Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
1.
J Biol Chem ; 296: 100372, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33548223

RESUMEN

Neural cell adhesion molecules 1 (NCAM1) and 2 (NCAM2) belong to the cell adhesion molecules of the immunoglobulin superfamily and have been shown to regulate formation, maturation, and maintenance of synapses. NCAM1 and NCAM2 undergo proteolysis, but the identity of all the proteases involved and how proteolysis is used to regulate their functions are not known. We report here that NCAM1 and NCAM2 are BACE1 substrates in vivo. NCAM1 and NCAM2 overexpressed in HEK cells were both cleaved by metalloproteinases or BACE1, and NCAM2 was also processed by γ-secretase. We identified the BACE1 cleavage site of NCAM1 (at Glu 671) and NCAM2 (at Glu 663) using mass spectrometry and site-directed mutagenesis. Next, we assessed BACE1-mediated processing of NCAM1 and NCAM2 in the mouse brain during aging. NCAM1 and NCAM2 were cleaved in the olfactory bulb of BACE1+/+ but not BACE1-/- mice at postnatal day 10 (P10), 4 and 12 months of age. In the hippocampus, a BACE1-specific soluble fragment of NCAM1 (sNCAM1ß) was only detected at P10. However, we observed an accumulation of full-length NCAM1 in hippocampal synaptosomes in 4-month-old BACE1-/- mice. We also found that polysialylated NCAM1 (PSA-NCAM1) levels were increased in BACE1-/- mice at P10 and demonstrated that BACE1 cleaves both NCAM1 and PSA-NCAM1 in vitro. In contrast, we did not find evidence for BACE1-dependent NCAM2 processing in the hippocampus at any age analyzed. In summary, our data demonstrate that BACE1 differentially processes NCAM1 and NCAM2 depending on the region of brain, subcellular localization, and age in vivo.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Antígeno CD56/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Secretasas de la Proteína Precursora del Amiloide/fisiología , Animales , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/fisiología , Encéfalo/metabolismo , Antígeno CD56/fisiología , Moléculas de Adhesión Celular/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neuronas/metabolismo , Ácidos Siálicos/metabolismo , Análisis Espacio-Temporal , Sinapsis/metabolismo
2.
Pharmacol Res ; 160: 105186, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32898689

RESUMEN

Neuroplastic alterations are the key processes involved in adaptation and rehabilitation after all neurological injuries and pathologies. Being the central contributor to the developmental and adult neuroplasticity, the polysialylated form of Neural Cell Adhesion Molecule (PSA-NCAM) may prove to be a potential target to facilitate repair/regeneration after CNS injury and disease. Over the years, several experimental approaches have been developed to exploit the therapeutic potential of PSA-NCAM. Broadly, the studies focused on cell-transplantation strategies to alter PSA-NCAM properties at the injury site, injection of peptide based as well as synthetic PSA mimetics directly into the injury site or the application of PSA containing hydrogels and scaffolds as biomaterials. A comprehensive understanding of the PSA-based experimental approaches, as well as their pros and cons, is urgently required for successful implementation of this molecule in therapeutics. The current review, therefore, has been designed to give the readers a thorough account of all the diverse roles of PSA in the adult nervous system and the recent progress that has been made in developing PSA-based therapeutic approaches for neuroregeneration.


Asunto(s)
Moléculas de Adhesión de Célula Nerviosa/fisiología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Plasticidad Neuronal/fisiología , Ácidos Siálicos/farmacología , Animales , Humanos , Regeneración Nerviosa/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/genética
3.
Brain Res Bull ; 155: 92-101, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31812781

RESUMEN

The posterodorsal medial amygdala (MePD) has a high concentration of receptors for gonadal hormones, is a sexually dimorphic region and dynamically controls the reproductive behavior of both males and females. Neurotrophic factors can promote dendritic spine remodeling and change synaptic input strength in a region-specific manner. Here, we analyzed the gene and protein expression of brain-derived neurotrophic factor (BDNF), insulin-like growth factor-I (IGF-1), polysialylated neural cell adhesion molecule (PSA-NCAM) and Ephrin-A4 in the MePD of adult males and females in diestrus, proestrus and estrus using real-time qPCR and fluorescent immunohistochemistry. The first approach showed their amplification except for Igf1 and the latter revealed that BDNF, IGF-1, PSA-NCAM and Ephrin-A4 are expressed in the MePD of the adult rats. Protein expression of these neurotrophic factors showed no differences between groups. However, proestrus females displayed a higher number of labelled puncta than males for BDNF expression and diestrus females for IGF-1 expression. In conjunction, results indicate that IGF-1 might be released rather than synthetized in the MePD, and the expression of specific neurotrophic factors varies specifically during proestrus. The dynamic modulation of BDNF and IGF-1 during this cyclic phase is coincident with synaptic changes and spine density remodeling in the MePD, the disinhibition of gonadotrophin secretion for ovulation and the display of sexual behavior.


