Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurotrauma ; 37(3): 534-554, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31426714

RESUMEN

L1 cell adhesion molecule (L1CAM) supports spinal cord cellular milieu after contusion and compression lesions, contributing to neuroprotection, promoting axonal outgrowth, and reducing outgrowth-inhibitory molecules in lesion proximity. We extended investigations into L1CAM molecular targets and explored long-distance effects of L1CAM rostral and caudal to complete spinal cord transection (SCT) in adult rats. L1CAM overexpression in neurons and glia after Th10/Th11 SCT was achieved using adeno-associated viral vector serotype 5 (AAV5) injected into an L1-lumbar segment immediately after transection. At 5 weeks, a L1CAM mRNA profound decrease detected rostral and caudal to the transection site was alleviated by AAV5-L1CAM treatment, with increased endogenous L1CAM rostral to the SCT. Transected corticospinal tract fibers showed attenuated retraction after treatment, accompanied by a multi-segmental increase of lesion-reduced expression of adenylate cyclase 1 (Adcy1), synaptophysin, growth-associated protein 43, and myelin basic protein genes caudal to transection, and Adcy1 rostral to transection. In parallel, chondroitin sulfate proteoglycan phosphacan elevated after SCT was downregulated after treatment. Low-molecular L1CAM isoforms generated after spinalization indicated the involvement of sheddases in L1CAM processing and long-distance effects. A disintegrin and metalloproteinase (ADAM)10 sheddase immunoreactivity, stronger in AAV5-L1CAM than AAV5- enhanced green fluorescent protein (EGFP)-transduced motoneurons indicated local ADAM10 upregulation by L1CAM. The results suggest that increased L1CAM availability and penetration of diffusible L1CAM fragments post-lesion induce both local and long-distance neuronal and glial responses toward better neuronal maintenance, neurite growth, and myelination. Despite the fact that intervention promoted beneficial molecular changes, kinematic analysis of hindlimb movements showed minor improvement, indicating that spinalized rats require longer L1CAM treatment to regain locomotor functions.


Asunto(s)
Regulación hacia Abajo/fisiología , Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Plasticidad Neuronal/fisiología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Traumatismos de la Médula Espinal/metabolismo , Regulación hacia Arriba/fisiología , Animales , Expresión Génica , Masculino , Molécula L1 de Adhesión de Célula Nerviosa/genética , Ratas , Ratas Wistar , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Traumatismos de la Médula Espinal/genética , Vértebras Torácicas/lesiones
2.
Mol Neurobiol ; 54(1): 495-510, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26742526

RESUMEN

Pleiotrophin (PTN) and midkine (MK) are secreted growth factors and cytokines, proposed to be significant neuromodulators with multiple neuronal functions. PTN and MK are generally related with cell proliferation, growth, and differentiation by acting through different receptors. PTN or MK, signaling through receptor protein tyrosine phosphatase ß/ζ (RPTPß/ζ), lead to the activation of extracellular signal-regulated kinases (ERKs) and thymoma viral proto-oncogene (Akt), which induce morphological changes and modulate addictive behaviors. Besides, there is increasing evidence that during the development of drug addiction, astrocytes contribute to the synaptic plasticity by synthesizing and releasing substances such as cytokines. In the present work, we studied the effect of acute morphine, chronic morphine, and morphine withdrawal on PTN, MK, and RPTPß/ζ expression and on their signaling pathways in the ventral tegmental area (VTA). Present results indicated that PTN, MK, and RPTPß/ζ levels increased after acute morphine injection, returned to basal levels during chronic opioid treatment, and were upregulated again during morphine withdrawal. We also observed an activation of astrocytes after acute morphine injection and during opiate dependence and withdrawal. In addition, immunofluorescence analysis revealed that PTN, but not MK, was overexpressed in astrocytes and that dopaminergic neurons expressed RPTPß/ζ. Interestingly, p-ERK 1/2 levels during chronic morphine and morphine withdrawal correlated RPTPß/ζ expression. All these observations suggest that the neuroprotective and behavioral adaptations that occur during opiate addiction could be, at least partly, mediated by these cytokines.


