Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Oncogene ; 33(26): 3451-62, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-23912459

ABSTRACT

Glioblastoma multiforme (GBM) are highly invasive and angiogenic malignancies with a median survival time from diagnosis of <15 months. Previous work has revealed robust overexpression of fibronectin (FN) mRNA in GBM, although immunohistochemical staining of FN in these tumors is typically associated with the angiogenic vasculature. Here we sought to examine the expression of tumor cell FN and address its possible involvement in the invasive phenotype of GBM. We found that FN was expressed and assembled into fibrillar arrays in human tumors and in established GBM lines. Cultured cells spontaneously formed dense cellular networks and spheroid-like domes. Depletion of FN by targeted-short hairpin RNA expression disrupted matrix assembly and multicellular network organization by exerting profound effects on cell adhesion and motility. Although FN depletion enhanced persistent directional migration of single cells, it compromised collective invasion of spheroids through a laminin-rich matrix and sensitized cells to ionizing radiation. In orthotopic grafts, FN depletion significantly reduced tumor growth and angiogenesis. Together our results show that FN produced by the tumor cells has a role in GBM pathophysiology and they provide insights into the implications that targeting FN interactions may have for combating this dreaded disease.


Subject(s)
Cell Adhesion/genetics , Fibronectins/metabolism , Glioblastoma/pathology , Animals , Basement Membrane/cytology , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Extracellular Matrix , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Integrin alpha5beta1/metabolism , Mice , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA Interference , RNA, Small Interfering , Spheroids, Cellular , Tumor Cells, Cultured
2.
Int J Obes (Lond) ; 33(12): 1348-55, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19773740

ABSTRACT

OBJECTIVES: The involvement of skeletal muscle mitochondrial uncoupling protein-3 (UCP3) in the control of energy expenditure in skeletal muscle and at the whole-body level is still a matter of debate. We previously reported that UCP3 downregulation is linked to an enhanced mitochondrial energy metabolism in rat skeletal muscle as a result of acute capsiate treatment. Here, we aimed at investigating noninvasively the effects of chronic capsiate ingestion on metabolic changes occurring in exercising gastrocnemius muscle and at the whole-body level. METHODS: We used an original experimental setup allowing a complete noninvasive investigation of gastrocnemius muscle function in situ using 31-phosphorus magnetic resonance spectroscopy. Whole-body fat composition was determined using magnetic resonance imaging and UCP3 gene expression was measured by quantitative real-time RT-PCR analysis. RESULTS: We found that a 14-day daily administration of capsiate (100 mg kg(-1) body weight) reduced UCP3 gene expression and increased phosphocreatine level at baseline and during the stimulation period in gastrocnemius muscle. During muscle stimulation, pH(i) showed a larger alkalosis in the capsiate group suggesting a lower glycolysis and a compensatory higher aerobic contribution to ATP production. Although the capsiate-treated rats were hyperphagic as compared to control animals, they showed a lower weight gain coupled to a decreased abdominal fat content. CONCLUSION: Overall, our data indicated that capsiate administration contributes to the enhancement of aerobic ATP production and the reduction of body fat content coupled to a UCP3 gene downregulation.


Subject(s)
Abdominal Fat/drug effects , Capsaicin/analogs & derivatives , Energy Metabolism/drug effects , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/drug effects , Uncoupling Agents/pharmacology , Abdominal Fat/metabolism , Animals , Capsaicin/administration & dosage , Capsaicin/pharmacology , Down-Regulation , Energy Metabolism/physiology , Female , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Oxidation-Reduction/drug effects , Rats , Uncoupling Agents/administration & dosage , Uncoupling Protein 3
3.
Am J Physiol Endocrinol Metab ; 292(5): E1474-82, 2007 May.
Article in English | MEDLINE | ID: mdl-17264228

ABSTRACT

Although it has been suggested that the skeletal muscle mitochondrial uncoupling protein-3 (UCP3) is involved in regulating energy expenditure, its role is still poorly understood. In the present study, we aimed at investigating noninvasively, using magnetic resonance techniques, metabolic changes occurring in exercising muscle as a result of capsiate treatment, which has been previously linked to UCP3 upregulation. We showed that capsiate ingestion strongly reduced UCP3 gene expression in rat gastrocnemius muscle. This large underexpression was accompanied by a significant increase in the rate of mitochondrial ATP production and phosphocreatine level both at rest and during muscle stimulation. Similarly, the stimulation-induced ATP fall and ADP accumulation were significantly less after capsiate administration than in untreated rats. The larger oxidative ATP production rate could not be explained by a proportional decrease in the anaerobic component, i.e., glycolysis and phosphocreatine breakdown. In addition, the mechanical performance was not affected by capsiate administration. Finally, the plasma free fatty acid (FFA) level increased in capsiate-treated rats, whereas no significant change was observed after muscle stimulation in the control group. Considering the corresponding enhanced UCP3 mRNA expression occurring in the control group after muscle stimulation, one can suggest that changes in FFA level and UCP3 mRNA expression are not mechanistically correlated. Overall, we have shown that capsiate administration induced a UCP3 downregulation coupled with an increased mitochondrial ATP synthesis, whereas the muscle force-generating capacity was unchanged. This suggests that a decrease in muscle efficiency and/or additional noncontractile ATP-consuming mechanisms result from UCP3 downregulation.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Capsaicin/analogs & derivatives , Ion Channels/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Capsaicin/pharmacology , Down-Regulation/drug effects , Electric Stimulation , Energy Metabolism , Fatty Acids, Nonesterified/blood , Female , Hydrogen-Ion Concentration , Ion Channels/biosynthesis , Ion Channels/genetics , Magnetic Resonance Spectroscopy , Mitochondria, Muscle/drug effects , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Muscle, Skeletal/drug effects , Phosphocreatine/metabolism , Rats , Rats, Sprague-Dawley , Uncoupling Protein 3
4.
Neurobiol Aging ; 26(1): 103-14, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15585350

ABSTRACT

Age-dependent cognitive impairments have been correlated with functional and structural modifications in the hippocampal formation. In particular, the brain endogenous steroid pregnenolone-sulfate (Preg-S) is a cognitive enhancer whose hippocampal levels have been linked physiologically to cognitive performance in senescent animals. However, the mechanism of its actions remains unknown. Because neurogenesis is sensitive to hormonal influences, we examined the effect of Preg-S on neurogenesis, a novel form of plasticity, in young and old rats. We demonstrate that in vivo infusion of Preg-S stimulates neurogenesis and the expression of the polysialylated forms of NCAM, PSA-NCAM, in the dentate gyrus of 3- and 20-month-old rats. These influences on hippocampal plasticity are mediated by the modulation of the gamma-aminobutyric acid receptor complex A (GABA(A)) receptors present on hippocampal neuroblasts. In vitro, Preg-S stimulates the division of adult-derived spheres suggesting a direct influence on progenitors. These data provide evidence that neurosteroids represent one of the local secreted signals controlling hippocampal neurogenesis. Thus, therapies which stimulate neurosteroidogenesis could preserve hippocampal plasticity and prevent the appearance of age-related cognitive disturbances.


Subject(s)
Gene Expression Regulation/drug effects , Hippocampus/cytology , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity/drug effects , Neurons/drug effects , Pregnenolone/pharmacology , Sialic Acids/metabolism , Age Factors , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Count/methods , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/physiology , Injections, Intraventricular/methods , Male , Microscopy, Immunoelectron/methods , Neurons/metabolism , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Time Factors
5.
Behav Brain Res ; 147(1-2): 31-9, 2003 Dec 17.
Article in English | MEDLINE | ID: mdl-14659567

ABSTRACT

Motor and cognitive phenotypes were assessed in mice deficient for the close homologue of the L1 adhesion molecule (CHL1). The CHL1-deficient mice displayed signs of decreased stress and a modification of exploratory behaviour. The mice also showed motor impairments on the Rotarod, but they were able to move as fast as controls in the alleys of a T-maze. The observed changes were assumed to be related to a deficit in attention. In addition, gender differences in CHL1 deficits were found and are discussed in view of a possible interaction with other cell adhesion molecules (CAMs) during development. The results are discussed in relation with motor and cognitive deficits in the human, caused by mutations of the distal part of the chromosome 3 which contains the CHL1 orthologue.


Subject(s)
Emotions/physiology , Mice, Knockout/physiology , Proteins/metabolism , Psychomotor Performance/physiology , Animals , Body Constitution/genetics , Body Weight/genetics , Cell Adhesion Molecules , Exploratory Behavior , Female , Male , Maze Learning/physiology , Membrane Proteins , Mice , Mice, Inbred C57BL , Proteins/genetics , Reaction Time , Running , Sex Factors , Time Factors
6.
J Neurosci Res ; 72(6): 679-90, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12774308

ABSTRACT

Injury to the nervous system results in reactive astrogliosis that is a critical determinant of neuronal regeneration. To analyze glial responses to mechanical injury and the role of the polysialic neural cell adhesion molecule (PSA-NCAM) in this process, we established primary glia cultures from newborn rat cerebral cortex. Scratching a confluent monolayer of primary glial cells resulted in two major events: rapid migration of oligodendrocyte progenitor-like (O-2A) cells into the wounded area and development of polarized morphology of type 1 astrocytes at the wound edge. Migrating O-2A progenitors had a bipolar morphology and exhibited A2B5 and O4 immunolabeling. Once these cells were established inside the wounded area, they lost A2B5 immunoreactivity and differentiated into glial fibrillary acidic protein-positive astrocytes. Migrating O-2A cells expressed PSA-NCAM, but type 1 astrocytes at the wound edge did not. Treatment of wounded cultures with Endo-N, which specifically removes PSA from the surface of cells, resulted in a significant decrease in O-2A cell migration into the wounded area and completely blocked the wound closure. Video time-lapse analysis showed that, in the presence of Endo-N, O-2A cells remained motile and migrated short distances but did not move away from the monolayer. These results demonstrate that O-2A progenitors contribute to reactive astrogliosis in culture and that PSA-NCAM is involved in this process by regulating cell migration.


Subject(s)
Cell Movement/physiology , Neural Cell Adhesion Molecule L1/biosynthesis , Neuroglia/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Sialic Acids/biosynthesis , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gliosis/metabolism , Neuroglia/cytology , Rats , Rats, Sprague-Dawley
7.
EMBO J ; 21(23): 6348-57, 2002 Dec 02.
Article in English | MEDLINE | ID: mdl-12456642

ABSTRACT

Mutations in the L1 gene induce a spectrum of human neurological disorders due to abnormal development of several brain structures and fiber tracts. Among its binding partners, L1 immunoglobulin superfamily adhesion molecule (Ig CAM) associates with neuropilin-1 (NP-1) to form a semaphorin3A (Sema3A) receptor and soluble L1 converts Sema3A-induced axonal repulsion into attraction. Using L1 constructs containing missense pathological mutations, we show here that this reversion is initiated by a specific trans binding of L1 to NP-1, but not to L1 or other Ig CAMs, and leads to activation of the NO/cGMP pathway. We identified the L1-NP-1-binding site in a restricted sequence of L1 Ig domain 1, as a peptide derived from this region could reverse Sema3A repulsive effects. A pathological L1 missense mutation located in this sequence specifically disrupts both L1-NP-1 complex formation and Sema3A reversion, suggesting that the cross-talk between L1 and Sema3A might participate in human brain development.


Subject(s)
Axons/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neuropilin-1/metabolism , Semaphorin-3A/metabolism , Animals , Cerebral Cortex/metabolism , Cyclic GMP/metabolism , Mice , Mutation , Neural Cell Adhesion Molecule L1/genetics , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Semaphorin-3A/pharmacology
8.
Neuroendocrinology ; 74(5): 335-46, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11694765

ABSTRACT

The neural cell adhesion molecule (NCAM) and its polysialylated isoform (PSA-NCAM) have been shown to influence the proliferation, differentiation and survival of different cell types. Here, we report the pattern of expression of NCAM and PSA-NCAM in the anterior lobe (AL) of the pituitary gland of the adult mouse. We demonstrate that the majority of cells express NCAM, while PSA-NCAM is retained mostly on corticotropes. Analysis of bromodeoxyuridine (BrdU) incorporation shows that the presence of PSA-NCAM on corticotropes is not related to proliferation but most likely to their functional properties. We subsequently analyzed defects induced by NCAM deficiency in adult NCAM knockout mice. In these mice, all secretory cell types in the AL are present and their distribution within the gland is similar to that in wild-type mice. However, proliferation of AL cells is significantly increased. In particular, more BrdU-positive cells are detected among somatotropes and mammotropes in NCAM-deficient mice. In addition, the percentages of secretory cells are changed: somatotropes are more numerous while the number of corticotropes is reduced. These data demonstrate the involvement of NCAM in the proper generation and/or maintenance of the different cell populations in the AL and suggest the importance of PSA in corticotrope functioning.


Subject(s)
Mutation/physiology , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/genetics , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Animals , Cell Count , Cell Differentiation/physiology , Growth Hormone/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/metabolism , Prolactin/metabolism , Reference Values , Sialic Acids/metabolism
9.
Eur J Neurosci ; 14(8): 1194-202, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11703448

ABSTRACT

Here we show a dual role of N-methyl-d-aspartate receptor (NMDAR) activation in controlling polysialylated neural cell adhesion molecule (PSA-NCAM) dynamic expression in the dorsal vagal complex (DVC), a gateway for many primary afferent fibres. In this structure the overall expression of PSA-NCAM decreases during the first 2 weeks after birth to persist only at synapses in the adult. Electrical stimulation of the vagal afferents causes a rapid increase of PSA-NCAM expression both in vivo and in acute slices before postnatal day (P) 14 whereas a similar stimulation induces a decrease after P15. Inhibition of NMDAR activity in vitro completely prevented these changes. These regulations depend on calmodulin activation and cGMP production at all stages. By contrast, blockade of neuronal nitric oxide synthase (nNOS) prevented these changes only after P10 in agreement with its late expression in the DVC. The pivotal role of NMDAR is also supported by the observation that chronic blockade induces a dramatic decrease in PSA-NCAM expression.


Subject(s)
Aging/physiology , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sialic Acids/metabolism , Solitary Nucleus/growth & development , Vagus Nerve/growth & development , Visceral Afferents/growth & development , Animals , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Free Radical Scavengers/pharmacology , GABA-A Receptor Antagonists , Glutamic Acid/metabolism , Immunohistochemistry , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Vagus Nerve/cytology , Vagus Nerve/metabolism , Visceral Afferents/cytology , Visceral Afferents/metabolism
10.
Infect Immun ; 69(11): 7130-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11598089

ABSTRACT

The capsular polysaccharide of group B Neisseria meningitidis is composed of a linear homopolymer of alpha(2-8) N-acetyl neuraminic acid or polysialic acid (PSA) that is also carried by isoforms of the mammalian neural cell adhesion molecule (NCAM), which is especially expressed on brain cells during development. Here we analyzed the ability of antibodies induced by the candidate vaccine N-propionyl polysaccharide tetanus toxoid conjugate to recognize PSA-NCAM. We hyperimmunized mice to produce a pool of antisera and a series of immunoglobulin G monoclonal antibodies and evaluated their self-reactivity profile by using a battery of tests (immunoprecipitation, immunoblotting, and immunofluorescence detection on live cells and human tissue sections) chosen for their sensitivity and specificity to detect PSA-NCAM in various environments. We also searched for the effects of the vaccine-induced antibodies in two functional assays involving cell lysis or cell migration. Although they were highly bactericidal, all the antibodies tested showed very low or no recognition of PSA-NCAM, in contrast to PSA-specific monoclonal antibodies used as controls. Different patterns of cross-reactions were revealed by the tests used, likely due to affinity and specificity differences among the populations of induced antibodies. Furthermore, neither cell lysis nor perturbation of migration was observed in the presence of the tested antibodies. Importantly, we showed that whereas enzymatic removal of PSA groups from the surfaces of live cells perturbed their migration, blocking them with PSA-specific antibodies was not functionally detrimental. Taken together, our data indicated that this candidate vaccine induced antibodies that could not demonstrate an immunopathologic effect.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Meningococcal Vaccines/immunology , Neisseria meningitidis/immunology , Neural Cell Adhesion Molecules/immunology , Polysaccharides, Bacterial/immunology , Sialic Acids/immunology , Vaccines, Conjugate/immunology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Bacterial Capsules , Cross Reactions , Female , Humans , Mice , Vaccination
11.
J Neurochem ; 78(4): 767-78, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11520897

ABSTRACT

Neural cell adhesion molecule (NCAM) and F3 are both axonal adhesion molecules which display homophilic (NCAM) or heterophilic (NCAM, F3) binding activities and participate in bidirectional exchange of information between neurones and glial cells. Engineered Fc chimeric molecules are fusion proteins that contain the extracellular part of NCAM or F3 and the Fc region of human IgG1. Here, we investigated the effect of NCAM-Fc and F3-Fc chimeras on Schwann cell (SC) migration. Binding sites were identified at the surface of cultured SCs by chimera coated fluorospheres. The functional effect of NCAM-Fc and F3-Fc binding was studied in two different SC migration models. In the first, migration is monitored at specific time intervals inside a 1-mm gap produced in a monolayer culture of SCs. In the second, SCs from a dorsal root ganglion explant migrate on a sciatic nerve cryosection. In both systems addition of the chimeras significantly increased the extent of SC migration and this effect could be prevented by the corresponding anti-NCAM or anti-F3 blocking antibodies. Furthermore, antiproteoglycan-type protein tyrosine phosphatase zeta/beta (RPTPzeta/beta) antibodies identified the presence of RPTPzeta/beta on SCs and prevented the enhancing effect of soluble F3 on SC motility by 95%. The F3-Fc coated Sepharose beads precipitated RPTPzeta/beta from SC lysates. Altogether these data point to RPTPzeta/beta is the putative F3 receptor on SCs. These results identify F3 and NCAM receptors on SC as potential mediators of signalling occurring between axons and glial cells during peripheral nerve development and regeneration.


Subject(s)
Axons/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/physiology , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Protein Tyrosine Phosphatases/metabolism , Schwann Cells/physiology , Animals , Binding Sites , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Cell Communication , Cell Fractionation , Cells, Cultured , Contactins , Fluorescent Dyes , Ganglia, Spinal/cytology , Humans , In Vitro Techniques , Isoenzymes/metabolism , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/genetics , Precipitin Tests , Protein Tyrosine Phosphatases/genetics , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Schwann Cells/metabolism , Sciatic Nerve/physiology , Signal Transduction
12.
J Neurosci ; 21(13): 4721-30, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11425899

ABSTRACT

Here we report that synapses in the adult dorsal vagal complex, a gateway for many primary afferent fibers, express a high level of the polysialylated neural cell adhesion molecule (PSA-NCAM). We show that electrical stimulation of the vagal afferents causes a rapid decrease of PSA-NCAM expression both in vivo and in acute slices. Inhibition of NMDA receptor activity completely prevented the decrease. Blockade of calmodulin activation, neuronal nitric oxide (NO) synthase, or soluble guanylyl cyclase and chelation of extracellular NO mimicked this inhibition. Our data provide a mechanistic framework for understanding how activity-linked stimulation of the NMDA-NO-cGMP pathway induces rapid changes in PSA-NCAM expression, which may be associated with long-term depression.


Subject(s)
Brain Stem/metabolism , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/biosynthesis , Nitric Oxide Synthase/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sialic Acids/biosynthesis , Synapses/metabolism , Animals , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Chelating Agents/pharmacology , Cyclic GMP/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , GAP-43 Protein/biosynthesis , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , In Vitro Techniques , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Neurons, Afferent/physiology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Transduction/physiology , Vagus Nerve/physiology
13.
Oncogene ; 20(8): 997-1004, 2001 Feb 22.
Article in English | MEDLINE | ID: mdl-11314035

ABSTRACT

PSA is an oncodevelopmental antigen usually expressed in human tumors with high metastatic potential. Here we set up a metastatic model in nude mice by using TE671 cells, which strongly express PSA-NCAM. We observed the formation of lung metastases when TE671 cells were injected intravenously, intramuscularly, and intraperitoneally, but not subcutaneously. Intraperitoneal injections also induced peritoneal carcinosis, ascites, and liver metastases. To evaluate the putative role of PSA in the metastatic process we used a specific cleavage of PSA on NCAM by endoneuraminidase-N on intraperitoneal primary tumors. Mice with primary intramuscular tumors were taken as control. Repeated injections of endoneuraminidase-N led to a decrease in PSA expression in primary intraperitoneal nodules and ascites but not in intramuscular primary tumors. Endoneuraminidase-N also increased the delay in ascitic formation and decreased the number of lung or liver metastases in the case of intraperitoneal tumors but not in the case of intramuscular tumors. When metastases occurred in endoneuraminidase-N injected animals, they strongly expressed PSA-NCAM. Therefore, we established a relationship between PSA expression on the surface of primary tumor cells and the metastatic process.


Subject(s)
Lung Neoplasms/secondary , Neoplasm Metastasis/physiopathology , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/metabolism , Rhabdomyosarcoma/secondary , Sialic Acids/metabolism , Animals , Ascites , Disease Models, Animal , Glycoside Hydrolases/metabolism , Humans , Mice , Mice, Nude
14.
Eur J Neurosci ; 13(7): 1391-402, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11298800

ABSTRACT

We show that the loss or inactivation of the polysialic acid (PSA) tail of neural cell adhesion molecule (NCAM) on rat cortical neurons in culture leads to reduced differentiation and survival. The mechanism by which this negative effect is mediated appears to involve the neuronal response to brain-derived neurotrophic factor (BDNF): (i) in the absence of PSA or in the presence of excess free PSA added to the culture medium, BDNF-induced cell signalling is reduced; (ii) the addition of exogenous BDNF to the medium reverses the effect of PSA loss or inactivation. These data suggest that PSA-NCAM, previously shown to modulate cell migration and plasticity, is needed for an adequate sensitivity of neurons to BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Cerebral Cortex/cytology , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/metabolism , Neurons/cytology , Sialic Acids/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Death/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Glycoside Hydrolases/metabolism , Humans , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/pharmacology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhabdomyosarcoma , Sialic Acids/genetics , Sialic Acids/pharmacology , Signal Transduction/physiology , Transfection , Tumor Cells, Cultured
15.
Mol Cell Neurosci ; 17(3): 561-76, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11273650

ABSTRACT

In vertebrates, interneurons of the olfactory bulb are continuously generated postnatally and throughout life at the subventricular zone of the forebrain. From there, the neuronal progenitors migrate tangentially in a typical chain-like structure to the olfactory bulb in which they differentiate as interneurons. We have used a mouse/chick xenograft strategy to explore the migration and differentiation potential of the mouse olfactory progenitors in a heterochronic and heterotypic environment. We compared the migration of primary cells derived from the subventricular zone of adult or newborn lateral ventricule with the behavior of in vitro amplified cells derived from the same structures. We show that in the chick environment, olfactory bulb progenitors from newborn brain tissue perform chain migration along the neural crest cell routes, whereas grafted neurosphere-derived-cells migrate as isolated cells. These results, together with in vitro observations, allow us to propose that neuronal chain migration is a community effect independent of environmental cues but which is closely regulated by the differentiation program of the cells. We established that the progenitor cells performing chain migration are already committed, while neurosphere-derived-cells are able to integrate and differentiate as components of the peripheral nervous system.


Subject(s)
Brain Tissue Transplantation , Interneurons/cytology , Interneurons/transplantation , Nerve Tissue Proteins , Neural Cell Adhesion Molecule L1 , Olfactory Bulb/cytology , Stem Cell Transplantation , Stem Cells/cytology , Age Factors , Animals , Animals, Newborn , Cell Differentiation/physiology , Cell Movement/physiology , Chick Embryo , Graft Survival/physiology , Intermediate Filament Proteins/analysis , Interneurons/chemistry , Mammals , Mice , Mice, Inbred Strains , Nestin , Neural Cell Adhesion Molecules/analysis , Sialic Acids/analysis , Stem Cells/chemistry , Transplantation, Heterologous , Tubulin/analysis , Tyrosine 3-Monooxygenase/analysis , Vimentin/analysis
16.
Neuron ; 27(2): 237-49, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10985345

ABSTRACT

In humans, defects of the corticospinal tract have been attributed to mutations in the gene encoding L1 CAM, a phenotype that is reproduced in L1-deficient mice. Using coculture assays, we report that Sema3A secreted from the ventral spinal cord repels cortical axons from wild-type but not from L1-deficient mice. L1 and neuropilin-1 (NP-1) form a stable complex, and their extracellular domains can directly associate. Thus, L1 is a component of the Sema3A receptor complex, and L1 mutations may disrupt Sema3A signaling in the growth cone, leading to guidance errors. Addition of soluble L1Fc chimeric molecules does not restore Sema3A responsiveness of L1-deficient axons; instead, it converts the repulsion of wild-type axons into an attraction, further supporting a function for L1 in the Sema3A transducing pathways within the growth cone.


Subject(s)
Axons/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Signal Transduction/physiology , Animals , Axons/drug effects , Cell Communication/genetics , Cell Communication/physiology , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Glycoproteins/pharmacology , Growth Cones/drug effects , Growth Cones/metabolism , Leukocyte L1 Antigen Complex , Macromolecular Substances , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Neural Cell Adhesion Molecules/genetics , Neuropilin-1 , Precipitin Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Semaphorin-3A , Signal Transduction/genetics , Spinal Cord/cytology , Spinal Cord/metabolism
17.
Proc Natl Acad Sci U S A ; 97(13): 7585-90, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10840047

ABSTRACT

Many factors have been shown to promote myelination, but few have been shown to be inhibitory. Here, we show that polysialylated-neural cell adhesion molecule (PSA-NCAM) can negatively regulate myelin formation. During development, PSA-NCAM is first expressed on all growing fibers; then, axonal expression is down-regulated and myelin deposition occurs only on PSA-NCAM-negative axons. Similarly, in cocultures of oligodendrocytes and neurons, PSA-NCAM expression on axons is initially high, but decreases as myelination proceeds. Importantly, if expression of PSA-NCAM is prematurely decreased in cultures, by either antibody-mediated internalization or enzymatic removal of the PSA moieties with endoneuraminidase N (endo-N), myelination increases 4- to 5-fold. In the optic nerve, premature cleavage of PSA moieties by intravitreous injection of endo-N also induces a transient increase in the number of myelinated internodes, but does not interfere with the onset of myelination. Previously, we showed that axonal electrical activity strongly induced myelination, which could be prevented by tetrodotoxin (TTX), an action potential blocker. Interestingly, removal of PSA moieties does not reverse the inhibition of myelination by TTX. Together, this suggests that myelination is tightly controlled by both positive (electrical activity) and negative (PSA-NCAM expression) regulatory signals.


Subject(s)
Central Nervous System/physiology , Myelin Sheath/metabolism , Neural Cell Adhesion Molecules/metabolism , Animals , Cells, Cultured , Down-Regulation , Membrane Potentials , Mice , Neurons/physiology
18.
Proc Natl Acad Sci U S A ; 97(8): 4315-20, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10760298

ABSTRACT

The neural cell adhesion molecule (NCAM) and its polysialylated form (PSA-NCAM) contribute to long-term potentiation (LTP) in the CA1 hippocampus. Here we report that the deficient LTP found in slices prepared from NCAM knockout mice and in organotypic slice cultures treated with Endo-N, an enzyme that cleaves the PSA moiety of NCAM, can be rescued by brain-derived neurotrophic factor (BDNF). This effect is not reproduced by nerve growth factor, but can be obtained with high concentrations of NT4/5. The effect of BDNF cannot be accounted for by modifications of N-methyl-D-aspartate receptor-dependent responses or of high-frequency bursts. PSA-NCAM, however, could directly interact with BDNF. Exogenous application of PSA residues or recombinant PSA-NCAM also prevents LTP. Furthermore trkB phosphorylation, and thus BDNF signaling, is reduced in both NCAM knockout mice and Endo-N-treated slice cultures. These results suggest that one action of PSA-NCAM could be to sensitize pyramidal neurons to BDNF, thereby modulating activity-dependent synaptic plasticity.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Neural Cell Adhesion Molecules/genetics , Animals , Culture Techniques , Hippocampus/metabolism , Mice , Mice, Knockout , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor, trkB/metabolism
19.
J Cell Biol ; 149(2): 491-502, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10769038

ABSTRACT

Paranodin/contactin-associated protein (caspr) is a transmembrane glycoprotein of the neurexin superfamily that is highly enriched in the paranodal regions of myelinated axons. We have investigated the role of its association with F3/contactin, a glycosylphosphatidyl inositol (GPI)-anchored neuronal adhesion molecule of the Ig superfamily. Paranodin was not expressed at the cell surface when transfected alone in CHO or neuroblastoma cells. Cotransfection with F3 resulted in plasma membrane delivery of paranodin, as analyzed by confocal microscopy and cell surface biotinylation. The region that mediates association with paranodin was mapped to the Ig domains of F3 by coimmunoprecipitation experiments. The association of paranodin with F3 allowed its recruitment to Triton X-100-insoluble microdomains. The GPI anchor of F3 was necessary, but not sufficient for surface expression of paranodin. F3-Ig, a form of F3 deleted of the fibronectin type III (FNIII) repeats, although GPI-linked and expressed at the cell surface, was not recovered in the microdomain fraction and was unable to promote cell surface targeting of paranodin. Thus, a cooperative effect between the GPI anchor, the FNIII repeats, and the Ig regions of F3 is required for recruitment of paranodin into lipid rafts and its sorting to the plasma membrane.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Membrane Glycoproteins/metabolism , Neuropeptides/metabolism , Animals , CHO Cells , COS Cells , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Cell Membrane/metabolism , Contactins , Cricetinae , Fluorescent Antibody Technique, Indirect , Glycosylphosphatidylinositols/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Neuroblastoma , Neuropeptides/chemistry , Neuropeptides/genetics , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid , Sequence Deletion , Transfection , Tumor Cells, Cultured
20.
Int J Dev Neurosci ; 18(2-3): 213-20, 2000.
Article in English | MEDLINE | ID: mdl-10715576

ABSTRACT

The Neural Cell Adhesion Molecule (NCAM) serves as a temporally and spatially regulated modulator of a variety of cell-cell interactions. This review summarizes recent results of studies aimed at understanding its regulation of expression and biological function, thereby focussing on its polysialylated isoforms (PSA-NCAM). The detailed analysis of the expression of PSA and NCAM in the hippocampal mossy fiber system and the morphological consequences of PSA-NCAM deficiency in mice support the notion that the levels of expression of NCAM are important not only for the regulation and maintenance of structural changes, such as migration, axonal growth and fasciculation, but also for activity-induced plasticity. There is evidence that PSA-NCAM can specifically contribute to a presynaptic form of plasticity, namely long-term potentiation at hippocampal mossy fiber synapses. This is consistent with previous observations that NCAM-deficient mice show deficits in spatial learning and exploratory behavior. Furthermore, our data points to an important role of the hypothalamic-pituitary-adrenal axis, which is the principle adaptive response of the organism to environmental challenges, in the control of PSA-NCAM expression in the hippocampal formation. In particular, we evidence an inhibitory influence of corticosterone on PSA-NCAM expression.


Subject(s)
Hippocampus/physiology , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/physiology , Neuronal Plasticity/physiology , Sialic Acids/physiology , Animals , Hippocampus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...