Asunto(s)
Complejo Nuclear Corticomedial/fisiología , Ciclo Estral , Factores de Crecimiento Nervioso/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/fisiología , Efrina-A4/análisis , Efrina-A4/fisiología , Femenino , Expresión Génica , Masculino , Moléculas de Adhesión de Célula Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Ratas Wistar , Caracteres Sexuales
4.
J Neurosci ; 37(41): 9828-9843, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-28871037

RESUMEN

The proper formation of synapses-specialized unitary structures formed between two neurons-is critical to mediating information flow in the brain. Synaptic cell adhesion molecules (CAMs) are thought to participate in the initiation of the synapse formation process. However, in vivo functional analysis demonstrates that most well known synaptic CAMs regulate synaptic maturation and plasticity rather than synapse formation, suggesting that either CAMs work synergistically in the process of forming synapses or more CAMs remain to be found. By screening for unknown CAMs using a co-culture system, we revealed that protein tyrosine phosphatase receptor type O (PTPRO) is a potent CAM that induces the formation of artificial synapse clusters in co-cultures of human embryonic kidney 293 cells and hippocampal neurons cultured from newborn mice regardless of gender. PTPRO was enriched in the mouse brain and localized to postsynaptic sites at excitatory synapses. The overexpression of PTPRO in cultured hippocampal neurons increased the number of synapses and the frequency of miniature EPSCs (mEPSCs). The knock-down (KD) of PTPRO expression in cultured neurons by short hairpin RNA (shRNA) reduced the number of synapses and the frequencies of the mEPSCs. The effects of shRNA KD were rescued by expressing either full-length PTPRO or a truncated PTPRO lacking the cytoplasmic domain. Consistent with these results, the N-terminal extracellular domain of PTPRO was required for its synaptogenic activity in the co-culture assay. Our data show that PTPRO is a synaptic CAM that serves as a potent initiator of the formation of excitatory synapses.SIGNIFICANCE STATEMENT The formation of synapses is critical for the brain to execute its function and synaptic cell adhesion molecules (CAMs) play essential roles in initiating the formation of synapses. By screening for unknown CAMs using a co-culture system, we revealed that protein tyrosine phosphatase receptor type O (PTPRO) is a potent CAM that induces the formation of artificial synapse clusters. Using loss-of-function and gain-of-function approaches, we show that PTPRO promotes the formation of excitatory synapses. The N-terminal extracellular domain of PTPRO was required for its synaptogenic activity in cultured hippocampal neurons and the co-culture assay. Together, our data show that PTPRO is a synaptic CAM that serves as a potent initiator of synapse formation.


Asunto(s)
Moléculas de Adhesión de Célula Nerviosa/fisiología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/fisiología , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Técnicas de Cocultivo , Potenciales Postsinápticos Excitadores/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Moléculas de Adhesión de Célula Nerviosa/genética , Técnicas de Placa-Clamp , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética
5.
Dev Biol ; 419(2): 285-297, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27618756