Asunto(s)
Proteínas Portadoras/biosíntesis , Citocinas/biosíntesis , Morfina/administración & dosificación , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Esquema de Medicación , Masculino , Midkina , Morfina/efectos adversos , Ratas , Ratas Wistar , Área Tegmental Ventral/efectos de los fármacos
3.
Alcohol Clin Exp Res ; 39(10): 1930-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26332441

RESUMEN

BACKGROUND: Alcohol exposure leads to changes in the extracellular matrix (ECM) in the brain, which profoundly impacts neuronal plasticity. Perineuronal nets (PNs) are specialized ECM structures that enclose subpopulations of neurons in the cortex. Adolescent exposure to alcohol induces long-lasting increases in the expression of PN components in the cortex in adult mice. However, it has not been determined whether binge alcohol exposure in young adults alters PNs. Here, we examined PNs and their core components in the insula and primary motor cortex after repeated binge-like ethanol (EtOH) consumption in adult mice. METHODS: The 4-day drinking in the dark (DID) procedure was performed in mice for 1 or 6 weeks to model binge alcohol consumption. The impact of EtOH drinking on PNs was examined by fluorescent staining of brain sections using a marker for PNs, Wisteria floribunda agglutinin (WFA). In another set of experiments, cortex was dissected and Western blots and real-time quantitative polymerase chain reaction were performed to evaluate the expression of the PN proteins aggrecan, brevican, and phosphacan. RESULTS: Binge-like EtOH drinking for 6 weeks caused a significant increase in PNs in the insula, as measured by WFA binding. Aggrecan, brevican, and phosphacan protein expression, and aggrecan mRNA expression, were also elevated in the insula after 6 weeks of EtOH drinking. In contrast, expression of PN components did not change after 1 week of DID. The increase in PNs appears to be specific to the insula, because alterations were not observed in the primary motor cortex. CONCLUSIONS: Our results provide the first evidence that insular PNs increase after long-term binge drinking. The insula mediates compulsive alcohol use. As PNs influence neuronal firing and plasticity, increased PNs in the insula after multiple binge cycles may contribute to restricted neuronal plasticity and lead to the development of compulsive alcohol use.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Corteza Cerebral/efectos de los fármacos , Etanol/administración & dosificación , Etanol/farmacología , Matriz Extracelular/efectos de los fármacos , Neuronas/efectos de los fármacos , Agrecanos/biosíntesis , Animales , Brevicano/biosíntesis , Corteza Cerebral/metabolismo , Masculino , Ratones , Corteza Motora/efectos de los fármacos , Corteza Motora/metabolismo , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis
4.
Neuromolecular Med ; 16(2): 457-72, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24519742

RESUMEN

To address the role of the transforming growth factor beta (TGFß)-Smad3 signaling pathway in dendrite growth and associated synaptogenesis, we used small inhibitory RNA to knockdown the Smad3 gene in either cultured neurons and or primary astrocytes. We found that TGFß1 treatment of primary neurons increased dendrite extensions and the number of synapsin-1-positive synapses. When Smad3 was knockdown in primary neurons, dendrite growth was inhibited and the number of synapsin-1-positive synapses reduced even with TGFß1 treatment. When astrocyte-conditioned medium (ACM), collected from TGFß1-treated astrocytes (TGFß1-stimulated ACM), was added to cultured neurons, dendritic growth was inhibited and the number of synapsin-1-positive puncta reduced. When TGFß1-stimulated ACM was collected from astrocytes with Smad3 knocked down, this conditioned media promoted the growth of dendrites and the number of synapsin-1-positive puncta in cultured neurons. We further found that TGFß1 signaling through Smad3 increased the expression of chondroitin sulfate proteoglycans, neurocan, and phosphacan in ACM. Application of chondroitinase ABC to the TGFß1-stimulated ACM reversed its inhibitory effects on the dendrite growth and the number of synapsin-1-positive puncta. On the other hand, we found that TGFß1 treatment caused a facilitation of Smad3 phosphorylation and translocation to the nucleus induced by status epilepticus (SE) in wild-type (Smad3(+/+)) mice, and this treatment also caused a promotion of γ-aminobutyric acid-ergic synaptogenesis impaired by SE in Smad3(+/+) as well as in Smad3(-/-) mice, but more dramatic promotion in Smad3(+/+) mice. Thus, we provide evidence for the first time that TGFß-Smad3 signaling pathways within neuron and astrocyte differentially regulate dendrite growth and synaptogenesis, and this pathway may be involved in the pathogenesis of some central nervous system diseases, such as epilepsy.


