MAPs) such as Tau, phosphorylated MAP2C and mode I-phosphorylated MAP1B compared with neurons cultured on control 3T3 cells. We also found increased expression of a specific marker of neuronal cell bodies and dendrites, high molecular weight MAP2 (HMW-MAP2). Interestingly, there was no accompanying up-regulation of a marker enriched within the neuronal somatodendritic domain, mode II-phosphorylated MAP1B. These data support the idea that secreted sema3C favors survival and neuritogenesis of cultured CGNs.


Subject(s)
Carrier Proteins/physiology , Cerebellum , Nerve Tissue Proteins/physiology , Neurites/physiology , Neurons/metabolism , Semaphorin-3A , 3T3 Cells , Animals , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebellum/cytology , Clone Cells , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/biosynthesis , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurites/drug effects , Neurons/cytology , Neurons/drug effects , Neuropilin-1/biosynthesis , Neuropilin-2/biosynthesis , Nucleotides, Cyclic/pharmacology , Rats , Transfection
15.
Biochim Biophys Acta ; 1582(1-3): 144-53, 2002 May 23.
Article in English | MEDLINE | ID: mdl-12069822

ABSTRACT

Neurite retraction is a crucial process during nervous system development and neurodegeneration. This process implies reorganization of the neuronal cytoskeleton. Some bioactive lipids such as lysophosphatidic acid (LPA) induce neurite retraction. The reorganization of the actin cytoskeleton during neurite retraction is one of the best-characterized effects of LPA. However, less information is available regarding the reorganization of the microtubule (MT) network in response to LPA in neuronal cells. Here, we first give an overview of the roles of cytoskeleton during neurite outgrowth, and subsequently, we review some of the data from different laboratories concerning LPA-induced cytoskeletal rearrangement in neuronal cells. We also summarize our own recent results about modifications of MTs during LPA-induced neurite retraction. We have shown that LPA induces changes in tubulin pools and increases in the phosphorylation levels of microtubule-associated proteins (MAPs), such as Tau. Tau hyperphosphorylation in response to LPA is mediated by the activation of glycogen synthase kinase-3 (GSK-3). The upregulation of GSK-3 activity by LPA seems to be a general process as it occurs in diverse neuronal cells of different species in correlation with the neurite retraction process.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cytoskeleton/ultrastructure , Lysophospholipids/physiology , Neurons/physiology , Animals , Cytoskeleton/physiology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Microtubule-Associated Proteins/physiology , Neurites/physiology , Neurons/ultrastructure , Tubulin/physiology
16.
J Neurosci Res ; 67(6): 713-9, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11891784

ABSTRACT

Several lines of evidence have indicated that changes in the structure of neuronal cytoskeleton provide the support for the dramatic morphological changes that occur during neuronal differentiation. It has been proposed that microtubule-associated proteins can contribute to the development of this phenomenon by controlling the dynamic properties of microtubules. In this report we have characterized the effect of the combined suppression of MAP1B and tau, and MAP1B and MAP2 on neuronal polarization in cultured hippocampal cells grown on a laminin-containing substrate. We have taken advantage of the use of a mouse line deficient in MAP1B expression obtained by the gene trapping approach. In addition to this engineered mice line we used the antisense oligonucleotide approach to induce the suppression of tau or MAP2, in wild type and MAP1B-deficient neurons. Together these results show a synergistic role for MAP1B/MAP2 and MAP1B/TAU.


Subject(s)
Cell Polarity/physiology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Cells, Cultured , Female , Fluorescent Antibody Technique , Hippocampus/cytology , Male , Mice , Mice, Knockout , Oligonucleotides, Antisense/pharmacology , Polylysine , Pregnancy , tau Proteins/genetics , tau Proteins/metabolism
17.
Neurosci Lett ; 314(1-2): 87-91, 2001 Nov 13.
Article in English | MEDLINE | ID: mdl-11698153

ABSTRACT

Fibronectin appears to be present in Senile Plaques of Alzheimer's disease brains. These senile or neuritic plaques are surrounded by dystrophic neurites, activated microglia and reactive astrocytes. The purpose of this work was to establish if a direct correlation exists between the production of Fibronectin (FN) by astrocytes and the presence of amyloid, analysing the modification of this protein produced after the treatment of cultured astrocytes with amyloid peptide (25-35). Our data showed that the addition of previously polymerised A beta-peptide to cultured astrocytes induced a marked increase in FN immunoreactivity that is in part dependent on phosphatases 2A or phosphatase 1, since was partially inhibited by okadaic acid. The increased amount of FN did not appear to be associated to any specific single isoform of which are mainly present in the rat brain. Our data suggest that in vivo FN accumulated in senile plaques may be the result, at least in part, of the response of reactive astrocyte to the presence of amyloid peptide. The importance of FN up-regulation in vivo, as part of a 'positive' response of the astrocytes to produce molecules that favours neurite outgrowth, is discussed.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Astrocytes/metabolism , Cell Differentiation/physiology , Cerebral Cortex/metabolism , Fibronectins/metabolism , Peptide Fragments/pharmacology , Plaque, Amyloid/metabolism , Alternative Splicing/physiology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/pathology , Cell Differentiation/drug effects , Cell Size/drug effects , Cell Size/physiology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Enzyme Inhibitors/pharmacology , Fibronectins/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/physiopathology , Growth Substances/metabolism , Immunohistochemistry , Neurites/drug effects , Neurites/metabolism , Neurites/pathology , Okadaic Acid/pharmacology , Peptide Fragments/metabolism , Plaque, Amyloid/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Vimentin/metabolism
18.
J Neurochem ; 78(3): 468-81, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483649

ABSTRACT

It has been extensively described that neuronal differentiation involves the signalling through neurotrophin receptors to a Ras-dependent mitogen-activated protein kinase (MAPK) cascade. However, signalling pathways from other neuritogenic factors have not been well established. It has been reported that cAMP may activate protein kinase (PKA), and it has been shown that PKA-mediated stimulation of MAPK pathway regulates not only neuritogenesis but also survival. However, extracellular regulated kinases (ERKs) mediated pathways are not sufficient to explain all the processes which occur in neuronal differentiation. Our present data show that: in cAMP-mediated neuritogenesis, using the SH-SY5Y human neuroblastoma cell line, there exists a link between the activation of PKA and stimulation of phosphatidylinositol 3-kinase (PI3K). Both kinase activities are essential to the initial elongation steps. Surprisingly, this neuritogenic process appears to be independent of ERKs. While the activity of PI3K is essential for elongation and maintenance of neurites, its inhibition causes retraction. In this neurite retraction process, GSK3 is activated. Using both a pharmacological approach and gene transfer of a dominant negative form of GSK3, we conclude that this induced retraction is a GSK3-dependent process which in turn appears to be a common target for transduction pathways involved in lysophosphatidic acid-mediated and PI3K-mediated neurite retraction.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Neurites/physiology , Phosphoinositide-3 Kinase Inhibitors , Sulfonamides , Androstadienes/pharmacology , Bucladesine/antagonists & inhibitors , Bucladesine/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Differentiation , Cell Fractionation , Chromones/pharmacology , Culture Media, Serum-Free , Cyclic AMP/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genes, Reporter , Glycogen Synthase Kinase 3 , Humans , Immunoblotting , Isoquinolines/pharmacology , Lithium Chloride/pharmacology , Microtubule-Associated Proteins/metabolism , Morpholines/pharmacology , Neurites/drug effects , Neurites/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Tubulin/metabolism , Tumor Cells, Cultured , Wortmannin
19.
Biotechniques ; 31(2): 394-402, 404-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11515376

ABSTRACT

Retroviral vectors have long been used in a wide variety of gene transfer applications but have certain drawbacks, such as small cargo size, limited tropism, and low titers. HSV expression vectors overcome these disadvantages, but, because they persist in target cells as nonreplicative episomes, they are not retained in all the progeny of dividing cells. Chimeric HSV/AAV products that can mediate transgene integration in human mitotic cells have been constructed, but, to date, genetic modification of dividing cells in animal models using HSV products has not been possible. Here, we report the construction of hybrid HSV/retroviral vectors that exhibit up to 50-fold higher transgene integration efficiency compared to vectors containing only HSV-1 components. Efficient integration of a retroviral transgene cassette encoding pac in human cells required expression of the Moloney murine leukemia virus gag-pol genes, but in murine cells, could also be mediated by endogenous activities, albeit at a lower level. Gene delivery was equally efficient in BHK21, a cell line resistant to retroviral infection, and transgene retention and expression were observed to be stable for least one month in Hs683 human glioma cells. These vectors have wide applications for the genetic modification of many cell types.


Subject(s)
Genetic Vectors , Herpesvirus 1, Human/genetics , Moloney murine leukemia virus/genetics , Mutagenesis, Insertional/methods , Transgenes/genetics , Animals , Antibiotics, Antineoplastic , Cell Line, Transformed , Chlorocebus aethiops , Cricetinae , Genes, gag , Genes, pol , Genetic Markers , Glioma , Humans , Kidney/cytology , Mitosis , Puromycin , Tumor Cells, Cultured , Viral Regulatory and Accessory Proteins/genetics , Virus Integration
20.
Mol Cell Neurosci ; 16(4): 408-21, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11085878

ABSTRACT

Microtubule-associated protein 1B (MAP1B) has been implicated in axogenesis in cultured cells. To gain insight into the functions that MAP1B plays in vivo, we analyzed a strain of Map1B mutant mice generated by a gene trapping approach. Homozygous mice die on the first day after birth, probably due to a severe abnormal development of the nervous system. They present alterations in the structure of several brain regions. The normal Map1B gene yields different protein isoforms from alternatively spliced transcripts. The smaller isoforms were present in wild type, hetero-, and homozygous mice, but their expression was higher in the mutants than in the wild-type. Moreover, trace amounts of MAP1B protein were also observed in Map1B homozygous mutants, indicating an alternative splicing around the gene trap insertion. Thus, the Map1B gene trapped mutation reported in this work did not generated a null mutant, but a mouse with a drastic deficiency in MAP1B expression. Analyses of these mice indicate the presence of several neural defects and suggest the participation of MAP1B in neuronal migration.


Subject(s)
Alternative Splicing/physiology , Genes, Lethal/physiology , Mice, Knockout , Microtubule-Associated Proteins/genetics , Animals , Animals, Newborn , Blotting, Northern , Blotting, Western , Exons , Gene Expression/physiology , Genotype , Heterozygote , Homozygote , Isomerism , Mice , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/chemistry , Nervous System/chemistry , Nervous System/embryology , Phenotype , RNA, Messenger/analysis , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...