RESUMEN

Collective cell migration is the coordinated movement of cells, which organize tissues during morphogenesis, repair and some cancers. The motile cell membrane of the advancing front in collective cell migration is termed the Leading Edge. The embryonic development of the vertebrate and Drosophila hearts are both characterized by the coordinated medial migration of a bilateral cluster of mesodermal cells. In Drosophila, the cardioblasts form cohesive bilateral rows that migrate collectively as a unit towards the dorsal midline to form the dorsal vessel. We have characterized the collective cell migration of cardioblasts as an in vivo quantitative model to study the behaviour of the Leading Edge. We investigated whether guidance signalling through Slit and Netrin pathways plays a role in cell migration during heart development. Through time-lapse imaging and quantitative assessment of migratory behaviour of the cardioblasts in loss-of-function mutants, we demonstrate that both Slit and Netrin mediated signals are autonomously and concomitantly required to maximize migration velocity, filopodial and lamellipodial activities. Additionally, we show that another Slit and Netrin receptor, Dscam1, the role of which during heart development was previously unknown, is required for both normal migration of cardioblasts and luminal expansion. Leading edge behaviour analysis revealed a dosage dependent genetic interaction between Slit and Netrin receptors suggesting that downstream signalling through these receptors converge on a common output that increases leading edge activity of the cardioblasts. Finally, we found that guidance signalling maintains the balance between epithelial and mesenchymal characteristics of the migrating cardioblasts.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/crecimiento & desarrollo , Corazón/embriología , Miocitos Cardíacos/citología , Seudópodos/fisiología , Células Madre/citología , Animales , Orientación del Axón , Moléculas de Adhesión Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Transición Epitelial-Mesenquimal , Proteínas Luminiscentes/análisis , Morfogénesis , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Receptores de Netrina , Netrina-1 , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/fisiología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores Inmunológicos/genética , Receptores Inmunológicos/fisiología , Transducción de Señal , Imagen de Lapso de Tiempo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología , Proteínas Roundabout
6.
Brain Struct Funct ; 221(3): 1591-605, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25596866

RESUMEN

The neural cell adhesion molecule NCAM and its association with the polysialic acid (PSA) are believed to contribute to brain structural plasticity that underlies memory formation. Indeed, the attachment of long chains of PSA to the glycoprotein NCAM down-regulates its adhesive properties by altering cell-cell interactions. In the brain, the biosynthesis of PSA is catalyzed by two polysialyltransferases, which are differentially regulated during lifespan. One of them, ST8SiaIV (PST), is predominantly expressed during adulthood whereas the other one, ST8SiaII (STX), dominates during embryonic and post-natal development. To understand the role played by ST8SiaIV during learning and memory and its underlying hippocampal plasticity, we used knockout mice deleted for the enzyme ST8SiaIV (PST-ko mice). At adult age, PST-ko mice show a drastic reduction of PSA-NCAM expression in the hippocampus and intact hippocampal adult neurogenesis. We found that these mice display impaired long-term but not short-term memory in both, spatial and non-spatial behavioral tasks. Remarkably, memory deficits of PST-ko mice were abolished by exposure to environmental enrichment that was also associated with an increased number of PSA-NCAM expressing new neurons in the dentate gyrus of these mice. Whether the presence of a larger pool of immature, likely plastic, new neurons favored the rescue of long-term memory in PST-ko mice remains to be determined. Our findings add new evidence to the role played by PSA in memory consolidation. They also suggest that PSA synthesized by PST critically controls the tempo of new neurons maturation in the adult hippocampus.


Asunto(s)
Ambiente Controlado , Hipocampo/enzimología , Memoria/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Sialiltransferasas/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neurogénesis , Plasticidad Neuronal , Sialiltransferasas/genética , Memoria Espacial/fisiología
7.
Lab Invest ; 92(9): 1297-309, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22732936

RESUMEN

Hepatocellular carcinoma (HCC) is a very angiogenic and malignant cancer. Conventional chemotherapy is poorly effective because of the abnormal structural organization of HCC-infiltrating vessels. In previous work, we demonstrated that HCC angiogenesis is driven by transforming growth factor beta-1(TGF-ß1)/CD105 axis, stimulating liver-derived microvascular endothelial cells (Ld-MECs) migration. As TGF-ß1 also affects mural cells (MCs) recruitment and maturation, we asked whether it may contribute to HCC-induced vascular abnormalities. HCC and adjacent non-neoplastic liver (nNL) biopsies obtained from 12 patients were analyzed by immunohistochemistry for angiogenic markers CD105, TGF-ß1, CD44 and vascular endothelial growth factor-a (VEGFa) and for MC markers NG2, α-smooth muscle actin (αSMA) and neural cell adhesion molecule (NCAM). The same markers were also investigated by immunocytochemistry on cultured HCC-derived stromal cells (HCC-StCs) and nNL-derived StCs (nNL-StCs) isolated from the same liver biopsies. Angiogenic factors released by StCs were analyzed by ELISA and the interaction between StCs and Ld-MECs by adhesion assay. Compared with nNL, HCC biopsies showed increased angiogenic markers and αSMA that was localized in vessels. By contrast, NG2 and NCAM were substantially localized in tumor cells but absent in vessels and stroma. Cultured HCC-StCs showed less expression of NG2, αSMA and NCAM. They also demonstrated a lower capacity to release angiogenic factors and adhered on Ld-MECs. HCC-StCs and nNL-StCs treated with TGF-ß1 or with of HepG2 (a human hepatoma cell line) derived conditioned medium (CM), down-modulated NCAM expression, whereas anti-NCAM antibodies significantly reduced the adhesion of StCs to Ld-MECs. By further blocking TGF-ß1 with anti-TGF-ß1 antibodies or with Ly-364947 (a specific inhibitor TGF-ß1-receptor) adhesion to Ld-MECs and NCAM expression respectively was partially restored. TGF-ß1 contributes to HCC-induced vascular alterations by affecting the interaction between HCC-StCs and Ld-MECs through a down-modulation of NCAM expression.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Regulación hacia Abajo , Neoplasias Hepáticas/metabolismo , Microvasos/anomalías , Moléculas de Adhesión de Célula Nerviosa/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Biomarcadores/metabolismo , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/patología , Neovascularización Patológica
8.
Mol Cell Endocrinol ; 348(1): 95-103, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-21846489

RESUMEN

There is abundant evidence to prove that the astrocytes are highly dynamic cell type in CNS and under physiological conditions such as reproduction, these cells display a remarkable structural plasticity especially at the level of their distal processes ensheathing the gonadotropin releasing hormone (GnRH) axon terminals. The morphology of GnRH axon terminals and astrocytes in the median eminence region of hypothalamus show activity dependent structural plasticity during different phases of estrous cycle. In the current study, we have assessed the functional contribution of ∞-2,8-linked polysialic acid (PSA) on neural cell adhesion molecule (PSA-NCAM) in this neuronal-glial plasticity using both in vitro and in vivo model systems. In vivo experiments were carried out after stereotaxic injection of endoneuraminidase enzyme (endo-N) near median eminence region of hypothalamus to specifically remove PSA residues on NCAM followed by localization of GnRH, PSA-NCAM and glial fibrillary acidic protein (GFAP) by immunostaining. Using in vitro model, structural remodeling of GnV-3 cells, (a conditionally immortalized GnRH cell line) co-cultured with primary astrocytes was studied after treating the cells with endo-N. Marked morphological changes were observed in GnRH axon terminals in proestrous phase rats and control GnV-3 cells as compared to endo-N treatment i.e. after removal of PSA. The specificity of endo-N treatment was also confirmed by studying the expression of PSA-NCAM by Western blotting in cultures treated with and without endo-N. Removal of PSA from surfaces with endo-N prevented stimulation associated remodeling of GnRH axon terminals as well as their associated glial cells under both in vivo and in vitro conditions. The current data confirms the permissive role of PSA to promote dynamic remodeling of GnRH axon terminals and their associated glia during reproductive cycle in rats.


Asunto(s)
Glicósido Hidrolasas/farmacología , Hormona Liberadora de Gonadotropina/fisiología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Ácidos Siálicos/metabolismo , Animales , Astrocitos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Ciclo Estral , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Glicósido Hidrolasas/administración & dosificación , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Microinyecciones , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neuroglía/citología , Neuroglía/efectos de los fármacos , Plasticidad Neuronal , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar
9.
Exp Neurol ; 233(2): 866-70, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22200540

RESUMEN

Spinal lamina II, where nociceptive C-fibers terminate, expresses high amounts of the polysialylated form of neural cell adhesion molecule (PSA-NCAM). While enzymatic removal of the PSA moiety from NCAM did not affect normal sensitivity to thermal stimuli, it exacerbated nerve injury-induced neuropathic hyperalgesia. The genetic removal of the NCAM core protein also did not alter thermal sensitivity. However in the presence of a peripheral nerve injury, NCAM-null mutants exhibited a complete suppression of thermal hyperalgesia. This strong NCAM mutant phenotype appears to involve the long form of NCAM's cytoplasmic domain, in that it is duplicated by selective genetic deletion of the NCAM-180 isoform. PSA appears therefore to provide a mechanism for modulation of chronic sensory overload, by means of attenuation of the activity of the NCAM-180 isoform, which reduces nociceptive transmission.