Asunto(s)
Astrocitos/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Proteína smad3/fisiología , Sinapsis/ultraestructura , Factor de Crecimiento Transformador beta1/fisiología , Transporte Activo de Núcleo Celular , Animales , Astrocitos/efectos de los fármacos , Astrocitos/ultraestructura , Células Cultivadas , Condroitina ABC Liasa/farmacología , Proteoglicanos Tipo Condroitín Sulfato/biosíntesis , Proteoglicanos Tipo Condroitín Sulfato/genética , Medios de Cultivo Condicionados/farmacología , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Neurocano/biosíntesis , Neurocano/genética , Neuronas/ultraestructura , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Proteína smad3/antagonistas & inhibidores , Proteína smad3/deficiencia , Proteína smad3/genética , Estado Epiléptico/metabolismo , Sinapsinas/análisis , Factor de Crecimiento Transformador beta1/farmacología
5.
Int J Cancer ; 135(5): 1101-9, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24496747

RESUMEN

While the cytogenetic and genetic characteristics of childhood acute lymphoblastic leukemias (ALL) are well studied, less clearly understood are the contributing epigenetic mechanisms that influence the leukemia phenotype. Our previous studies and others identified gene mutation (RAS) and DNA methylation (FHIT) to be associated with the most common cytogenetic subgroup of childhood ALL, high hyperdiploidy (having five more chromosomes). We screened DNA methylation profiles, using a genome-wide high-dimension platform of 166 childhood ALLs and 6 normal pre-B cell samples and observed a strong association of DNA methylation status at the PTPRG locus in human samples with levels of PTPRG gene expression as well as with RAS gene mutation status. In the 293 cell line, we found that PTPRG expression induces dephosphorylation of ERK, a downstream RAS target that may be critical for mutant RAS-induced cell growth. In addition, PTPRG expression is upregulated by RAS activation under DNA hypomethylating conditions. An element within the PTPRG promoter is bound by the RAS-responsive transcription factor RREB1, also under hypomethylating conditions. In conclusion, we provide evidence that DNA methylation of the PTPRG gene is a complementary event in oncogenesis induced by RAS mutations. Evidence for additional roles for PTPR family member genes is also suggested. This provides a potential therapeutic target for RAS-related leukemias as well as insight into childhood ALL etiology and pathophysiology.


Asunto(s)
Metilación de ADN/genética , Regulación Leucémica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Activación Transcripcional/genética , Proteínas ras/genética , Transformación Celular Neoplásica/genética , Niño , Preescolar , Proteínas de Unión al ADN/genética , Activación Enzimática , Epigénesis Genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Mutación , Fosforilación/genética , Regiones Promotoras Genéticas , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Factores de Transcripción/genética , Proteínas ras/metabolismo
6.
Neurochem Res ; 38(6): 1285-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624942

RESUMEN

Complex glycan structures and their respective carrier molecules are often expressed in a cell type specific manner. Thus, glycans can be used for the enrichment of specific cell types such as neural precursor cells (NPCs). We have recently shown that the monoclonal antibodies 487(LeX) and 5750(LeX) differentially detect the LewisX (LeX) glycan on NPCs in the developing mouse forebrain. Here, we analysed the staining pattern of both antibodies during late embryonic mouse spinal cord development. At E13.5 both antibodies strongly label the central canal region. Along these lines they detect the LeX glycan primarily on Nestin-positive NPCs at that age. Moreover, we show that spinal cord NPCs cultured as free floating neurospheres display a high immunoreactivity to both antibodies. In that context, we also demonstrate that the 487(LeX) antibody can be used to deplete a subpopulation of neurosphere forming NPCs from a mixed E13.5 spinal cord cell suspension. Towards the end of embryogenesis the overall immunoreactivity to both antibodies increases and the staining appears very diffuse. However, the 5750(LeX) antibody still labels the central canal region. The increase in immunoreactivity correlates with an expression increase of the extracellular matrix molecules Tenascin C and Receptor Protein Tyrosine Phosphatase ß/ζ, two potential LeX carrier proteins. In line with this, immunoprecipitation analyses confirmed Tenascin C as a LeX carrier protein in the embryonic mouse spinal cord. However, the immunoreactivity to both antibodies appears only to be marginally affected in the absence of Tenascin C, arguing against Tenascin C being the major LeX carrier. In conclusion our study gives some novel insights into the complex expression of LeX glycans and potential carrier proteins during the development of the mouse spinal cord.


Asunto(s)
Antígeno Lewis X/biosíntesis , Células-Madre Neurales/metabolismo , Polisacáridos/biosíntesis , Médula Espinal/embriología , Animales , Células Cultivadas , Ratones , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Médula Espinal/metabolismo , Tenascina/biosíntesis
7.
Stem Cells Dev ; 21(18): 3351-62, 2012 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-22834539

RESUMEN

Neurospheres are widely used to propagate and investigate neural stem cells (NSCs) and neural progenitors (NPs). However, the exact cell types present within neurospheres are still unknown. To identify cell types, we used single-cell mRNA profiling of 48 genes in 187 neurosphere cells. Using a clustering algorithm, we identified 3 discrete cell populations within neurospheres. One cell population [cluster unsorted (US) 1] expresses high Bmi1 and Hes5 and low Myc and Klf12. Cluster US2 shows intermediate expression of most of the genes analyzed. Cluster US3 expresses low Bmi1 and Hes5 and high Myc and Klf12. The mRNA profiles of these 3 cell populations correlate with a developmental timeline of early, intermediate, and late NPs, as seen in vivo from the mouse brain. We enriched the cell population for neurosphere-forming cells (NFCs) using morphological criteria of forward scatter (FSC) and side scatter (SSC). FSC/SSC(high) cells generated 2.29-fold more neurospheres than FSC/SSC(low) cells at clonal density. FSC/SSC(high) cells were enriched for NSCs and Lewis-X(+ve) cells, possessed higher phosphacan levels, and were of a larger cell size. Clustering of both FSC/SSC(high) and FSC/SSC(low) cells identified an NFC cluster. Significantly, the mRNA profile of the NFC cluster drew close resemblance to that of early NPs. Taken together, data suggest that the neurosphere culture system can be used to model central nervous system development, and that early NPs are the cell population that gives rise to neurospheres. In future work, it may be possible to further dissect the NFCs and reveal the molecular signature for NSCs.


Asunto(s)
Encéfalo/citología , Células-Madre Neurales/clasificación , ARN Mensajero/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Células Cultivadas , Sistema Nervioso Central/embriología , Perfilación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/biosíntesis , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Complejo Represivo Polycomb 1/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Proteínas Represoras/biosíntesis
8.
Alcohol Clin Exp Res ; 35(10): 1812-21, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21595703

RESUMEN

BACKGROUND: Hypothalamic glial-neuronal communications are important for the activation of luteinizing hormone releasing hormone (LHRH) secretion at the time of puberty. As we have shown that alcohol (ALC) diminishes prepubertal LHRH secretion and delays puberty, we first assessed the effects of short-term ALC administration on the basal expression of a specific gene family involved in glial-neuronal communications. Second, as insulin-like growth factor-1 (IGF-1) is a critical regulator of LHRH secretion and the pubertal process, we then assessed whether IGF-1 could induce the expression of these signaling genes and determine whether ALC can block this affect. METHODS: Immature female rats were fed a liquid diet containing ALC for 6 days beginning when 27 days old. Control animals received either the companion isocaloric liquid diet or rat chow and water. Animals were decapitated on day 33, in the late juvenile stage of development. Medial basal hypothalamic (MBH) tissues were obtained for gene and protein analyses of glial receptor protein tyrosine phosphatase-ß (RPTPß) and the 2 neuronal components, contactin and contactin-associated protein 1 (Caspr1). In the second experiment, IGF-1 was administered into the third ventricle (3V) and the MBH removed 6 hours after peptide delivery, and the above-mentioned 3 genes were analyzed by real-time PCR. To determine whether this action was affected by ALC, immature female rats were administered either ALC (3 g/kg) or water via gastric gavage at 0900 hours. At 1030 hours, the ALC and control groups were subdivided such that half of the animals were injected into the 3V with IGF-1 and the other half with an equal volume of saline. Rats were killed 6 hours after the IGF-1 injection and MBHs collected. RESULTS: Real-time PCR showed that when compared with control animals, ALC caused a marked decrease (p < 0.001) in the basal expression of the RPTPß gene, but did not affect the expression of either contactin or Caspr1. Likewise, analysis by Western blotting demonstrated that ALC caused suppressed (p < 0.001) levels of the RPTPß protein, with the expressions of both contactin and Caspr1 proteins being unaltered. In the second experiment, results showed that only the RPTPß gene was stimulated (p < 0.05) by IGF-1 in the MBH 6 hours after peptide delivery. Assessments revealed that the IGF-1 induced increase (p < 0.01) in the expression of the RPTPß gene was blocked by the presence of ALC. CONCLUSIONS: Prepubertal ALC exposure is capable of interfering with hypothalamic glial-neuronal communications by suppressing the synthesis of the glial product, RPTPß, which is required for binding to the contactin-Caspr1 complex on LHRH neuronal terminals, thus suggesting that this action of ALC contributes to its detrimental effects on the pubertal process.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Hipotálamo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Depresores del Sistema Nervioso Central/metabolismo , Contactina 1/análisis , Contactina 1/biosíntesis , Contactina 1/genética , Etanol/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/biosíntesis , Humanos , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/genética , Hormona Luteinizante/antagonistas & inhibidores , Neuroglía , ARN/análisis , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/análisis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Maduración Sexual/fisiología , Transducción de Señal
9.
Hippocampus ; 21(1): 81-92, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20014386

RESUMEN

This study documents the spatial and temporal expression of three structurally related chondroitin sulfated proteoglycans (CSPGs) during synaptic regeneration induced by brain injury. Using the unilateral entorhinal cortex (EC) lesion model of adaptive synaptogenesis, we documented mRNA and protein profiles of phosphacan and its two splice variants, full length receptor protein tyrosine phosphatase ß (RPTPß) and the short transmembrane receptor form (sRPTPß), at 2, 7, and 15 days postlesion. We report that whole hippocampal sRPTPß protein and mRNA are persistently elevated over the first two weeks after UEC. As predicted, this transmembrane family member was localized adjacent to synaptic sites in the deafferented neuropil and showed increased distribution over that zone following lesion. By contrast, whole hippocampal phosphacan protein was not elevated with deafferentation; however, its mRNA was increased during the period of sprouting and synapse formation (7d). When the zone of synaptic reorganization was sampled using molecular layer/granule cell (ML/GCL) enriched dissections, we observed an increase in phosphacan protein at 7d, concurrent with the observed hippocampal mRNA elevation. Immunohistochemistry also showed a shift in phosphacan distribution from granule cell bodies to the deafferented ML at 2 and 7d postlesion. Phosphacan and sRPTPß were not colocalized with glial fibrillary acid protein (GFAP), suggesting that reactive astrocytes were not a major source of either proteoglycan. While transcript for the developmentally prominent full length RPTPß was also increased at 2 and 15d, its protein was not detected in our adult samples. These results indicate that phosphacan and RPTPß splice variants participate in both the acute degenerative and long-term regenerative phases of reactive synaptogenesis. These results suggest that increase in the transmembrane sRPTPß tyrosine phosphatase activity is critical to this plasticity, and that local elevation of extracellular phosphacan influences dendritic organization during synaptogenesis.


Asunto(s)
Lesiones Encefálicas/metabolismo , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Animales , Western Blotting , Corteza Entorrinal/lesiones , Hipocampo/metabolismo , Inmunohistoquímica , Masculino , Microscopía Confocal , Microscopía Electrónica de Transmisión , Isoformas de Proteínas/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sinapsis/metabolismo
10.
Transl Psychiatry ; 1: e8, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-22832403

RESUMEN

Schizophrenia is a serious and chronic mental disorder, in which both genetic and environmental factors have a role in the development of the disease. Neuregulin-1 (NRG1) is one of the most established genetic risk factors for schizophrenia, and disruption of NRG1 signaling has been reported in this disorder. We reported previously that NRG1/ErbB4 signaling is inhibited by receptor phosphotyrosine phosphatase-ß/ζ (RPTP ß/ζ) and that the gene encoding RPTPß/ζ (PTPRZ1) is genetically associated with schizophrenia. In this study, we examined the expression of RPTPß/ζ in the brains of patients with schizophrenia and observed increased expression of this gene. We developed mice overexpressing RPTPß/ζ (PTPRZ1-transgenic mice), which showed reduced NRG1 signaling, and molecular and cellular changes implicated in the pathogenesis of schizophrenia, including altered glutamatergic, GABAergic and dopaminergic activity, as well as delayed oligodendrocyte development. Behavioral analyses also demonstrated schizophrenia-like changes in the PTPRZ1-transgenic mice, including reduced sensory motor gating, hyperactivity and working memory deficits. Our results indicate that enhanced RPTPß/ζ signaling can contribute to schizophrenia phenotypes, and support both construct and face validity for PTPRZ1-transgenic mice as a model for multiple schizophrenia phenotypes. Furthermore, our results implicate RPTPß/ζ as a therapeutic target in schizophrenia.


Asunto(s)
Trastornos del Conocimiento/genética , Regulación Enzimológica de la Expresión Génica , Fenotipo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Esquizofrenia/genética , Regulación hacia Arriba/genética , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Esquizofrenia/enzimología , Esquizofrenia/metabolismo , Transducción de Señal/genética , Adulto Joven
11.
J Cell Mol Med ; 15(11): 2353-63, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21129151

RESUMEN

Substantial genetic evidence suggests that chromosome 11q is involved in regulating initiation and progression of malignant melanomas. Mutations of the MEN1 gene, located in chromosome 11q13, predispose individuals to the multiple endocrine neoplasia type 1 (MEN1) familial syndrome. MEN1 patients develop primary malignant melanoma, suggesting a potential link between MEN1 syndrome and development of melanomas, but the precise molecular mechanism is poorly understood. Here we show that the MEN1 gene suppresses malignant phenotypes of melanoma cells through multiple signalling pathways. Ectopic expression of menin, the product of MEN1 gene, significantly inhibited melanoma cell proliferation and migration in vitro and in vivo. The inhibition was partly achieved through suppressing expression of growth factor pleiotrophin (PTN) and receptor protein tyrosine phosphatase (RPTP) ß/ζ, accompanied with the reduced expression of phosphatidylinositol 3-kinase (pI3K) and decreased phosphorylation of focal adhesion kinase (FAK) and extracellular signal regulated kinase (ERK1/2). Interestingly, reduced expression of menin was associated with hypermethylation of the CpG islands of the MEN1 promoter in melanoma cells. Taken together, these findings suggest a previously unappreciated function for menin in suppressing malignant phenotypes of melanomas and unravel a novel mechanism involving in regulating PTN signalling by menin in development and progression of melanomas.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Melanoma Experimental/metabolismo , Melanoma/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/biosíntesis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Islas de CpG/genética , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Melanoma/patología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Fosfatidilinositol 3-Quinasa/biosíntesis , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Regiones Promotoras Genéticas , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Transducción de Señal , Trasplante Heterólogo
12.
ASN Neuro ; 2(3): e00039, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20689858

RESUMEN

The heparan sulfate proteoglycan glypican-1, the chondroitin sulfate proteoglycan phosphacan/RPTP (receptor protein-tyrosine phosphatase)-zeta/beta and the extracellular matrix protein tenascin-C were all found to be expressed by neural stem cells and by neural cells derived from them. Expression of proteoglycans and tenascin-C increased after retinoic acid induction of SSEA1-positive ES (embryonic stem) cells to nestin-positive neural stem cells, and after neural differentiation, proteoglycans and tenascin-C are expressed by both neurons and astrocytes, where they surround cell bodies and processes and in certain cases show distinctive expression patterns. With the exception of tenascin-C (whose expression may decrease somewhat), expression levels do not change noticeably during the following 2 weeks in culture. The significant expression, by neural stem cells and neurons and astrocytes derived from them, of two major heparan sulfate and chondroitin sulfate proteoglycans of nervous tissue and of tenascin-C, a high-affinity ligand of phosphacan/RPTP-zeta/beta, indicates that an understanding of their specific functional roles in stem cell neurobiology will be important for the therapeutic application of this new technology in facilitating nervous tissue repair and regeneration.


Asunto(s)
Células Madre Embrionarias/enzimología , Glipicanos/biosíntesis , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Tenascina/biosíntesis , Animales , Bovinos , Línea Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Ligandos , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Células-Madre Neurales/citología , Neuronas/citología , Ratas
13.
Int J Cancer ; 124(8): 1785-93, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19058221

RESUMEN

Pleiotrophin (PTN) is a secreted growth factor involved in angiogenesis and tumor growth. We have recently shown that low concentrations of hydrogen peroxide (HP) stimulate PTN expression, through activation of the transcription factor AP-1. In the present work, we studied the possible involvement of endothelial nitric oxide synthase (eNOS) and the role of nitric oxide (NO) in the regulation of PTN expression, as well as involvement of the latter in the NO-induced human endothelial and prostate cancer cell migration. Inhibition of eNOS or the downstream effector soluble guanylate cyclase (sGC) completely suppressed HP-induced AP-1 activities that lead to PTN expression and cell migration. The NO donor sodium nitroprusside (SNP) through activation of sGC significantly and concentration-dependently increased expression of PTN, through transcriptional activation of the corresponding gene. Moreover, SNP had no effect on the migration of stably transfected prostate cancer cells that do not express PTN and knockdown of PTN receptor protein tyrosine phosphatase beta/zeta (RPTPbeta/zeta) completely abolished SNP-induced cell migration. NO added exogenously or produced endogenously by low concentrations of HP through stimulation of sGC activates extracellular signal-regulated kinase[1/2] (ERK[1/2]) and leads to PTN expression and cell migration. On the other hand, p38, which also intervenes in the up-regulation of PTN expression by low concentrations of HP, seems to act upstream of eNOS and does not intervene in the SNP-induced PTN expression and cell migration. The above data suggest that PTN through its receptor RPTPbeta/zeta is a mediator of the stimulatory effects of eNOS/NO on human endothelial and prostate cancer cell migration.


