Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ASN Neuro ; 14: 17590914221123138, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36164936

RESUMEN

A major limitation of mechanistic studies in aging brains is the lack of routine methods to robustly visualize and discriminate the cellular distribution of tissue antigens using fluorescent immunohistochemical multi-labeling techniques. Although such approaches are routine in non-aging brains, they are not consistently feasible in the aging brain due to the progressive accumulation of autofluorescent pigments, particularly lipofuscin, which strongly excite and emit over a broad spectral range. Consequently, aging research has relied upon colorimetric antibody techniques, where discrimination of tissue antigens is often challenging. We report the application of a simple, reproducible, and affordable protocol using multispectral light-emitting diodes (mLEDs) exposure for the reduction/elimination of lipofuscin autofluorescence (LAF) in aging brain tissue from humans, non-human primates, and mice. The mLEDs lamp has a broad spectral range that spans from the UV to infrared range and includes spectra in the violet/blue and orange/red. After photo quenching, the LAF level was markedly reduced when the tissue background fluorescence before and after mLEDs exposure was compared (p < 0.0001) across the spectral range. LAF elimination was estimated at 95 ± 1%. This approach permitted robust specific fluorescent immunohistochemical co-visualization of commonly studied antigens in aging brains. We also successfully applied this method to specifically visualize CD44 variant expression in aging human cerebral white matter using RNAscope fluorescent in-situ hybridization. Photo quenching provides an attractive means to accelerate progress in aging research by increasing the number of molecules that can be topologically discriminated by fluorescence detection in brain tissue from normative or pathological aging.


Asunto(s)
Envejecimiento , Encéfalo , Receptores de Hialuranos , Primates , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Encéfalo/metabolismo , Variación Genética , Humanos , Receptores de Hialuranos/genética , Inmunohistoquímica , Hibridación in Situ , Lipofuscina/química , Ratones , Primates/genética
2.
Glia ; 68(2): 263-279, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31490574

RESUMEN

Myelination delay and remyelination failure following insults to the central nervous system (CNS) impede axonal conduction and lead to motor, sensory and cognitive impairments. Both myelination and remyelination are often inhibited or delayed due to the failure of oligodendrocyte progenitor cells (OPCs) to mature into myelinating oligodendrocytes (OLs). Digestion products of the glycosaminoglycan hyaluronan (HA) have been implicated in blocking OPC maturation, but how these digestion products are generated is unclear. We tested the possibility that hyaluronidase activity is directly linked to the inhibition of OPC maturation by developing a novel modified flavonoid that functions as a hyaluronidase inhibitor. This compound, called S3, blocks some but not all hyaluronidases and only inhibits matrix metalloproteinase activity at high concentrations. We find that S3 reverses HA-mediated inhibition of OPC maturation in vitro, an effect that can be overcome by excess recombinant hyaluronidase. Furthermore, we find that hyaluronidase inhibition by S3 accelerates OPC maturation in an in vitro model of perinatal white matter injury. Finally, blocking hyaluronidase activity with S3 promotes functional remyelination in mice with lysolecithin-induced demyelinating corpus callosum lesions. All together, these findings support the notion that hyaluronidase activity originating from OPCs in CNS lesions is sufficient to prevent OPC maturation, which delays myelination or blocks remyelination. These data also indicate that modified flavonoids can act as selective inhibitors of hyaluronidase activity and can promote OPC maturation, making them excellent candidates to accelerate myelination or promote remyelination following perinatal and adult CNS insults.


Asunto(s)
Enfermedades Desmielinizantes/patología , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/metabolismo , Remielinización/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Neurogénesis/fisiología , Células Madre/metabolismo
3.
J Clin Invest ; 128(5): 2025-2041, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29664021

RESUMEN

Cerebral white matter injury (WMI) persistently disrupts myelin regeneration by oligodendrocyte progenitor cells (OPCs). We identified a specific bioactive hyaluronan fragment (bHAf) that downregulates myelin gene expression and chronically blocks OPC maturation and myelination via a tolerance-like mechanism that dysregulates pro-myelination signaling via AKT. Desensitization of AKT occurs via TLR4 but not TLR2 or CD44. OPC differentiation was selectively blocked by bHAf in a maturation-dependent fashion at the late OPC (preOL) stage by a noncanonical TLR4/TRIF pathway that induced persistent activation of the FoxO3 transcription factor downstream of AKT. Activated FoxO3 selectively localized to oligodendrocyte lineage cells in white matter lesions from human preterm neonates and adults with multiple sclerosis. FoxO3 constraint of OPC maturation was bHAf dependent, and involved interactions at the FoxO3 and MBP promoters with the chromatin remodeling factor Brg1 and the transcription factor Olig2, which regulate OPC differentiation. WMI has adapted an immune tolerance-like mechanism whereby persistent engagement of TLR4 by bHAf promotes an OPC niche at the expense of myelination by engaging a FoxO3 signaling pathway that chronically constrains OPC differentiation.


Asunto(s)
Proteína Forkhead Box O3/inmunología , Tolerancia Inmunológica , Células Precursoras de Oligodendrocitos/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Animales , ADN Helicasas/genética , ADN Helicasas/inmunología , Femenino , Proteína Forkhead Box O3/genética , Humanos , Ratones , Ratones Noqueados , Neurregulina-1/genética , Neurregulina-1/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Células Precursoras de Oligodendrocitos/patología , Factor de Transcripción 2 de los Oligodendrocitos/genética , Factor de Transcripción 2 de los Oligodendrocitos/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Receptores Toll-Like/genética , Factores de Transcripción/genética , Factores de Transcripción/inmunología
4.
Dev Biol ; 413(2): 173-87, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27067865

RESUMEN

The Olig2 basic-helix-loop-helix transcription factor promotes oligodendrocyte specification in early neural progenitor cells (NPCs), including radial glial cells, in part by recruiting SWI/SNF chromatin remodeling complexes to the enhancers of genes involved in oligodendrocyte differentiation. How Olig2 expression is regulated during oligodendrogliogenesis is not clear. Here, we find that the Brg1 subunit of SWI/SNF complexes interacts with a proximal Olig2 promoter and represses Olig2 transcription in the mouse cortex at E14, when oligodendrocyte progenitors (OPCs) are not yet found in this location. Brg1 does not interact with the Olig2 promoter in the E14 ganglionic eminence, where NPCs differentiate into Olig2-positive OPCs. Consistent with these findings, Brg1-null NPCs demonstrate precocious expression of Olig2 in the cortex. However, these cells fail to differentiate into OPCs. We further find that Brg1 is necessary for neuroepithelial-to-radial glial cell transition, but not neuronal differentiation despite a reduction in expression of the pro-neural transcription factor Pax6. Collectively, these and earlier findings support a model whereby Brg1 promotes neurogenic radial glial progenitor cell specification but is dispensable for neuronal differentiation. Concurrently, Brg1 represses Olig2 expression and the specification of OPCs, but is required for OPC differentiation and oligodendrocyte maturation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , ADN Helicasas/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/fisiología , Oligodendroglía/citología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Línea Celular Tumoral , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Neurogénesis , Factor de Transcripción 2 de los Oligodendrocitos , Regiones Promotoras Genéticas , Células Madre/citología
5.
Ann Neurol ; 73(2): 266-80, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23463525

RESUMEN

OBJECTIVE: Oligodendrocyte progenitor cells (OPCs) recruited to demyelinating lesions often fail to mature into oligodendrocytes (OLs) that remyelinate spared axons. The glycosaminoglycan hyaluronan (HA) accumulates in demyelinating lesions and has been implicated in the failure of OPC maturation and remyelination. We tested the hypothesis that OPCs in demyelinating lesions express a specific hyaluronidase, and that digestion products of this enzyme inhibit OPC maturation. METHODS: Mouse OPCs grown in vitro were analyzed for hyaluronidase expression and activity. Gain of function studies were used to define the hyaluronidases that blocked OPC maturation. Mouse and human demyelinating lesions were assessed for hyaluronidase expression. Digestion products from different hyaluronidases and a hyaluronidase inhibitor were tested for their effects on OPC maturation and functional remyelination in vivo. RESULTS: OPCs demonstrated hyaluronidase activity in vitro and expressed multiple hyaluronidases, including HYAL1, HYAL2, and PH20. HA digestion by PH20 but not other hyaluronidases inhibited OPC maturation into OLs. In contrast, inhibiting HA synthesis did not influence OPC maturation. PH20 expression was elevated in OPCs and reactive astrocytes in both rodent and human demyelinating lesions. HA digestion products generated by the PH20 hyaluronidase but not another hyaluronidase inhibited remyelination following lysolecithin-induced demyelination. Inhibition of hyaluronidase activity lead to increased OPC maturation and promoted increased conduction velocities through lesions. INTERPRETATION: We determined that PH20 is elevated in demyelinating lesions and that increased PH20 expression is sufficient to inhibit OPC maturation and remyelination. Pharmacological inhibition of PH20 may therefore be an effective way to promote remyelination in multiple sclerosis and related conditions.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Hialuronoglucosaminidasa/metabolismo , Regeneración Nerviosa/fisiología , Células-Madre Neurales/enzimología , Oligodendroglía/citología , Potenciales de Acción/fisiología , Animales , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inducido químicamente , Inhibidores Enzimáticos/farmacología , Femenino , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/antagonistas & inhibidores , Hialuronoglucosaminidasa/genética , Lisofosfatidilcolinas/toxicidad , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Oligodendroglía/fisiología
6.
Int J Cancer ; 132(12): 2767-77, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23197309

RESUMEN

Malignant rhabdoid tumors (MRTs) are rare, aggressive cancers occuring in young children primarily through inactivation of the SNF5(INI1, SMARCB1) tumor suppressor gene. We and others have demonstrated that mice heterozygous for a Snf5 null allele develop MRTs with partial penetrance. We have also shown that Snf5(+/-) mice that lack expression of the pRb family, due to TgT121 transgene expression, develop MRTs with increased penetrance and decreased latency. Here, we report that altering the genetic background has substantial effects upon MRT development in Snf5(+/--) and TgT121 ;Snf5(+/-) mice, with a mixed F1 background resulting in increased latency and the appearance of brain tumors. We also report the establishment of the first mouse MRT cell lines that recapitulate many features of their human counterparts. Our studies provide further insight into the genetic influences on MRT development as well as provide valuable new cell culture and genetically engineered mouse models for the study of CNS-MRT etiology.


Asunto(s)
Línea Celular Tumoral , Tumor Rabdoide/genética , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Neoplasias del Plexo Coroideo/genética , Neoplasias del Plexo Coroideo/patología , Proteínas Cromosómicas no Histona/genética , Modelos Animales de Enfermedad , Genotipo , Humanos , Cariotipo , Ratones , Ratones Transgénicos , Fenotipo , Tumor Rabdoide/patología , Proteína SMARCB1
7.
J Biol Chem ; 287(40): 33237-51, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22865853

RESUMEN

The extravasation of lymphocytes across central nervous system (CNS) vascular endothelium is a key step in inflammatory demyelinating diseases including multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). The glycosaminoglycan hyaluronan (HA) and its receptor, CD44, have been implicated in this process but their precise roles are unclear. We find that CD44(-/-) mice have a delayed onset of EAE compared with wild type animals. Using an in vitro lymphocyte rolling assay, we find that fewer slow rolling (<1 µm/s) wild type (WT) activated lymphocytes interact with CD44(-/-) brain vascular endothelial cells (ECs) than with WT ECs. We also find that CD44(-/-) ECs fail to anchor HA to their surfaces, and that slow rolling lymphocyte interactions with WT ECs are inhibited when the ECs are treated with a pegylated form of the PH20 hyaluronidase (PEG-PH20). Subcutaneous injection of PEG-PH20 delays the onset of EAE symptoms by ~1 day and transiently ameliorates symptoms for 2 days following disease onset. These improved symptoms correspond histologically to degradation of HA in the lumen of CNS blood vessels, decreased demyelination, and impaired CD4(+) T-cell extravasation. Collectively these data suggest that HA tethered to CD44 on CNS ECs is critical for the extravasation of activated T cells into the CNS providing new insight into the mechanisms promoting inflammatory demyelinating disease.


Asunto(s)
Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Células Endoteliales/citología , Receptores de Hialuranos/biosíntesis , Ácido Hialurónico/química , Linfocitos/citología , Animales , Encéfalo/metabolismo , Enfermedades Desmielinizantes/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Exones , Femenino , Receptores de Hialuranos/genética , Inflamación , Rodamiento de Leucocito , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
8.
Neurobiol Aging ; 33(4): 830.e13-24, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21872361

RESUMEN

The glycosaminoglycan hyaluronan (HA) accumulates in central nervous system lesions where it limits astrogliosis but also inhibits oligodendrocyte progenitor cell (OPC) maturation. The role of hyaluronan in normative brain aging has not been previously investigated. Here, we tested the hypothesis that HA accumulates in the aging nonhuman primate brain. We found that HA levels significantly increase with age in the gray matter of rhesus macaques. HA accumulation was linked to age-related increases in the transcription of HA synthase-1 (HAS1) expressed by reactive astrocytes but not changes in the expression of other HAS genes or hyaluronidases. HA accumulation was accompanied by increased expression of CD44, a transmembrane HA receptor. Areas of gray matter with elevated HA in older animals demonstrated increased numbers of olig2(+) OPCs, consistent with the notion that HA may influence OPC expansion or maturation. Collectively, these data indicate that HAS1 and CD44 are transcriptionally upregulated in astrocytes during normative aging and are linked to HA accumulation in gray matter.


Asunto(s)
Envejecimiento , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Regulación de la Expresión Génica/fisiología , Ácido Hialurónico/metabolismo , Factores de Edad , Animales , Femenino , Glucuronosiltransferasa/metabolismo , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Hialuronano Sintasas , Ácido Hialurónico/genética , Macaca fascicularis , Macaca mulatta , Masculino , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , ARN Mensajero/metabolismo
9.
J Neurosci Res ; 89(6): 840-51, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21394759

RESUMEN

Fxyd1 encodes a trans-membrane protein that modulates Na(+) ,K(+) -ATPase activity and is a substrate for multiple protein kinases. Fxyd1 expression is repressed by methyl CpG-binding protein 2 (Mecp2) in the frontal cortex (FC) but not in the cerebellum (CB) of the mouse brain. Consistently with these observations, FXYD1 mRNA abundance is increased in the FC of Rett syndrome (RTT) patients with MECP2 mutations. Because Fxyd1 is implicated in the regulation of neuronal excitability, understanding how Fxyd1 expression is controlled is important. Here we report that basal expression of Fxyd1a and Fxyd1b, the two main alternatively spliced forms of Fxyd1 mRNA, is lower in the FC than in the CB. This difference is accompanied by increased Mecp2 recruitment to the promoter region of these two Fxyd1 mRNA forms. DNA methylation of both promoters is more frequent in the FC than in the CB, and in both cases the most frequently methylated CpG dinucleotides are adjacent to [A/T](4) sequences required for high-affinity Mecp2 binding. Consistently with these features of epigenetic silencing, histone 3 acetylated at lysines 9 and 14 (H3K9/14ac) and histone 3 methylated at lysine 4 (H3K4me3), both activating histone marks, were associated with the Fxyd1 promoter to a lesser degree in the FC than in the CB. These results indicate that differential Fxyd1 expression in these two brain regions is, at least in part, regulated by an epigenetic mechanism involving increased DNA methylation of the two alternative Fxyd1 promoters, enhanced Mecp2 recruitment, and reduced association of activating histones.


Asunto(s)
Encéfalo/metabolismo , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Fosfoproteínas/metabolismo , Animales , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Metilación de ADN , Femenino , Histonas/metabolismo , Proteínas de la Membrana/genética , Proteína 2 de Unión a Metil-CpG/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/genética , ARN Mensajero/metabolismo
10.
Hum Mol Genet ; 16(6): 640-50, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17309881

RESUMEN

Rett syndrome (RTT) is an X-linked neurodevelopmental disorder linked to heterozygous de novo mutations in the MECP2 gene. MECP2 encodes methyl-CpG-binding protein 2 (MeCP2), which represses gene transcription by binding to 5-methylcytosine residues in symmetrically positioned CpG dinucleotides. Direct MeCP2 targets underlying RTT pathogenesis remain largely unknown. Here, we report that FXYD1, which encodes a transmembrane modulator of Na(+), K(+) -ATPase activity, is elevated in frontal cortex (FC) neurons of RTT patients and Mecp2-null mice. Increasing neuronal FXDY1 expression is sufficient to reduce dendritic arborization and spine formation, hallmarks of RTT neuropathology. Mecp2-null mouse cortical neurons have diminished Na(+),K(+)-ATPase activity, suggesting that aberrant FXYD1 expression contributes to abnormal neuronal activity in RTT. MeCP2 represses Fxyd1 transcription through direct interactions with sequences in the Fxyd1 promoter that are methylated in FC neurons. FXYD1 is therefore a MeCP2 target gene whose de-repression may directly contribute to RTT neuronal pathogenesis.


Asunto(s)
Encéfalo/metabolismo , Proteínas de la Membrana/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Fosfoproteínas/genética , Síndrome de Rett/genética , Animales , Estudios de Casos y Controles , Metilación de ADN , Femenino , Expresión Génica , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Regiones Promotoras Genéticas , Síndrome de Rett/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
11.
Dev Biol ; 289(2): 372-83, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16330018

RESUMEN

Epigenetic alterations in cell-type-specific gene expression control the transition of neural stem cells (NSCs) from predominantly neurogenic to predominantly gliogenic phases of differentiation, but how this switch occurs is unclear. Here, we show that brahma-related gene 1 (Brg1), an ATP-dependent chromatin remodeling factor, is required for the repression of neuronal commitment and the maintenance of NSCs in a state that permits them to respond to gliogenic signals. Loss of Brg1 in NSCs in conditional brg1 mutant mice results in precocious neuronal differentiation, such that cells in the ventricular zone differentiate into post-mitotic neurons before the onset of gliogenesis. As a result, there is a dramatic failure of astrocyte and oligodendrocyte differentiation in these animals. The ablation of brg1 in gliogenic progenitors in vitro also prevents growth-factor-induced astrocyte differentiation. Furthermore, proteins implicated in the maintenance of stem cells, including Sox1, Pax6 and Musashi-1, are dramatically reduced in the ventricular zones of brg1 mutant mice. We conclude that Brg1 is required to repress neuronal differentiation in NSCs as a means of permitting glial cell differentiation in response to gliogenic signals, suggesting that Brg1 regulates the switch from neurogenesis to gliogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Neuroglía/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/fisiología , Organogénesis/fisiología , Células Madre/metabolismo , Factores de Transcripción/fisiología , Animales , Astrocitos/metabolismo , Diferenciación Celular/genética , Linaje de la Célula , Células Cultivadas , Corteza Cerebral/metabolismo , ADN Helicasas , Femenino , Masculino , Ratones , Ratones Mutantes , Neuroglía/citología , Neuroglía/fisiología , Neuronas/citología , Proteínas Nucleares/genética , Fenotipo , Transducción de Señal , Células Madre/citología , Factores de Transcripción/genética
12.
Nat Med ; 11(9): 966-72, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16086023

RESUMEN

Demyelination is the hallmark of numerous neurodegenerative conditions, including multiple sclerosis. Oligodendrocyte progenitors (OPCs), which normally mature into myelin-forming oligodendrocytes, are typically present around demyelinated lesions but do not remyelinate affected axons. Here, we find that the glycosaminoglycan hyaluronan accumulates in demyelinated lesions from individuals with multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. A high molecular weight (HMW) form of hyaluronan synthesized by astrocytes accumulates in chronic demyelinated lesions. This form of hyaluronan inhibits remyelination after lysolecithin-induced white matter demyelination. OPCs accrue and do not mature into myelin-forming cells in demyelinating lesions where HMW hyaluronan is present. Furthermore, the addition of HMW hyaluronan to OPC cultures reversibly inhibits progenitor-cell maturation, whereas degrading hyaluronan in astrocyte-OPC cocultures promotes oligodendrocyte maturation. HMW hyaluronan may therefore contribute substantially to remyelination failure by preventing the maturation of OPCs that are recruited to demyelinating lesions.


Asunto(s)
Astrocitos/fisiología , Enfermedades Desmielinizantes/fisiopatología , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Oligodendroglía/fisiología , Animales , Encefalomielitis Autoinmune Experimental/fisiopatología , Humanos , Ratones , Esclerosis Múltiple/fisiopatología , Células Madre/fisiología
13.
Cancer Res ; 65(9): 3542-7, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867346

RESUMEN

Brahma (Brm) and brahma-related gene-1 (Brg1) are mammalian homologues of SWI/SNF chromatin-remodeling factor subunits that can regulate both transcriptional activation and repression. Both Brg1 and Brm are mutated or deleted in numerous cancer cell lines, leading to the altered expression of genes that influence cell proliferation and metastasis. Here, we find that the promoters of two such genes, CD44 and E-cadherin, are hypermethylated in cells that have lost Brg1 or Brm. In two carcinoma cell lines that lack functional Brg1 and Brm, CD44 and E-cadherin expression are induced by the demethylating agent 5-aza-2'-deoxycytidine. Transfection with either Brg1 or Brm also induces CD44 and E-cadherin transcription and protein expression in these cells, as well as loss of methylation at sequences in the promoters of both genes. Chromatin immunoprecipitation assays show that Brg1 and Brm associate with these regions of the CD44 and E-cadherin promoters, suggesting that SWI/SNF protein complexes may directly influence the loss of DNA methylation. In vivo, Brm-deficient mice also show methylation and silencing of the CD44 promoter. Collectively, these data implicate loss of SWI/SNF-mediated transcriptional activation as a novel mechanism to increase DNA methylation in cancer cells and provide insight into the mechanisms underlying aberrant gene induction and repression during tumor progression.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Metilación de ADN , Factores de Transcripción/fisiología , Activación Transcripcional/fisiología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas , Células HeLa , Humanos , Receptores de Hialuranos/genética , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
14.
J Biol Chem ; 280(12): 11790-7, 2005 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-15659388

RESUMEN

Biallelic mutations in the neurofibromatosis 2 (NF2) gene are linked to schwannoma and meningioma tumorigenesis. Cells with NF2 mutations exhibit elevated levels of phosphorylated extracellular signal-regulated kinase (ERK) and aberrant cell-cell and cell-matrix contacts. The NF2 gene product, merlin, associates with adherens junction protein complexes, suggesting that part of its function as a tumor suppressor involves regulating cell junctions. Here, we find that a novel PDZ protein, called erbin, binds directly to the merlin-binding partner, EBP0, and regulates adherens junction dissociation through a MAP kinase-dependent mechanism. Reducing erbin expression using a targeted siRNA in primary cultures of Schwann cells results in altered cell-cell interactions, disruption of E-cadherin adherens junctions, increased cell proliferation, and elevated levels of phosphorylated ERK, all phenotypes observed in cells that lack merlin. Reduction of erbin expression also results in the dissociation of merlin from adherens junction proteins and an increase in the levels of phosphorylated merlin. These phenotypes can be rescued if cells with reduced levels of erbin are treated with a pharmacological inhibitor of ERK kinase. Collectively, these data indicate that erbin regulates MAP kinase activation in Schwann cells and suggest that erbin links merlin to both adherens junction protein complexes and the MAP kinase signaling pathway.


Asunto(s)
Uniones Adherentes/fisiología , Cadherinas/fisiología , Proteínas Portadoras/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neurofibromina 2/fisiología , Células de Schwann/ultraestructura , Uniones Adherentes/química , Animales , Butadienos/farmacología , Comunicación Celular , Proliferación Celular , Células Cultivadas , Activación Enzimática , Péptidos y Proteínas de Señalización Intracelular , Sistema de Señalización de MAP Quinasas , Ratones , Nitrilos/farmacología , Fosforilación , Ratas , Células de Schwann/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...