Asunto(s)
Hiperalgesia/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Ácidos Siálicos/fisiología , Animales , Hiperalgesia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Molécula L1 de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Traumatismos de los Nervios Periféricos/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Ácidos Siálicos/genética
10.
PLoS One ; 6(10): e26026, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22043302

RESUMEN

BACKGROUND: Diabetic retinopathy and retinopathy of prematurity are diseases caused by pathological angiogenesis in the retina as a consequence of local hypoxia. The underlying mechanism for epiretinal neovascularization (tuft formation), which contributes to blindness, has yet to be identified. Neural cell adhesion molecule (N-CAM) is expressed by Müller cells and astrocytes, which are in close contact with the retinal vasculature, during normal developmental angiogenesis. METHODOLOGY/PRINCIPAL FINDINGS: Notably, during oxygen induced retinopathy (OIR) N-CAM accumulated on astrocytes surrounding the epiretinal tufts. Here, we show that N-CAM ablation results in reduced vascular tuft formation due to reduced endothelial cell proliferation despite an elevation in VEGFA mRNA expression, whereas retinal developmental angiogenesis was unaffected. CONCLUSION/SIGNIFICANCE: We conclude that N-CAM exhibits a regulatory function in pathological angiogenesis in OIR. This is a novel finding that can be of clinical relevance in diseases associated with proliferative vasculopathy.


Asunto(s)
Retinopatía Diabética/etiología , Hipoxia/complicaciones , Neovascularización Patológica , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neovascularización Retiniana , Animales , Proliferación Celular , Células Endoteliales/patología , Ratones , Moléculas de Adhesión de Célula Nerviosa/análisis , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/patología , Factor A de Crecimiento Endotelial Vascular/genética
11.
C R Biol ; 334(12): 855-62, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22123087

RESUMEN

The desert rodents Psammomys obesus and Gerbillus tarabuli live under extreme conditions and overcome food and water shortage by modes of food and fluid intake specific to each species. Using immunohistochemistry and electron microscopy, we found that the hypothalamic magnocellular nuclei, and in particular, their vasopressinergic component, is highly and similarly developed in Psammomys and Gerbillus. In comparison to other rodents, the hypothalamus in both species contains more magnocellular VP neurons that, together with oxytocin neurons, accumulate in distinct and extensive nuclei. As in dehydrated rodents, many magnocellular neurons contained both neuropeptides. A striking feature of the hypothalamic magnocellular system of Psammomys and Gerbillus was its display of ultrastructural properties related to heightened neurosecretion, namely, a significant reduction in glial coverage of neuronal somata and dendrites in the hypothalamic nuclei. There were many neuronal elements whose surfaces were directly juxtaposed and shared the same synapses. Their magnocellular nuclei also showed a high level of sialylated isoform of the Neural Cell Adhesion Molecule (PSA-NCAM) that underlies their capacity for neuronal and glial plasticity. These species thus offer striking models of structural neuronal and glial plasticity linked to natural conditions of heightened neurosecretion.


Asunto(s)
Núcleo Basal de Meynert/citología , Núcleo Basal de Meynert/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Neuropéptidos/fisiología , Equilibrio Hidroelectrolítico/fisiología , Animales , Núcleo Basal de Meynert/ultraestructura , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Gerbillinae , Inmunohistoquímica , Masculino , Microscopía Electrónica , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neuroglía/ultraestructura , Plasticidad Neuronal/fisiología , Neuronas/ultraestructura , Oxitocina/fisiología , Neurohipófisis/metabolismo , Neurohipófisis/fisiología , Neurohipófisis/ultraestructura , Fijación del Tejido , Vasopresinas/fisiología
12.
Brain Res ; 1374: 27-35, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21167820

RESUMEN

IgLONs are a family of four GPI-anchored cell adhesion molecules that regulate neurite outgrowth, synaptogenesis and may act as tumour suppressor genes. IgLONs are thought to function as monomers or homodimers and we have proposed that IgLONs also act as heterodimeric complexes termed Dimeric IgLONs or DIgLONs. Here we show that the initiation of neurite outgrowth is inhibited from a subset of chick embryonic day (E) 7 or 8 forebrain neurons when they are cultured on CHO cell lines expressing DIgLON:CEPU-1-OBCAM and DIgLON:CEPU-1-LAMP but not on CHO cells that express single IgLONs CEPU-1 or OBCAM. Surprisingly at the younger age of E6 forebrain neurons do not respond to DIgLONs. Since there is little difference in expression of IgLONs on the surface of chick forebrain neurons at these two ages we suggest IgLONs alone are not the receptor on the responding forebrain neurons. A DIgLON heterodimeric recombinant protein DIgLON:CEPU-1-OBCAM-Fc also blocked neurite outgrowth from E8 chick forebrain neurons. However, when IgLONs were removed from the surface of these E8 neurons they no longer responded to DIgLON:CEPU-1-OBCAM-Fc substrate, indicating that IgLONs form at least a component of the neuronal cell receptor complex involved in this inhibition of neurite outgrowth. Inhibitors pertussis toxin and Y27632 reversed the inhibition of neurite outgrowth on a DIgLON:CEPU-1-OBCAM and DIgLON:CEPU-1-LAMP substrate. This suggests the involvement of a G-protein coupled receptor and activation of Rho A. In summary we provide evidence that DIgLON:CEPU-1-OBCAM and DIgLON:CEPU-1-LAMP complexes regulate initiation of neurite outgrowth on forebrain neurons via an IgLON-containing receptor complex.


Asunto(s)
Proteínas Aviares/fisiología , Inhibidores de Crecimiento/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Inhibición Neural/fisiología , Neuritas/fisiología , Prosencéfalo/fisiología , Animales , Células CHO , Línea Celular Tumoral , Embrión de Pollo , Cricetinae , Cricetulus , Inmunoglobulinas/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/fisiología , Ratones , Neuronas/fisiología
13.
J Neurosci ; 30(11): 3995-4003, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20237270

RESUMEN

Cells generated in the subventricular zone give rise to neuroblasts that migrate to the olfactory bulb (OB) along the rostral migratory stream (RMS). The polysialylated form of neural cell adhesion molecule (PSA-NCAM) is expressed by these cells, and has been shown to both promote their migration and suppress differentiation induced by NCAM. In the present study, enzymatic removal of PSA from these neuroblasts using PSA-specific endoneuraminidase has been found not only to disrupt the tangential migration and cellular organization of the RMS, but also to cause a massive dispersion of BrdU (5-bromo-2'-deoxyuridine)-labeled neuroblasts into surrounding brain regions, including cortex and striatum. These dispersed cells are capable of differentiation, some into mature neurons, and could potentially be of value in the repair of CNS injury. Although the removal of PSA by genetic deletion of NCAM also results in a smaller OB and a swollen RMS, the cells do not escape the RMS in large numbers. These findings suggest that the presence of NCAM without PSA plays a role in the dispersion process, possibly by inducing a new pattern of migration associated with NCAM-dependent differentiation.


Asunto(s)
Movimiento Celular/fisiología , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/fisiología , Neuronas/citología , Neuronas/fisiología , Ácidos Siálicos/deficiencia , Células Madre/citología , Células Madre/fisiología , Animales , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/fisiología , Bromodesoxiuridina , Diferenciación Celular/fisiología , Ventrículos Cerebrales/metabolismo , Glicósido Hidrolasas/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/fisiología , Células Madre/metabolismo
14.
J Neurosci ; 30(11): 4171-83, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20237287

RESUMEN

The neural cell adhesion molecule (NCAM) is the predominant carrier of alpha2,8 polysialic acid (PSA) in the mammalian brain. Abnormalities in PSA and NCAM expression are associated with schizophrenia in humans and cause deficits in hippocampal synaptic plasticity and contextual fear conditioning in mice. Here, we show that PSA inhibits opening of recombinant NMDA receptors composed of GluN1/2B (NR1/NR2B) or GluN1/2A/2B (NR1/NR2A/NR2B) but not of GluN1/2A (NR1/NR2A) subunits. Deficits in NCAM/PSA increase GluN2B-mediated transmission and Ca(2+) transients in the CA1 region of the hippocampus. In line with elevation of GluN2B-mediated transmission, defects in long-term potentiation in the CA1 region and contextual fear memory in NCAM/PSA-deficient mice are abrogated by application of a GluN2B-selective antagonist. Furthermore, treatment with the glutamate scavenger glutamic-pyruvic transaminase, ablation of Ras-GRF1 (a mediator of GluN2B signaling to p38 MAPK), or direct inhibition of hyperactive p38 MAPK can restore impaired synaptic plasticity in brain slices lacking PSA/NCAM. Thus, PSA carried by NCAM regulates plasticity and learning by inhibition of the GluN2B-Ras-GRF1-p38 MAPK signaling pathway. These findings implicate carbohydrates carried by adhesion molecules in modulating NMDA receptor signaling in the brain and demonstrate reversibility of cognitive deficits associated with ablation of a schizophrenia-related adhesion molecule.


Asunto(s)
Aprendizaje/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Ácidos Siálicos/fisiología , Animales , Región CA1 Hipocampal/fisiología , Células CHO , Cricetinae , Cricetulus , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
15.
J Am Soc Nephrol ; 21(4): 666-77, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20167703

RESUMEN

The neuronal adhesion protein Dragon acts as a bone morphogenetic protein (BMP) coreceptor that enhances BMP signaling. Given the importance of BMP signaling in nephrogenesis and its putative role in the response to injury in the adult kidney, we studied the localization and function of Dragon in the kidney. We observed that Dragon localized predominantly to the apical surfaces of tubular epithelial cells in the thick ascending limbs, distal convoluted tubules, and collecting ducts of mice. Dragon expression was weak in the proximal tubules and glomeruli. In mouse inner medullary collecting duct (mIMCD3) cells, Dragon generated BMP signals in a ligand-dependent manner, and BMP4 is the predominant endogenous ligand for the Dragon coreceptor. In mIMCD3 cells, BMP4 normally signaled through BMPRII, but Dragon enhanced its signaling through the BMP type II receptor ActRIIA. Dragon and BMP4 increased transepithelial resistance (TER) through the Smad1/5/8 pathway. In epithelial cells isolated from the proximal tubule and intercalated cells of collecting ducts, we observed coexpression of ActRIIA, Dragon, and BMP4 but not BMPRII. Taken together, these results suggest that Dragon may enhance BMP signaling in renal tubular epithelial cells and maintain normal renal physiology.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Células Epiteliales/fisiología , Proteínas del Tejido Nervioso/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Animales , Células Cultivadas , Riñón , Ratones , Transducción de Señal , Urotelio/citología
16.
Haematologica ; 95(6): 884-91, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20015889

RESUMEN

BACKGROUND: We previously established a mesenchymal stem cell line (FMS/PA6-P) from the bone marrow adherent cells of fetal mice. The cell line expresses a higher level of neural cell adhesion molecule and shows greater hematopoiesis-supporting capacity in mice than other murine stromal cell lines. DESIGN AND METHODS: Since there is 94% homology between human and murine neural cell adhesion molecule, we examined whether FMS/PA6-P cells support human hematopoiesis and whether neural cell adhesion molecules expressed on FMS/PA6-P cells contribute greatly to the human hematopoiesis-supporting ability of the cell line. RESULTS: When lineage-negative cord blood mononuclear cells were co-cultured on the FMS/PA6-P cells, a significantly greater hematopoietic stem cell-enriched population (CD34(+)CD38(-) cells) was obtained than in the culture without the FMS/PA6-P cells. Moreover, when lineage-negative cord blood mononuclear cells were cultured on FMS/PA6-P cells and transplanted into SCID mice, a significantly larger proportion of human CD45(+) cells and CD34(+)CD38(-) cells were detected in the bone marrow of SCID mice than in the bone marrow of SCID mice that had received lineage-negative cord blood mononuclear cells cultured without FMS/PA6-P cells. Furthermore, we found that direct cell-to-cell contact between the lineage-negative cord blood mononuclear cells and the FMS/PA6-P cells was essential for the maximum expansion of the mononuclear cells. The addition of anti-mouse neural cell adhesion molecule antibody to the culture significantly inhibited their contact and the proliferation of lineage-negative cord blood mononuclear cells. CONCLUSIONS: These findings suggest that neural cell adhesion molecules expressed on FMS/PA6-P cells play a crucial role in the human hematopoiesis-supporting ability of the cell line.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Línea Celular , Linaje de la Célula/fisiología , Técnicas de Cocultivo , Humanos , Ratones , Ratones Endogámicos ICR , Ratones SCID , Especificidad de la Especie
17.
Neurochem Int ; 53(5): 137-47, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18656513

RESUMEN

Although a large number of signalling cascades are known to be activated downstream of NCAM, only little is known regarding the hierarchical relationship between the involved molecules in the individual cascades and the level of cross talk between the cascades. Here, we evaluated the requirement of putative upstream signalling cascades for the phosphorylation of the kinases extracellular signal-regulated kinase (ERK) and Akt and the transcription factor cyclic adenosine monophosphate (cAMP) response-element binding protein (CREB) following stimulation of NCAM in rat cerebellar granule neurons with an NCAM ligand, the C3d peptide. NCAM-mediated ERK phosphorylation depended on activation of the fibroblast growth factor receptor (FGFR), Src-family kinases, MEK (MAP and ERK kinase) and G(0)/G(i)-proteins, whereas NCAM-mediated CREB phosphorylation depended on the activity of Src-family kinases and MEK. NCAM-specific Akt phosphorylation depended on cyclic guanosine monophosphate (cGMP) and phosphatidylinositide 3-kinase (PI3K). All three phosphorylation events were independent of activation of the signalling molecules phospholipase C, protein kinase C, protein kinase A, and CamKII, which all have been demonstrated previously to be involved in NCAM signalling. For comparison, we also evaluated the role of upstream signalling cascades on fibroblast growth factor 2 (FGF2)-mediated phosphorylation of ERK, Akt, and CREB and found that FGF2 required the activity of both FGFR and Src-family kinases for phosphorylation of ERK, Akt, and CREB. MEK was required for phosphorylation of ERK and CREB, but not Akt, whereas G(0)/G(i)-proteins were necessary for phosphorylation of Akt and CREB, and cGMP was necessary for Akt phosphorylation. We thus demonstrate that even though NCAM and FGF2 have many signalling features in common, and even though both are known to activate FGFR, there are a number of differences in the intracellular signalling network activated by the NCAM ligand C3d and the FGFR ligand FGF2.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Células Cultivadas , Cerebelo/citología , Neuronas/metabolismo , Fosforilación , Ratas , Ratas Wistar , Transducción de Señal
18.
Mol Cell Neurosci ; 37(4): 781-93, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18289872

RESUMEN

Neural cell adhesion molecule, NCAM, is an important regulator of neuronal process outgrowth and synaptic plasticity. Transgenic mice that overexpress the soluble NCAM extracellular domain (NCAM-EC) have reduced GABAergic inhibitory and excitatory synapses, and altered behavioral phenotypes. Here, we examined the role of dysregulated NCAM shedding, modeled by overexpression of NCAM-EC, on development of GABAergic basket interneurons in the prefrontal cortex. NCAM-EC overexpression disrupted arborization of basket cells during the major period of axon/dendrite growth, resulting in decreased numbers of GAD65- and synaptophysin-positive perisomatic synapses. NCAM-EC transgenic protein interfered with interneuron branching during early postnatal stages when endogenous polysialylated (PSA) NCAM was converted to non-PSA isoforms. In cortical neuron cultures, soluble NCAM-EC acted as a dominant inhibitor of NCAM-dependent neurite branching and outgrowth. These findings suggested that excess soluble NCAM-EC reduces perisomatic innervation of cortical neurons by perturbing axonal/dendritic branching during cortical development.


Asunto(s)
Interneuronas/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Corteza Prefrontal/embriología , Corteza Prefrontal/fisiología , Sinapsis/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Células Cultivadas , Humanos , Interneuronas/citología , Ratones , Ratones Transgénicos , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Moléculas de Adhesión de Célula Nerviosa/genética , Corteza Prefrontal/citología , Estructura Terciaria de Proteína/genética , Sinapsis/genética
19.
Dev Dyn ; 237(1): 276-85, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18095350

RESUMEN

Modulation of cell-cell adhesion is crucial for regulating neuronal migration and maintenance of structural plasticity in the embryonic and mature brain. Such modulation can be obtained by the enzymatic attachment of polysialic acid (PSA) to the neural cell adhesion molecule (NCAM) by means of the polysialyltransferases STX and PST. Thus, differential expression of STX and PST is likely to be responsible for varying functions of PSA-NCAM during neuronal differentiation, maintenance, plasticity, and regeneration. We have isolated the zebrafish homologues of STX (St8sia2) and PST (St8sia4) and demonstrate that their expression in the embryonic and adult nervous system is often confined to regions of neuronal migration. Moreover, in the adult cerebellum, the complementary expression pattern of both polysialyltransferases suggests a function in regulating cerebellar neuronal plasticity. Enzymatic removal of PSA in the embryonic cerebellum results in impaired neuronal migration, suggesting that PSA-NCAM is a key regulator of motility for cerebellar neuronal progenitors.


Asunto(s)
Movimiento Celular/genética , Neuronas/metabolismo , Sialiltransferasas/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Adhesión Celular/genética , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Cerebelo/embriología , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neuronas/citología , Neuronas/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ácidos Siálicos/metabolismo , Sialiltransferasas/metabolismo , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA