Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
2.
Neuroscience ; 239: 173-95, 2013 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-23022538

RESUMEN

One hypothesis to account for the onset and severity of neurological disorders is the loss of trophic support. Indeed, changes in the levels and activities of brain-derived neurotrophic factor (BDNF) occur in numerous neurodegenerative and neuropsychiatric diseases. A deficit promotes vulnerability whereas a gain of function facilitates recovery by enhancing survival, synapse formation and synaptic plasticity. Implementation of 'BDNF therapies', however, faces numerous methodological and pharmacokinetic issues. Identifying BDNF mimetics that activate the BDNF receptor or downstream targets of BDNF signaling represent an alternative approach. One mechanism that shows great promise is to study the interplay of BDNF and glucocorticoid hormones, a major class of natural steroid secreted during stress reactions and in synchrony with circadian rhythms. While small amounts of glucocorticoids support normal brain function, excess stimulation by these steroid hormones precipitates stress-related affective disorders. To date, however, because of the paucity of knowledge of underlying cellular mechanisms, deleterious effects of glucocorticoids are not prevented following extreme stress. In the present review, we will discuss the complementary roles shared by BDNF and glucocorticoids in synaptic plasticity, and delineate possible signaling mechanisms mediating these effects.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/metabolismo , Glucocorticoides/metabolismo , Plasticidad Neuronal/fisiología , Transducción de Señal/fisiología , Animales , Humanos , Neuronas/metabolismo
3.
Br J Pharmacol ; 170(8): 1449-58, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24528237

RESUMEN

The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties from the IUPHAR database. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. This compilation of the major pharmacological targets is divided into seven areas of focus: G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors & Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and GRAC and provides a permanent, citable, point-in-time record that will survive database updates.


Asunto(s)
Bases de Datos Farmacéuticas , Terapia Molecular Dirigida , Farmacología , Humanos , Ligandos , Preparaciones Farmacéuticas/química
4.
Biochem Soc Trans ; 34(Pt 4): 607-11, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16856873

RESUMEN

Regulation of cell survival decisions and neuronal plasticity by neurotrophins are mediated by two classes of receptors, Trks (tropomyosin receptor kinases) and p75, the first discovered member of the tumour necrosis factor receptor superfamily. The p75 receptor participates with the TrkA receptor in the formation of high-affinity nerve growth factor-binding sites to promote survival under limiting concentrations of neurotrophins. Activation of Trk receptors leads to increased phosphorylation of Shc (Src homology and collagen homology), phospholipase C-gamma and novel adaptor molecules, such as the ARMS (ankyrin-rich membrane spanning)/Kidins220 protein. Small ligands that interact with G-protein-coupled receptors can also activate Trk receptor kinase activity. Transactivation of Trk receptors and their downstream signalling pathways raise the possibility of using small molecules to elicit neuroprotective effects for the treatment of neurodegenerative diseases. Like amyloid precursor protein and Notch, p75 is a substrate for gamma-secretase cleavage. The p75 receptor undergoes an alpha-secretase-mediated release of the extracellular domain followed by a gamma-secretase-mediated intramembrane cleavage. Cleavage of p75 may represent a general mechanism for transmitting signals as an independent receptor and as a co-receptor for other signalling systems.


Asunto(s)
Receptor de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular , Humanos , Ligandos , FN-kappa B/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor de Factor de Crecimiento Nervioso/genética , Receptor trkA/genética , Receptor trkA/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
5.
J Neurobiol ; 49(3): 224-34, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11745660

RESUMEN

Myelin in the mammalian central nervous system (CNS) is produced by oligodendrocytes, most of which arise from oligodendrocyte precursor cells (OPCs) during late embryonic and early postnatal development. Both external and internal cues have been implicated in regulating OPC exit from the cell cycle and differentiation into oligodendrocytes. In this study, we demonstrate that differentiation of cultured OPCs into mature oligodendrocytes is associated with lower levels of activity of telomerase, the ribonucleoprotein that synthesizes telomeric DNA at the ends of chromosomes. Differentiation is also associated with lower levels of mRNA encoding the catalytic subunit of telomerase (TERT), whereas no difference is seen in the expression of its telomeric template RNA component (TR). These data suggest a possible role for telomerase during normal growth and differentiation of oligodendrocytes that may be relevant to the mechanism of myelination in the CNS.


Asunto(s)
Oligodendroglía/fisiología , Telomerasa/metabolismo , Animales , Bromodesoxiuridina , Diferenciación Celular , Células Cultivadas , Isoenzimas/aislamiento & purificación , Isoenzimas/metabolismo , Masculino , Microscopía Fluorescente , Vaina de Mielina/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telomerasa/aislamiento & purificación , Moldes Genéticos
6.
J Neurobiol ; 49(1): 62-78, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11536198

RESUMEN

During development of the central nervous system, oligodendrocyte progenitor cells differentiate into mature myelinating cells. The molecular signals that promote this process, however, are not well defined. One molecule that has been implicated in oligodendrocyte differentiation is the Src family kinase Fyn. In order to probe the function of Fyn in this system, a yeast two hybrid screen was performed. Using Fyn as bait, p190 RhoGAP was isolated in the screen of an oligodendrocyte cDNA library. Coimmunoprecipitation and in vitro binding assays verified that p190 RhoGAP bound to the Fyn SH2 domain. Phosphorylation of p190 required active Fyn tyrosine kinase and was increased threefold upon differentiation of primary oligodendrocytes. Moreover, complex formation between p190 and p120 RasGAP occurred in differentiated oligodendrocytes. p190 RhoGAP activity is known to regulate the RhoGDP:RhoGTP ratio. Indeed, expression of dominant negative Rho in primary oligodendrocytes caused a hyperextension of processes. Conversely, constitutively activated Rho caused reduced process formation. These findings define a pathway in which Fyn activity regulates the phosphorylation of p190, leading to an increase in RhoGAP activity with a subsequent increase in RhoGDP, which in turn, regulates the morphological changes that accompany oligodendrocyte differentiation.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Nucleares/metabolismo , Oligodendroglía/citología , Proteínas Proto-Oncogénicas/metabolismo , Tirosina/metabolismo , Proteínas de Fase Aguda/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas Nucleares/fisiología , Oligodendroglía/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-fyn , Ratas , Ratas Sprague-Dawley , Proteínas Represoras , Especificidad por Sustrato , Proteína Activadora de GTPasa p120/fisiología
7.
J Neurosci ; 21(16): 5854-63, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11487608

RESUMEN

In addition to promoting cell survival, neurotrophins also can elicit apoptosis in restricted cell types. Recent results indicate that nerve growth factor (NGF) can induce Schwann cell death via engagement of the p75 neurotrophin receptor. Here we describe a novel interaction between the p75 receptor and receptor-interacting protein 2, RIP2 (RICK/CARDIAK), that accounts for the ability of neurotrophins to choose between a survival-versus-death pathway. RIP2, an adaptor protein with a serine threonine kinase and a caspase recruitment domain (CARD), is highly expressed in dissociated Schwann cells and displays an endogenous association with p75. RIP2 binds to the death domain of p75 via its CARD domain in an NGF-dependent manner. The introduction of RIP2 into Schwann cells deficient in RIP2 conferred NGF-dependent nuclear transcription factor-kappaB (NF-kappaB) activity and decreased the cell death induced by NGF. Conversely, the expression of a dominant-negative version of RIP2 protein resulted in a loss of NGF-induced NF-kappaB induction and increased NGF-mediated cell death. These results indicate that adaptor proteins like RIP2 can provide a bifunctional switch for cell survival or cell death decisions mediated by the p75 neurotrophin receptor.


Asunto(s)
Caspasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Factor de Transcripción Activador 2 , Animales , Apoptosis/fisiología , Western Blotting , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Genes Dominantes , Glutatión Transferasa/genética , Proteínas Fluorescentes Verdes , Humanos , Riñón/citología , Riñón/metabolismo , Ligandos , Proteínas Luminiscentes/genética , FN-kappa B/metabolismo , Factor de Crecimiento Nervioso/farmacología , Unión Proteica/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína/fisiología , Proteínas/genética , Proteínas/metabolismo , Ratas , Receptor de Factor de Crecimiento Nervioso , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Receptores de Factor de Crecimiento Nervioso/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células de Schwann/citología , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Factor 6 Asociado a Receptor de TNF , Factores de Transcripción/metabolismo , Transfección
8.
J Biol Chem ; 276(35): 32687-95, 2001 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-11435417

RESUMEN

Ligand-induced receptor oligomerization is an established mechanism for receptor-tyrosine kinase activation. However, numerous receptor-tyrosine kinases are expressed in multicomponent complexes with other receptors that may signal independently or alter the binding characteristics of the receptor-tyrosine kinase. Nerve growth factor (NGF) interacts with two structurally unrelated receptors, the Trk A receptor-tyrosine kinase and p75, a tumor necrosis factor receptor family member. Each receptor binds independently to NGF with predominantly low affinity (K(d) = 10(-9) m), but they produce high affinity binding sites (K(d) = 10(-11) m) upon receptor co-expression. Here we provide evidence that the number of high affinity sites is regulated by the ratio of the two receptors and by specific domains of Trk A and p75. Co-expression of Trk A containing mutant transmembrane or cytoplasmic domains with p75 yielded reduced numbers of high affinity binding sites. Similarly, co-expression of mutant p75 containing altered transmembrane and cytoplasmic domains with Trk A also resulted in predominantly low affinity binding sites. Surprisingly, extracellular domain mutations of p75 that abolished NGF binding still generated high affinity binding with Trk A. These results indicate that the transmembrane and cytoplasmic domains of Trk A and p75 are responsible for high affinity site formation and suggest that p75 alters the conformation of Trk A to generate high affinity NGF binding.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Sitios de Unión , Línea Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Cinética , Ligandos , Modelos Moleculares , Conformación Proteica , Receptor de Factor de Crecimiento Nervioso , Receptor trkA/química , Receptor trkA/genética , Receptores de Factor de Crecimiento Nervioso/química , Receptores de Factor de Crecimiento Nervioso/genética , Proteínas Recombinantes de Fusión/metabolismo , Spodoptera , Transfección
9.
J Neurosci ; 21(13): 4572-81, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11425885

RESUMEN

Dorsal root ganglion (DRG) neurons first express kainate receptor subunits, predominantly GluR5, during embryonic development. In the DRG and throughout the nervous system, substantial editing of GluR5 mRNA occurs with developmental maturation (Bernard et al., 1999). The accompanying change in Ca(2+) permeability of functional kainate receptors that is the predicted outcome of this developmental regulation of mRNA editing has not been investigated. Here we report that kainate receptors on DRG neurons from late embryonic and newborn rats are predominantly Ca(2+) permeable but then become fully Ca(2+) impermeable later in the first postnatal week. Using multiple markers for nociceptor subpopulations, we show that this switch in Ca(2+) permeability is not caused by the appearance of a new subpopulation of nociceptors with different receptor properties. Instead, the change in Ca(2+) permeability matches the time course of post-transcriptional RNA editing of GluR5 at the Q/R site within the pore of the channel, indicating that the change is probably caused by developmentally regulated RNA editing. We also report that, on the basis of the strong correlation of receptor expression with expression of the surface markers LA4, isolectin B4, and LD2, kainate receptors are present on C-fiber-type neurons projecting to lamina II of spinal cord dorsal horn. These results raise the possibility that kainate receptors in their Ca(2+)-permeable form serve a developmental role in synapse formation between this population of C-fibers and their targets in the spinal cord dorsal horn. Thereafter, the receptors may serve a new function that does not require Ca(2+) permeability.


Asunto(s)
Calcio/metabolismo , Nociceptores/metabolismo , Receptores de Ácido Kaínico/biosíntesis , Animales , Antígenos de Diferenciación/biosíntesis , Diferenciación Celular , Permeabilidad de la Membrana Celular/fisiología , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ácido Kaínico/farmacología , Fibras Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nociceptores/citología , Células del Asta Posterior/citología , Edición de ARN , Ratas , Receptores de Ácido Kaínico/genética
10.
Curr Opin Neurobiol ; 11(3): 281-6, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11399425
11.
Nature ; 411(6840): 957-62, 2001 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-11418861

RESUMEN

Tissue injury generates endogenous factors that heighten our sense of pain by increasing the response of sensory nerve endings to noxious stimuli. Bradykinin and nerve growth factor (NGF) are two such pro-algesic agents that activate G-protein-coupled (BK2) and tyrosine kinase (TrkA) receptors, respectively, to stimulate phospholipase C (PLC) signalling pathways in primary afferent neurons. How these actions produce sensitization to physical or chemical stimuli has not been elucidated at the molecular level. Here, we show that bradykinin- or NGF-mediated potentiation of thermal sensitivity in vivo requires expression of VR1, a heat-activated ion channel on sensory neurons. Diminution of plasma membrane phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2) levels through antibody sequestration or PLC-mediated hydrolysis mimics the potentiating effects of bradykinin or NGF at the cellular level. Moreover, recruitment of PLC-gamma to TrkA is essential for NGF-mediated potentiation of channel activity, and biochemical studies suggest that VR1 associates with this complex. These studies delineate a biochemical mechanism through which bradykinin and NGF produce hypersensitivity and might explain how the activation of PLC signalling systems regulates other members of the TRP channel family.


Asunto(s)
Bradiquinina/fisiología , Factor de Crecimiento Nervioso/fisiología , Fosfatidilinositol 4,5-Difosfato/fisiología , Receptores de Droga/fisiología , Animales , Línea Celular , Electrofisiología , Activación Enzimática , Femenino , Calor , Masculino , Ratones , Nociceptores/metabolismo , Oocitos/fisiología , Dolor , Proteína Quinasa C/metabolismo , Receptor trkA/fisiología , Receptores de Droga/genética , Transducción de Señal , Fosfolipasas de Tipo C/fisiología , Xenopus laevis
12.
Oncogene ; 20(10): 1229-34, 2001 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11313867

RESUMEN

The TrkA NGF receptor extracellular region contains three leucine repeats flanked by cysteine clusters and two immunoglobulin-like domains that are required for specific ligand binding. Deletion of the immunoglobulin-like domains abolishes NGF binding and causes ligand independent activation of the receptor. Here we report a specific mutation that increases the binding affinity of the TrkA receptor for NGF. A change of proline 203 to alanine (P203A) in the linker region between the leucine repeats and the first Ig-like domain increased NGF binding by decreasing the ligand rate of dissociation. This mutated receptor was appropriately expressed on the cell surface and promoted ligand-independent neurite outgrowth in PC12nnr5 cells. The mutant receptor was capable of spontaneous dimerization and was constitutively phosphorylated in the absence of ligand. Moreover, expression of TrkA-P203A receptor in fibroblasts induced DNA synthesis and transformation and generated tumours in nude mice. These data suggest that domains outside of the immunoglobulin-like structure contribute to ligand binding and constitutive activation of Trk receptors.


Asunto(s)
Mutación , Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Ensayo de Unidades Formadoras de Colonias , Ligandos , Mutagénesis Sitio-Dirigida , Neuritas/fisiología , Células PC12/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína/genética , Ratas , Relación Estructura-Actividad
13.
Mol Biol Cell ; 12(3): 615-27, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11251075

RESUMEN

NGF initiates the majority of its neurotrophic effects by promoting the activation of the tyrosine kinase receptor TrkA. Here we describe a novel interaction between TrkA and GIPC, a PDZ domain protein. GIPC binds to the juxtamembrane region of TrkA through its PDZ domain. The PDZ domain of GIPC also interacts with GAIP, an RGS (regulators of G protein signaling) protein. GIPC and GAIP are components of a G protein-coupled signaling complex thought to be involved in vesicular trafficking. In transfected HEK 293T cells GIPC, GAIP, and TrkA form a coprecipitable protein complex. Both TrkA and GAIP bind to the PDZ domain of GIPC, but their binding sites within the PDZ domain are different. The association of endogenous GIPC with the TrkA receptor was confirmed by coimmunoprecipitation in PC12 (615) cells stably expressing TrkA. By immunofluorescence GIPC colocalizes with phosphorylated TrkA receptors in retrograde transport vesicles located in the neurites and cell bodies of differentiated PC12 (615) cells. These results suggest that GIPC, like other PDZ domain proteins, serves to cluster transmembrane receptors with signaling molecules. When GIPC is overexpressed in PC12 (615) cells, NGF-induced phosphorylation of mitogen-activated protein (MAP) kinase (Erk1/2) decreases; however, there is no effect on phosphorylation of Akt, phospholipase C-gamma1, or Shc. The association of TrkA receptors with GIPC and GAIP plus the inhibition of MAP kinase by GIPC suggests that GIPC may provide a link between TrkA and G protein signaling pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al GTP/metabolismo , Neuropéptidos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor trkA/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Humanos , Sustancias Macromoleculares , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Factor de Crecimiento Nervioso/farmacología , Neuronas/citología , Neuronas/metabolismo , Neuropéptidos/química , Neuropéptidos/genética , Células PC12 , Fosforilación , Estructura Terciaria de Proteína , Ratas , Receptor trkA/química , Receptor trkA/genética , Transducción de Señal , Técnicas del Sistema de Dos Híbridos
14.
EMBO Rep ; 2(1): 27-34, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11252720

RESUMEN

Differentiation of most cell types requires both establishment of G1 arrest and the induction of a program related to achieving quiescence. We have chosen to study the differentiation of oligodendrocyte cells to determine the role of p27 and p21 in this process. Here we report that both p27 and p21 are required for the appropriate differentiation of these cells. p27 is required for proper withdrawal from the cell cycle, p21 is not. Instead, p21 is required for the establishment of the differentiation program following growth arrest. Similar observations were made in vivo. We show that p21-/- cells withdraw from the cell cycle similar to wild-type cells; however, early in animal life, the brain is hypomyelinated, inferring that the loss of p21 delayed myelination in the cerebellum. We found that we could complement or bypass the differentiation failure in p21-/- cells with either PD98059, an inhibitor of Mek1, or by transducing them with a tat-p16ink4a protein. We concluded that the two cdk inhibitors serve non-redundant roles in this program of differentiation, with p27 being responsible for arrest and p21 having a function in differentiation independent of its ability to control exit from the cell cycle.


Asunto(s)
Proteínas de Ciclo Celular , Ciclo Celular , Ciclinas/fisiología , Proteínas Supresoras de Tumor , Animales , Animales Recién Nacidos , Antimetabolitos/farmacología , Encéfalo/metabolismo , Bromodesoxiuridina/farmacología , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Genotipo , Inmunohistoquímica , Cinética , MAP Quinasa Quinasa 1 , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Oligodendroglía/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Factores de Tiempo , Transducción Genética
15.
Proc Natl Acad Sci U S A ; 98(6): 3555-60, 2001 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-11248116

RESUMEN

Neurotrophins regulate neuronal cell survival and synaptic plasticity through activation of Trk receptor tyrosine kinases. Binding of neurotrophins to Trk receptors results in receptor autophosphorylation and downstream phosphorylation cascades. Here, we describe an approach to use small molecule agonists to transactivate Trk neurotrophin receptors. Activation of TrkA receptors in PC12 cells and TrkB in hippocampal neurons was observed after treatment with adenosine, a neuromodulator that acts through G protein-coupled receptors. These effects were reproduced by using the adenosine agonist CGS 21680 and were counteracted with the antagonist ZM 241385, indicating that this transactivation event by adenosine involves adenosine 2A receptors. The increase in Trk activity could be inhibited by the use of the Src family-specific inhibitor, PP1, or K252a, an inhibitor of Trk receptors. In contrast to other G protein-coupled receptor transactivation events, adenosine used Trk receptor signaling with a longer time course. Moreover, adenosine activated phosphatidylinositol 3-kinase/Akt through a Trk-dependent mechanism that resulted in increased cell survival after nerve growth factor or brain-derived neurotrophic factor withdrawal. Therefore, adenosine acting through the A(2A) receptors exerts a trophic effect through the engagement of Trk receptors. These results provide an explanation for neuroprotective actions of adenosine through a unique signaling mechanism and raise the possibility that small molecules may be used to elicit neurotrophic effects for the treatment of neurodegenerative diseases.


Asunto(s)
Factores de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Receptores Purinérgicos P1/metabolismo , Transducción de Señal , Adenosina/metabolismo , Adenosina/farmacología , Animales , Línea Celular , Células Cultivadas , Activación Enzimática , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Agonistas del Receptor Purinérgico P1 , Ratas , Receptor de Adenosina A2A , Receptor trkA/antagonistas & inhibidores
16.
J Neurosci ; 21(3): RC125, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11157096

RESUMEN

Nerve growth factor (NGF) initiates its trophic effects by long-range signaling through binding, internalization, and transport of a ligand-receptor complex from the axon terminal to the cell body. However, the mechanism by which retrograde transport of NGF takes place has not been elucidated. Here we describe an interaction between the Trk receptor tyrosine kinase and a 14 kDa light chain of cytoplasmic dynein. After transfection in human embryonic kidney 293 cells, this 14 kDa dynein light chain was found to bind to TrkA, TrkB, and TrkC receptors. Mapping experiments indicated that the 14 kDa dynein light chain binds to the distal region of the TrkA juxtamembrane domain. Coimmunoprecipitation experiments in vivo indicate that Trk receptors are in a complex with the 14 kDa light chain and 74 kDa intermediate chain of dynein. Confirming the physiological relevance of this association, a marked accumulation of Trk with the 14 kDa and the 74 kDa dynein components was observed after ligation of the sciatic nerve. The association of Trk receptors with components of cytoplasmic dynein suggests that transport of neurotrophins during vesicular trafficking may occur through a direct interaction of the Trk receptor with the dynein motor machinery.


Asunto(s)
Citoplasma/metabolismo , Proteínas de Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos , Proteínas Motoras Moleculares/metabolismo , Proteínas Nucleares , Receptor trkA/metabolismo , Animales , Células Cultivadas , Humanos , Riñón/citología , Riñón/metabolismo , Ligadura , Proteínas de Microtúbulos/genética , Mutagénesis Sitio-Dirigida , Neuronas/citología , Neuronas/metabolismo , Pruebas de Precipitina , Unión Proteica/fisiología , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Nervio Ciático/fisiología , Eliminación de Secuencia , Transfección , Técnicas del Sistema de Dos Híbridos , Región del Complejo T del Genoma
17.
J Neurosci ; 21(1): 176-85, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11150334

RESUMEN

Appropriate development of nervous system connectivity involves a variety of processes, including neuronal life-and-death decisions, differentiation, axon guidance and migration, and synaptogenesis. Although these activities likely require specialized signaling events, few substrates unique to these neurotrophic functions have been identified. Here we describe the cloning of ankyrin repeat-rich membrane spanning (ARMS), which encodes a novel downstream target of neurotrophin and ephrin receptor tyrosine kinases, Trk and Eph, respectively. The amino acid sequence of ARMS is highly conserved from nematode to human, suggesting an evolutionarily conserved role for this protein. The ARMS protein consists of 1715 amino acids containing four putative transmembrane domains, multiple ankyrin repeats, a sterile alpha motif domain, and a potential PDZ-binding motif. In the rat, ARMS is specifically expressed in the developing nervous system and in highly plastic areas of the adult brain, regions enriched in Trks and Eph receptors. ARMS can physically associate with TrkA and p75 neurotrophin receptors. Moreover, endogenous ARMS protein is tyrosine phosphorylated after neurotrophin treatment of pheochromocytoma 12 cells and primary hippocampal neurons or ephrin B treatment of NG108-15 cells, demonstrating that ARMS is a downstream target for both neurotrophin and ephrin receptors.


Asunto(s)
Secuencia Conservada/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Fosfoproteínas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Secuencias de Aminoácidos/genética , Animales , Repetición de Anquirina/genética , Línea Celular , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Clonación Molecular , Humanos , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/farmacología , Especificidad de Órganos , Células PC12 , Fosforilación , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/genética , Receptor EphA1 , Receptor de Factor de Crecimiento Nervioso , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Homología de Secuencia de Aminoácido
18.
Mol Cell Biol ; 21(3): 893-901, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11154276

RESUMEN

The Akt family of serine/threonine-directed kinases promotes cellular survival in part by phosphorylating and inhibiting death-inducing proteins. Here we describe a novel functional interaction between Akt and apoptosis signal-regulating kinase 1 (ASK1), a mitogen-activated protein kinase kinase kinase. Akt decreased ASK1 kinase activity stimulated by both oxidative stress and overexpression in 293 cells by phosphorylating a consensus Akt site at serine 83 of ASK1. Activation of the phosphoinositide 3-kinase (PI3-K)/Akt pathway also inhibited the serum deprivation-induced activity of endogenous ASK1 in L929 cells. An association between Akt and ASK1 was detected in cells by coimmunoprecipitation. Phosphorylation by Akt inhibited ASK1-mediated c-Jun N-terminal kinase and activating transcription factor 2 activities in intact cells. Finally, activation of the PI3-K/Akt pathway reduced apoptosis induced by ASK1 in a manner dependent on phosphorylation of serine 83 of ASK1. These results provide the first direct link between Akt and the family of stress-activated kinases.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , Apoptosis , Línea Celular , Supervivencia Celular , Células HeLa , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , MAP Quinasa Quinasa Quinasa 5 , Quinasas Quinasa Quinasa PAM/química , Quinasas Quinasa Quinasa PAM/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Serina/química , Transducción de Señal , Especificidad por Sustrato
19.
Mol Cell Biol ; 20(16): 5908-16, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10913174

RESUMEN

The extracellular region of the nerve growth factor (NGF) receptor, TrkA, contains two immunoglobulin (Ig)-like domains that are required for specific ligand binding. We have investigated the possible role of these two Ig-like domains in receptor dimerization and activation by using different mutants of the TrkA extracellular region. Deletions of each Ig-like domain, of both, and of the entire extracellular region were made. To probe the structural constraints on ligand-independent receptor dimerization, chimeric receptors were generated by swapping the Ig-like domains of the TrkA receptor for the third or fourth Ig-like domain of c-Kit. We also introduced single-amino-acid changes in conserved residues within the Ig-like domains of TrkA. Most of these TrkA variants did not bind NGF, and their expression in PC12nnr5 cells, which lack endogenous TrkA, promoted ligand-independent neurite outgrowth. Some TrkA mutant receptors induced malignant transformation of Rat-1 cells, as assessed by measuring proliferation in the absence of serum, anchorage-independent growth, and tumorigenesis in nude mice. These mutants exhibited constitutive phosphorylation and spontaneous dimerization consistent with their biological activities. Our data suggest that spontaneous dimerization of TrkA occurs when the structure of the Ig-like domains is altered, implying that the intact domains inhibit receptor dimerization in the absence of NGF.


Asunto(s)
Receptor trkA/química , Sustitución de Aminoácidos , Animales , Sitios de Unión , Ligandos , Ratones , Factor de Crecimiento Nervioso/metabolismo , Unión Proteica , Receptor trkA/genética , Receptor trkA/metabolismo , Relación Estructura-Actividad
20.
J Neurosci ; 20(12): 4627-34, 2000 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10844032

RESUMEN

Schwann cell proliferation is regulated by multiple growth factors and axonal signals. However, the molecules that control growth arrest of Schwann cells are not well defined. Here we describe regulation of the cyclin-dependent kinase-2 (CDK2) protein, an enzyme that is necessary for the transition from G1 to S phase. Levels of CDK2 protein were elevated in proliferating Schwann cells cultured in serum and forskolin. However, when cells were grown with either serum-free media or at high densities, CDK2 levels declined to low levels. The decrease in CDK2 levels was associated with growth arrest of Schwann cells. The modulation of CDK2 appears to be regulated at the transcriptional level, because CDK2 mRNA levels and its promoter activity both decline during cell cycle arrest. Furthermore, analysis of the CDK2 promoter suggests that Sp1 DNA binding sites are essential for maximal activation in Schwann cells. Together, these data suggest that CDK2 may represent a significant target of developmental signals that regulate Schwann cell proliferation and that this regulation is mediated, in part, through regulation of Sp1 transcriptional activity.


Asunto(s)
Quinasas CDC2-CDC28 , Ciclo Celular/fisiología , Quinasas Ciclina-Dependientes/metabolismo , Neuronas/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Células de Schwann/citología , Células de Schwann/fisiología , Animales , Animales Recién Nacidos , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Células Cultivadas , Colforsina/farmacología , Medio de Cultivo Libre de Suero , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/genética , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Regulación Enzimológica de la Expresión Génica , Neuritas/fisiología , Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA