Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Anal Chem ; 94(35): 11967-11972, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35998076

RESUMEN

One of the main challenges in cancer management relates to the discovery of reliable biomarkers, which could guide decision-making and predict treatment outcome. In particular, the rise and democratization of high-throughput molecular profiling technologies bolstered the discovery of "biomarker signatures" that could maximize the prediction performance. Such an approach was largely employed from diverse OMICs data (i.e., genomics, transcriptomics, proteomics, metabolomics) but not from epitranscriptomics, which encompasses more than 100 biochemical modifications driving the post-transcriptional fate of RNA: stability, splicing, storage, and translation. We and others have studied chemical marks in isolation and associated them with cancer evolution, adaptation, as well as the response to conventional therapy. In this study, we have designed a unique pipeline combining multiplex analysis of the epitranscriptomic landscape by high-performance liquid chromatography coupled to tandem mass spectrometry with statistical multivariate analysis and machine learning approaches in order to identify biomarker signatures that could guide precision medicine and improve disease diagnosis. We applied this approach to analyze a cohort of adult diffuse glioma patients and demonstrate the existence of an "epitranscriptomics-based signature" that permits glioma grades to be discriminated and predicted with unmet accuracy. This study demonstrates that epitranscriptomics (co)evolves along cancer progression and opens new prospects in the field of omics molecular profiling and personalized medicine.


Asunto(s)
Glioma , ARN , Biomarcadores , Glioma/diagnóstico , Glioma/genética , Humanos , Metabolómica/métodos , Análisis Multivariante , Proteómica/métodos
2.
Sci Rep ; 10(1): 5504, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32218467

RESUMEN

IDH1-mutated gliomas are slow-growing brain tumours which progress into high-grade gliomas. The early molecular events causing this progression are ill-defined. Previous studies revealed that 20% of these tumours already have transformation foci. These foci offer opportunities to better understand malignant progression. We used immunohistochemistry and high throughput RNA profiling to characterize foci cells. These have higher pSTAT3 staining revealing activation of JAK/STAT signaling. They downregulate RNAs involved in Wnt signaling (DAAM2, SFRP2), EGFR signaling (MLC1), cytoskeleton and cell-cell communication (EZR, GJA1). In addition, foci cells show reduced levels of RNA coding for Ethanolamine-Phosphate Phospho-Lyase (ETNPPL/AGXT2L1), a lipid metabolism enzyme. ETNPPL is involved in the catabolism of phosphoethanolamine implicated in membrane synthesis. We detected ETNPPL protein in glioma cells as well as in astrocytes in the human brain. Its nuclear localization suggests additional roles for this enzyme. ETNPPL expression is inversely correlated to glioma grade and we found no ETNPPL protein in glioblastomas. Overexpression of ETNPPL reduces the growth of glioma stem cells indicating that this enzyme opposes gliomagenesis. Collectively, these results suggest that a combined alteration in membrane lipid metabolism and STAT3 pathway promotes IDH1-mutated glioma malignant progression.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Liasas de Carbono-Oxígeno/genética , Glioma/genética , Glioma/metabolismo , Isocitrato Deshidrogenasa/genética , Factor de Transcripción STAT3/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Regulación hacia Abajo , Perfilación de la Expresión Génica , Glioma/patología , Humanos , Inmunohistoquímica , Metabolismo de los Lípidos , Mutación , Fosforilación , Transducción de Señal
3.
Physiol Res ; 67(Suppl 1): S257-S264, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29947545

RESUMEN

The endothelin axis (endothelins and their receptors) is strongly involved in physiological and pathological processes. ET-1 plays a crucial role in particular in tumor diseases. Endothelin-1 receptors (ET(A) and ET(B)) are deregulated and overexpressed in several tumors such as melanoma and glioma. We studied the binding of 24 monoclonal antibodies directed against human ET(B) receptors (hET(B)) to different melanoma cell lines. Few of these mAbs bound to all the melanoma cell lines. One of them, rendomab B49, bound to ET(B) receptors expressed at the surface of human glioma stem cells. More recently, we produced new antibodies directed against human ET(A) receptor (hET(A)). Several antibodies have been isolated and have been screened on different tumoral cells lines. As for the mAbs directed against the hET(B) receptor only some of new antibodies directed against ET(A) receptor are capable to bind the human tumoral cell lines. Rendomab A63 directed against hET(A) is one of them. We report the specificity and binding properties of these mAbs and consider their potential use in diagnosis by an in vivo imaging approach.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antagonistas de los Receptores de la Endotelina A/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Antagonistas de los Receptores de la Endotelina A/administración & dosificación , Endotelina-1/genética , Endotelina-1/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Unión Proteica/fisiología , Receptor de Endotelina A/genética , Receptor de Endotelina B/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Glia ; 66(2): 239-255, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29027701

RESUMEN

Diffuse low grade gliomas (DLGG, grade II gliomas) are slowly-growing brain tumors that often progress into high grade gliomas. Most tumors have a missense mutation for IDH1 combined with 1p19q codeletion in oligodendrogliomas or ATRX/TP53 mutations in astrocytomas. The phenotype of tumoral cells, their environment and the pathways activated in these tumors are still ill-defined and are mainly based on genomics and transcriptomics analysis. Here we used freshly-resected tumors to accurately characterize the tumoral cell population and their environment. In oligodendrogliomas, cells express the transcription factors MYT1, Nkx2.2, Olig1, Olig2, Sox8, four receptors (EGFR, PDGFRα, LIFR, PTPRZ1) but not the co-receptor NG2 known to be expressed by oligodendrocyte progenitor cells. A variable fraction of cells also express the more mature oligodendrocytic markers NOGO-A and MAG. DLGG cells are also stained for the young-neuron marker doublecortin (Dcx) which is also observed in oligodendrocytic cells in nontumoral human brain. In astrocytomas, MYT1, PDGFRα, PTPRZ1 were less expressed whereas Sox9 was prominent over Sox8. The phenotype of DLGG cells is overall maintained in culture. Phospho-array screening showed the absence of EGFR and PDGFRα phosphorylation in DLGG but revealed the strong activation of p44/42 MAPK/ERK which was present in a fraction of tumoral cells but also in nontumoral cells. These results provide evidence for the existence of close relationships between the cellular phenotype and the mutations found in DLGG. The slow proliferation of these tumors may be associated with the absence of activation of PDGFRα/EGFR receptors.


Asunto(s)
Neoplasias Encefálicas/genética , Receptores ErbB/genética , Glioma/genética , Isocitrato Deshidrogenasa/genética , Mutación/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proteína Doblecortina , Receptores ErbB/metabolismo , Femenino , Glioma/metabolismo , Glioma/patología , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Humanos , Isocitrato Deshidrogenasa/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Clasificación del Tumor/métodos , Proteínas Nucleares , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Transcripción , Células Tumorales Cultivadas , Adulto Joven
5.
Br J Cancer ; 107(9): 1488-97, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23033007

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) cells are resistant to anticancer drugs. Cancer stem cells (CSCs) are a key mediator of chemoresistance. We have reported that disulfiram (DS), an aldehyde dehydrogenase (ALDH) inhibitor, targets breast CSC-like cells. In this study, the effect of DS and combination of DS and gemcitabine (dFdC) on GBM cells and GBM stem-like cells was investigated. METHODS: 1-(4,5-Dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT), combination index (CI)-isobologram, western blot, luciferase reporter gene assay, electrophoretic mobility-shift assay and ALDH analysis were used in this study. RESULTS: Disulfiram is cytotoxic in GBM cell lines in a copper (Cu)-dependent manner. Disulfiram/copper enhances the cytotoxicity of dFdC. Combination index-isobologram analysis indicates a synergistic effect between DS/Cu and dFdC. Disulfiram/copper induces reactive oxygen species (ROS), activates JNK and p38 pathways and inhibits nuclear factor-kappa B activity in GBM cell lines. Disulfiram/copper may trigger intrinsic apoptotic pathway via modulation of the Bcl2 family. Disulfiram/copper abolishes stem-like cell population in GBM cell lines. CONCLUSION: Our findings indicate that the cytotoxicity of DS/Cu and the enhancing effect of DS/Cu on the cytotoxicity of dFdC in GBM stem-like cells may be caused by induction of ROS and inhibition of both ALDH and the NFkB pathway. Both DS and dFdC can traverse the blood-brain barrier. Further study may lead them into GBM chemotherapy.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Cobre/farmacología , Disulfiram/farmacología , Glioblastoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Aldehído Deshidrogenasa/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sinergismo Farmacológico , Glioblastoma/enzimología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Gemcitabina
6.
Cell Death Dis ; 2: e218, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-21993394

RESUMEN

Serotonergic innervation of the spinal cord in mammals has multiple roles in the control of motor, sensory and visceral functions. In rats, functional consequences of spinal cord injury at thoracic level can be improved by a substitutive transplantation of serotonin (5-HT) neurons or regeneration under the trophic influence of grafted stem cells. Translation to either pharmacological and/or cellular therapies in humans requires the mapping of the spinal cord 5-HT innervation and its receptors to determine their involvement in specific functions. Here, we have performed a preliminary mapping of serotonergic processes and serotonin-lA (5-HT(1A)) receptors in thoracic and lumbar segments of the human spinal cord. As in rodents and non-human primates, 5-HT profiles in human spinal cord are present in the ventral horn, surrounding motoneurons, and also contact their presumptive dendrites at lumbar level. 5-HT(1A) receptors are present in the same area, but are more densely expressed at lumbar level. 5-HT profiles are also present in the intermediolateral region, where 5-HT(1A) receptors are absent. Finally, we observed numerous serotonergic profiles in the superficial part (equivalent of Rexed lamina II) of the dorsal horn, which also displayed high levels of 5-HT(1A) receptors. These findings pave the way for local specific therapies involving cellular and/or pharmacological tools targeting the serotonergic system.


Asunto(s)
Receptor de Serotonina 5-HT1A/metabolismo , Neuronas Serotoninérgicas/citología , Médula Espinal/anatomía & histología , Adolescente , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ratas , Neuronas Serotoninérgicas/metabolismo , Médula Espinal/metabolismo , Adulto Joven
7.
J Neurosci Res ; 86(9): 1916-26, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18335522

RESUMEN

Adult human and rodent brains contain neural stem and progenitor cells, and the presence of neural stem cells in the adult rodent spinal cord has also been described. Here, using electron microscopy, expression of neural precursor cell markers, and cell culture, we investigated whether neural precursor cells are also present in adult human spinal cord. In well-preserved nonpathological post-mortem human adult spinal cord, nestin, Sox2, GFAP, CD15, Nkx6.1, and PSA-NCAM were found to be expressed heterogeneously by cells located around the central canal. Ultrastructural analysis revealed the existence of immature cells close to the ependymal cells, which display characteristics of type B and C cells found in the adult rodent brain subventricular region, which are considered to be stem and progenitor cells, respectively. Completely dissociated spinal cord cells reproducibly formed Sox2(+) nestin(+) neurospheres containing proliferative precursor cells. On differentiation, these generate glial cells and gamma-aminobutyric acid (GABA)-ergic neurons. These results provide the first evidence for the existence in the adult human spinal cord of neural precursors with the potential to differentiate into neurons and glia. They represent a major interest for endogenous regeneration of spinal cord after trauma and in degenerative diseases.


Asunto(s)
Neuroglía/citología , Neuronas/citología , Médula Espinal/citología , Médula Espinal/fisiología , Células Madre/citología , Adulto , Animales , Biomarcadores/metabolismo , Muerte Encefálica , Técnicas de Cultivo de Célula , Diferenciación Celular , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Células Madre/fisiología , Donantes de Tejidos
8.
Mol Cell Neurosci ; 24(1): 198-213, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14550780

RESUMEN

NTERA2 cells are a human neural cell line generating neurons after exposure to retinoic acid and, as such, are widely used as a model of neurogenesis. We report that these cells form spheres when grown in serum-free medium supplemented with basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF). These spheres were found to express markers of radial glial cells such as, Pax6, glutamate transporter (GLAST), tenascin C, brain lipid-binding protein (BLBP), and the 3CB2 antigen. On plating on an adhesive substrate, NTERA2 spheres generate a large percentage of immature neurons (30-50%) together with a minority of cells of the oligodendrocyte lineage. Thus NTERA2 cells share properties with neural stem cells. However, at variance with the latter, we found that they produce their own bFGF implicated in an autocrine or paracrine proliferative loop and that they do not generate astrocytes after differentiation. These results provide an interesting model to study radial glial cells and their role in human neurogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Neuroglía/metabolismo , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/fisiología , Biomarcadores , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Microscopía Electrónica , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/ultraestructura , Neuronas/ultraestructura , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/ultraestructura , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/ultraestructura , Células Madre/efectos de los fármacos , Células Madre/ultraestructura
9.
J Neurosci Res ; 65(3): 195-207, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11494354

RESUMEN

During brain development, the neuroepithelium generates neurons and glial cells. Proliferation and differentiation of neuroepithelial cells are controlled by a complex combination of secreted factors and more intrinsic or local mechanisms, such as lateral inhibition and asymmetric division. To obtain further insights into the signals governing neuroepithelial cell fate, we used the immortomouse to derive conditionally immortalised cell lines from mouse E10 neuroepithelium. We isolated a nestin-positive basic fibroblast growth factor (bFGF)-responsive cell line (SVE10-23) which mostly differentiate into astrocytes when cocultured with primary cortical cells. We found that, by simply lowering the cell density, SVE10-23 cells embarked on oligodendrocytic differentiation as indicated by the strong expression of galactocerebroside C and 2'3'-cyclic nucleotide 3'-phosphodiesterase. Apoptosis accompanied the differentiation, and all cells died within 1 week. We present here evidence that direct interactions between cells are the main mechanism regulating this oligodendrocytic differentiation. We demonstrate that SVE10-23 cells contact or proximity inhibit their differentiation, prevent apoptosis, and promote their proliferation. Similarly, others nestin-positive precursor cell lines and nonimmortalised bFGF-grown E10 cells were found to spontaneously differentiate at low density, thus generalising the idea that neural precursor fate is regulated by direct cell-cell interactions. The SVE10-23 cell line provides a valuable tool with which to study further the molecular components implicated in this mode of regulation.


Asunto(s)
Apoptosis/fisiología , Comunicación Celular , Oligodendroglía/citología , Hidrolasas Diéster Fosfóricas , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa , 2',3'-Nucleótido Cíclico Fosfodiesterasas/biosíntesis , Animales , Astrocitos/citología , Biomarcadores , Diferenciación Celular , Línea Celular Transformada , Linaje de la Célula , Corteza Cerebral/citología , Técnicas de Cocultivo , Inhibición de Contacto , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Galactosilceramidas/biosíntesis , Proteína Ácida Fibrilar de la Glía/análisis , Proteínas de Filamentos Intermediarios/análisis , Ratones , Ratones Transgénicos , Proteína Básica de Mielina/análisis , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Oligodendroglía/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
10.
Neuroreport ; 12(10): 2237-41, 2001 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-11447341

RESUMEN

MHP36 is a nestin bFGF-dependent cell line isolated from embryonic hippocampus using a thermolabile form of SV40 T antigen. When grafted in ischemic hippocampus MHP36 cells differentiate and alleviate the cognitive deficit associated with the lesion. We report here in vitro features of MHP36 cells. First, we found that T Ag expression was not necessary for MHP36 growth as cells cultured at the nonpermissive temperature carry on proliferating at a normal rate, Second, we observed that part of MHP36 cells spontaneously differentiate into astrocytes when bFGF is removed at39 degrees C. This differentiation was increased 4-fold by leukemia inhibitory factor. Third, we found that the majority of cells spontaneously expressed oligodendrocytic markers (CNPase, A2B5, GalC) when cultured at low density.


Asunto(s)
Astrocitos/fisiología , Neuroglía/citología , Oligodendroglía/fisiología , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Astrocitos/efectos de los fármacos , Proteínas Portadoras/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Transgénicos , Neuroglía/efectos de los fármacos , Neuroglía/fisiología , Oligodendroglía/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/fisiología
11.
J Biol Chem ; 272(39): 24371-9, 1997 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-9305895

RESUMEN

Two novel K+ channel alpha subunits, named Kv9.1 and Kv9.2, have been cloned. The Kv9.2 gene is situated in the 8q22 region of the chromosome. mRNAs for these two subunits are highly and selectively expressed in the nervous system. High levels of expressions are found in the olfactory bulb, cerebral cortex, hippocampal formation, habenula, basolateral amygdaloid nuclei, and cerebellum. Interestingly Kv9.1 and Kv9.2 colocalized with Kv2.1 and/or Kv2.2 alpha subunits in several regions of the brain. Neither Kv9.1 nor Kv9.2 have K+ channel activity by themselves, but both modulate the activity of Kv2.1 and Kv2.2 channels by changing kinetics and levels of expression and by shifting the half-inactivation potential to more polarized values. This report also analyzes the changes in electrophysiological properties of Kv2 subunits induced by Kv5.1 and Kv6.1, two other modulatory subunits. Each modulatory subunit has its own specific properties of regulation of the functional Kv2 subunits, and they can lead to extensive inhibitions, to large changes in kinetics, and/or to large shifts in the voltage dependencies of the inactivation process. The increasing number of modulatory subunits for Kv2.1 and Kv2.2 provides an amazingly new capacity of functional diversity.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Células COS , Clonación Molecular , ADN Complementario , Técnica del Anticuerpo Fluorescente , Activación del Canal Iónico , Ratones , Datos de Secuencia Molecular , Fosforilación , Filogenia , Canales de Potasio/química , Canales de Potasio/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido
12.
J Biol Chem ; 272(13): 8774-80, 1997 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-9079713

RESUMEN

The Kv8.1 subunit is unable to generate K+ channel activity in Xenopus oocytes or in COSm6 cells. The Kv8.1 subunit expressed at high levels acts as a specific suppressor of the activity of Kv2 and Kv3 channels in Xenopus oocytes (Hugnot, J. P., Salinas, M., Lesage, F., Guillemare, E., Weille, J., Heurteaux, C., Mattéi, M. G., and Lazdunski, M. (1996) EMBO J. 15, 3322-3331). At lower levels, Kv8.1 associates with Kv2.1 and Kv2.2 to form hybrid Kv8.1/Kv2 channels, which have new biophysical properties and more particularly modified properties of the inactivation process as compared with homopolymers of Kv2.1 or Kv2.2 channels. The same effects have been seen by coexpressing the Kv8.1 subunit and the Kv2.2 subunit in COSm6 cells. In these cells, Kv8.1 expressed alone remains in intracellular compartments, but it can reach the plasma membrane when it associates with Kv2.2, and it then also forms new types of Kv8.1/Kv2. 2 channels. Present results indicate that Kv8.1 when expressed at low concentrations acts as a modifier of Kv2.1 and Kv2.2 activity, while when expressed at high concentrations in oocytes it completely abolishes Kv2.1, Kv2.2, or Kv3.4 K+ channel activity. The S6 segment of Kv8.1 is atypical and contains the structural elements that modify inactivation of Kv2 channels.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Canales de Potasio de Tipo Rectificador Tardío , Técnica del Anticuerpo Fluorescente Indirecta , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Alineación de Secuencia , Canales de Potasio Shab , Fracciones Subcelulares/metabolismo , Xenopus
13.
Genomics ; 39(1): 113-6, 1997 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9027495

RESUMEN

K+ channels are essential for a variety of cellular functions in both excitable and nonexcitable cells, and K+ channel gene alteration has been recently described in cardiac and neurological disorders. To explore further the relations between hereditary human diseases and K+ channels, we isolated from a human cosmid library the gene encoding the inwardly rectifying K+ channel alpha-subunit Kir 2.2 (KCNJ12). PCR analysis performed on this clone indicates that the entire open reading frame is contained in one unique exon. A polymorphic (CA)16 sequence was localized 2.2 kb upstream of the ATG start codon. Fluorescence in situ hybridization on human metaphases assigns the gene to band 17p11.1. The implication of a deletion of the Kir 2.2 gene in the Smith-Magenis syndrome, which is also localized at 17p11, is unlikely since a Kir 2.2-linked microsatellite sequence could be amplified from the DNA of a Smith-Magenis syndrome affected patient bearing a 17p interstitial deletion.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 17 , Repeticiones de Dinucleótido , Polimorfismo Genético , Canales de Potasio de Rectificación Interna , Canales de Potasio/genética , Anomalías Múltiples/genética , Animales , Línea Celular , Clonación Molecular , Cricetinae , Exones , Femenino , Eliminación de Gen , Humanos , Hibridación Fluorescente in Situ , Intrones , Masculino , Ratones , Datos de Secuencia Molecular , Síndrome
14.
EMBO J ; 15(13): 3322-31, 1996 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-8670833

RESUMEN

Outward rectifier K+ channels have a characteristic structure with six transmembrane segments and one pore region. A new member of this family of transmembrane proteins has been cloned and called Kv8.1. Kv8.1 is essentially present in the brain where it is located mainly in layers II, IV and VI of the cerebral cortex, in hippocampus, in CA1-CA4 pyramidal cell layer as well in granule cells of the dentate gyrus, in the granule cell layer and in the Purkinje cell layer of the cerebellum. The Kv8.1 gene is in the 8q22.3-8q24.1 region of the human genome. Although Kv8.1 has the hallmarks of functional subunits of outward rectifier K+ channels, injection of its cRNA in Xenopus oocytes does not produce K+ currents. However Kv8.1 abolishes the functional expression of members of the Kv2 and Kv3 subfamilies, suggesting that the functional role of Kv8.1 might be to inhibit the function of a particular class of outward rectifier K+ channel types. Immunoprecipitation studies have demonstrated that inhibition occurs by formation of heteropolymeric channels, and results obtained with Kv8.1 chimeras have indicated that association of Kv8.1 with other types of subunits is via its N-terminal domain.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Bloqueadores de los Canales de Potasio , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Corteza Cerebral/metabolismo , Mapeo Cromosómico , Cromosomas Humanos Par 8 , Clonación Molecular , ADN Complementario , Hipocampo/metabolismo , Humanos , Activación del Canal Iónico , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Oocitos , Canales de Potasio/genética , Pruebas de Precipitina , ARN Complementario , Homología de Secuencia de Aminoácido , Canales de Potasio Shaw , Xenopus laevis
15.
Biochem Biophys Res Commun ; 212(2): 657-63, 1995 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-7626080

RESUMEN

The family of G-protein-activated inward-rectifiers K+ channels presently comprise at least 3 cloned members called GIRK1, GIRK2 and GIRK3. A close structural parent of GIRK channels has recently been described as being an ATP-sensitive K+ channel. This paper shows that Xenopus expression of this new channel that we call GIRK4 does not produce an ATP-inhibitable activity with a pharmacological activation by pinacidil as previously described but instead a G-protein activated inward-rectifier. While oocyte expression of single subunits is infrequent and relatively modest in intensity, expression of GIRK1,2, GIRK1,4 and GIRK2,4 mixtures leads to routine and robust expression of K+ channels indicating that heterologous subunit assembly is necessary for activity. Activity of GIRK1,2, GIRK1,4 and GIRK2,4 channels required the presence of ATP acting as an activator at the cytoplasmic face and is further activated by the beta gamma subunits.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Corazón/fisiología , Neuronas/fisiología , Canales de Potasio de Rectificación Interna , Canales de Potasio/química , Canales de Potasio/fisiología , Adenosina Trifosfato/farmacología , Animales , Conductividad Eléctrica , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Expresión Génica , Técnicas de Transferencia de Gen , Sustancias Macromoleculares , Oocitos/metabolismo , Canales de Potasio/genética , ARN Complementario , Relación Estructura-Actividad , Xenopus
16.
FEBS Lett ; 353(1): 37-42, 1994 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-7926018

RESUMEN

MbIRK3, mbGIRK2 and mbGIRK3 K+ channels cDNAs have been cloned from adult mouse brain. These cDNAs encode polypeptides of 445, 414 and 376 amino acids, respectively, which display the hallmarks of inward rectifier K+ channels, i.e. two hydrophobic membrane-spanning domains M1 and M2 and a pore-forming domain H5. MbIRK3 shows around 65% amino acid identity with IRK1 and rbIRK2 and only 50% with ROMK1 and GIRK1. On the other hand, mbGIRK2 and mbGIRK3 are more similar to GIRK1 (60%) than to ROMK1 and IRK1 (50%). Northern blot analysis reveals that these three novel clones are mainly expressed in the brain. Xenopus oocytes injected with mbIRK3 and mbGIRK2 cRNAs display inward rectifier K(+)-selective currents very similar to IRK1 and GIRK1, respectively. As expected from the sequence homology, mbGIRK2 cRNA directs the expression of G-protein coupled inward rectifier K+ channels which has been observed through their functional coupling with co-expressed delta-opioid receptors. These results provide the first evidence that the GIRK family, as the IRK family, is composed of multiple genes with members specifically expressed in the nervous system.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Unión al GTP/metabolismo , Canales de Potasio de Rectificación Interna , Canales de Potasio/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Ratones , Datos de Secuencia Molecular , Canales de Potasio/metabolismo , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Distribución Tisular , Xenopus
17.
Nucleic Acids Res ; 22(18): 3685-8, 1994 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-7937077

RESUMEN

The rat HBP1 cDNA was cloned by its capacity to suppress the potassium transport-defective phenotype of mutant Saccharomyces cerevisiae cells. HBP1 cDNA encodes a 513 amino acids protein which, unexpectedly, does not share any homology with K+ transporters or K+ channels. However, a search in protein databases reveals that HBP1 contains a putative DNA-binding domain called HMG-box. Northern blot analysis shows that HBP1 is expressed in a variety of tissues and that in adipocyte and myogenic cell lines, its expression is directly related to differentiation. Taken together, the results suggest that the rat HBP1 is a new member of the HMG class of transcriptional regulators involved in cell differentiation pathways.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/genética , Potasio/metabolismo , Proteínas Represoras , Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/genética , Adipocitos/citología , Secuencia de Aminoácidos , Animales , Química Encefálica , Diferenciación Celular , Línea Celular , Clonación Molecular , ADN Complementario , Expresión Génica , Biblioteca de Genes , Proteína HMGB1 , Proteínas del Grupo de Alta Movilidad/fisiología , Transporte Iónico , Datos de Secuencia Molecular , Músculos/citología , Especificidad de Órganos , ARN Mensajero/biosíntesis , Ratas , Saccharomyces cerevisiae/genética , Análisis de Secuencia de ADN , Factores de Transcripción/fisiología
18.
J Biol Chem ; 268(32): 24283-9, 1993 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8226976

RESUMEN

The mouse Kv1-5 K+ channel cDNA has been cloned from heart. This channel was highly expressed in heart and, to a lesser extent, in other tissues, including brain and thymus. Two alternatively spliced isoforms were found. The longer form encoded a 602-amino acid protein, while in the short form (Kv1-5 delta 5'), the first 200 amino acids lying upstream the transmembrane segment S1 were deleted. RNase protection experiments showed that both Kv1-5 mRNA isoforms are present in the mouse tissues examined, the longer form being predominant. The short mRNA (Kv1-5 delta 5') arose by an unusual splicing event within the exonic sequence. An additional short cDNA clone (Kv1-5 delta 3') that codes for a carboxyl-terminal truncated protein has been isolated. The gene coding sequence contained a single exon and has been mapped on human chromosome 12 (p13) and on mouse chromosome 6 (band F). Expression in Xenopus oocytes revealed that the long (Kv1-5) and the amino-terminal deleted (Kv1-5 delta 5') isoforms elicited similar K+ currents with a drastically decreased efficacy for Kv1-5 delta 5'. The carboxyl-terminal truncated Kv1-5 delta 3' clone was not functional but inhibited the expression of the long isoform.


Asunto(s)
Miocardio/metabolismo , Canales de Potasio/genética , ARN Mensajero/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , ADN Recombinante , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Oocitos , Xenopus
19.
Neuromuscul Disord ; 3(5-6): 519-24, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8186704

RESUMEN

The pattern of expression of two distal transcripts initiated in the 62nd intron of the dystrophin gene was investigated under different circumstances; (i) during the development of different rat tissues these transcripts and Dp71, a protein encoded by one of them, increased with brain development and decreased with muscle development; (ii) in cultured glial and neuronal cells, the distal promoter was coactivated with tissue-specific upstream promoters, the muscle-type promoter in glial cells and the brain-type promoter in neuronal cells, which suggests that activity of the upstream promoter does not interfere with activity of the distal promoter; (iii) in lymphoblasts of DMD patients with various deletions of the dystrophin gene, the most distal of which included the 56th intron, the production of the distal transcript was not perturbed.


Asunto(s)
Distrofina/biosíntesis , Distrofina/genética , Expresión Génica , Intrones , Distrofias Musculares/genética , Animales , Secuencia de Bases , Niño , Cartilla de ADN , Eliminación de Gen , Humanos , Hígado/metabolismo , Linfocitos/metabolismo , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Ratas , Transcripción Genética
20.
Biochem Biophys Res Commun ; 192(1): 69-74, 1993 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-8476435

RESUMEN

The dystrophin whose defect is responsible for Duchenne and Becker muscular dystrophies is present in muscle, brain and cerebellum. We describe here the detection of dystrophin in human cultured skin fibroblasts, L809 cells and murine 3T6 cell line. Dystrophin transcripts initiated at the muscle specific first exon can also be amplified by cDNA-PCR from various fibroblastic cells. The expression of the dystrophin gene in fibroblasts could account for some abnormalities observed in patient's fibroblast cultures.


Asunto(s)
Distrofina/genética , Animales , Secuencia de Bases , Química Encefálica , Línea Celular , Células Cultivadas , ADN , Fibroblastos/metabolismo , Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Músculos/química , Reacción en Cadena de la Polimerasa , Empalme del ARN , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...