Asunto(s)
Proteínas Portadoras/biosíntesis , Citocinas/biosíntesis , Células Endoteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Óxido Nítrico/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Guanilato Ciclasa/metabolismo , Humanos , Masculino , Modelos Biológicos
14.
Am J Pathol ; 173(1): 77-92, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18556777

RESUMEN

Ischemic brain injury causes tissue damage and neuronal death. The deficits can often be permanent because adult neurons fail to regenerate. One barrier to neuronal regeneration is the formation of the glial scar, a repair mechanism that is otherwise necessary to seal off necrotic areas. The process of gliosis has been well described, but the mechanisms regulating the robust production of scar components after injury remain poorly understood. Here we show that the early growth response 1 transcriptional factor (Egr-1, also called Krox24, Zif268, and NGFI-A) is expressed in astrocytes in the ventricular wall, corpus callosum, and striatum of normal mouse brain. After experimental stroke caused by permanent occlusion of the middle cerebral artery, Egr-1 was expressed long term in reactive astrocytes that accumulate around the injury site. Gain- and loss-of-function studies in primary astrocytes indicated that Egr-1 regulates the transcription of chondroitin sulfate proteoglycans genes, the main extracellular matrix proteins of the glial scar. Egr-1 bound to a site within the phosphacan promoter and transactivated its expression. Egr-1-deficient mice accumulated lower levels of phosphacan RNA and protein than wild-type mice after stroke, but there were no measurable differences in neurite outgrowth toward the infarct area between the two groups. Our findings suggest that Egr-1 is an important component of the transcriptional network regulating genes involved in gliosis after ischemic injury.


Asunto(s)
Astrocitos/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Regulación de la Expresión Génica/fisiología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Accidente Cerebrovascular/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Técnica del Anticuerpo Fluorescente , Expresión Génica , Gliosis/metabolismo , Gliosis/patología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Ratas , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Accidente Cerebrovascular/patología , Transfección
15.
J Neurosci Res ; 86(6): 1316-23, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18044762

RESUMEN

The accelerated senescence-prone SAMP10 mouse strain is a model for age-dependent neurodegeneration and is characterized by brain atrophy and deficits in learning and memory. Because perineuronal nets play an important role in the synaptic plasticity of adult brains, we examined the distributions of molecules that constitute perineuronal nets in SAMP10 mouse brain samples and compared them with those in control SAMR1 mouse samples. Proteoglycan-related monoclonal antibody 6B4 (MAb6B4) clearly immunostained perineuronal nets in SAMR1 mice cortices, but the corresponding immunostaining in SAMP10 mice was very faint. MAb6B4 recognizes phosphacan/PTPzeta in immature brains. However, this antibody recognized several protein bands, including a 400-kDa core glycoprotein from chondroitin sulfate proteoglycan in homogenates of mature cortices from SAMR1 mice. The 400-kDa band was also recognized by antiaggrecan antibodies. The aggrecan core glycoprotein band was also detectable in samples from SAMP10 mice, but this glycoprotein was faintly immunostained by MAb6B4. Because MAb6B4 recognized the same set of protein bands that the monoclonal antibody Cat-315 recognized in mature cerebral cortices of SAMR1 mice, the MAb6B4 epitope appears to be closely related to that of Cat-315 and presumably represents a novel type of oligosaccharide that attaches to aggrecans. The Cat-315 epitope colocalized with aggrecan in perineuronal nets from SAMR1 mouse brain samples, whereas its expression was prominently reduced in SAMP10 mouse brain samples. The biological significance of the MAb6B4/Cat-315 epitope in brain function and its relationship to the neurodegeneration and learning disabilities observed in SAMP10 mice remain to be elucidated.


Asunto(s)
Agrecanos/biosíntesis , Agrecanos/inmunología , Envejecimiento/fisiología , Anticuerpos Monoclonales , Corteza Cerebral/metabolismo , Red Nerviosa/metabolismo , Agrecanos/química , Animales , Western Blotting , Proteoglicanos Tipo Condroitín Sulfato/química , Proteoglicanos Tipo Condroitín Sulfato/inmunología , Modelos Animales de Enfermedad , Epítopos/biosíntesis , Epítopos/inmunología , Inmunohistoquímica , Ratones , Ratones Endogámicos , Degeneración Nerviosa , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/química , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA