Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
EMBO J ; 41(15): e110721, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35730718

RESUMEN

ΔfosB is an alternatively spliced product of the FosB gene that is essential for dopamine-induced reward pathways and that acts as a master switch for addiction. However, the molecular mechanisms of its generation and regulation by dopamine signaling are unknown. Here, we report that dopamine D1 receptor signaling synergizes with the activin/ALK4/Smad3 pathway to potentiate the generation of ΔFosB mRNA in medium spiny neurons (MSNs) of the nucleus accumbens (NAc) via activation of the RNA-binding protein PCBP1, a regulator of mRNA splicing. Concurrent activation of PCBP1 and Smad3 by D1 and ALK4 signaling induced their interaction, nuclear translocation, and binding to sequences in exon-4 and intron-4 of FosB mRNA. Ablation of either ALK4 or PCBP1 in MSNs impaired ΔFosB mRNA induction and nuclear translocation of ΔFosB protein in response to repeated co-stimulation of D1 and ALK4 receptors. Finally, ALK4 is required in NAc MSNs of adult mice for behavioral sensitization to cocaine. These findings uncover an unexpected mechanism for ΔFosB generation and drug-induced sensitization through convergent dopamine and ALK4 signaling.


Asunto(s)
Cocaína , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Activinas Tipo I/metabolismo , Empalme Alternativo , Animales , Cocaína/metabolismo , Cocaína/farmacología , Dopamina/metabolismo , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo
2.
J Biol Chem ; 297(2): 100916, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34175311

RESUMEN

The p75 neurotrophin receptor (p75NTR) is a critical mediator of neuronal death and tissue remodeling and has been implicated in various neurodegenerative diseases and cancers. The death domain (DD) of p75NTR is an intracellular signaling hub and has been shown to interact with diverse adaptor proteins. In breast cancer cells, binding of the adaptor protein TRADD to p75NTR depends on nerve growth factor and promotes cell survival. However, the structural mechanism and functional significance of TRADD recruitment in neuronal p75NTR signaling remain poorly understood. Here we report an NMR structure of the p75NTR-DD and TRADD-DD complex and reveal the mechanism of specific recognition of the TRADD-DD by the p75NTR-DD mainly through electrostatic interactions. Furthermore, we identified spatiotemporal overlap of p75NTR and TRADD expression in developing cerebellar granule neurons (CGNs) at early postnatal stages and discover the physiological relevance of the interaction between TRADD and p75NTR in the regulation of canonical NF-κB signaling and cell survival in CGNs. Our results provide a new structural framework for understanding how the recruitment of TRADD to p75NTR through DD interactions creates a membrane-proximal platform, which can be efficiently regulated by various neurotrophic factors through extracellular domains of p75NTR, to propagate downstream signaling in developing neurons.


Asunto(s)
FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/química , Receptores de Factor de Crecimiento Nervioso/metabolismo , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Animales , Dominio de Muerte , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptor de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Proteína de Dominio de Muerte Asociada a Receptor de TNF/química
3.
Cell Tissue Res ; 382(1): 71-82, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32737575

RESUMEN

The discovery in the late 1990s of the partnership between the RET receptor tyrosine kinase and the GFRα family of GPI-anchored co-receptors as mediators of the effects of GDNF family ligands galvanized the field of neurotrophic factors, firmly establishing a new molecular framework besides the ubiquitous neurotrophins. Soon after, however, it was realized that many neurons and brain areas expressed GFRα receptors without expressing RET. These observations led to the formulation of two new concepts in GDNF family signaling, namely, the non-cell-autonomous functions of GFRα molecules, so-called trans signaling, as well as cell-autonomous functions mediated by signaling receptors distinct from RET, which became known as RET-independent signaling. To date, the best studied RET-independent signaling pathway for GDNF family ligands involves the neural cell adhesion molecule NCAM and its association with GFRα co-receptors. Among the many functions attributed to this signaling system are neuronal migration, neurite outgrowth, dendrite branching, spine formation, and synaptogenesis. This review summarizes our current understanding of this and other mechanisms of RET-independent signaling by GDNF family ligands and GFRα receptors, as well as their physiological importance.


Asunto(s)
Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Movimiento Celular , Humanos , Ligandos , Transducción de Señal
4.
J Biol Chem ; 295(7): 2034-2042, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31919095

RESUMEN

Prolonged cold exposure stimulates the formation of brownlike adipocytes expressing UCP1 (uncoupling-protein-1) in subcutaneous white adipose tissue which, together with classical brown adipose tissue, contributes to maintaining body temperature in mammals through nonshivering thermogenesis. The mechanisms that regulate the formation of these cells, alternatively called beige or brite adipocytes, are incompletely understood. Here we report that mice lacking CD137, a cell surface protein used in several studies as a marker for beige adipocytes, showed elevated levels of thermogenic markers, including UCP1, increased numbers of beige adipocyte precursors, and expanded UCP1-expressing cell clusters in inguinal white adipose tissue after chronic cold exposure. CD137 knockout mice also showed enhanced cold resistance. These results indicate that CD137 functions as a negative regulator of "browning" in white adipose tissue and call into question the use of this protein as a functional marker for beige adipocytes.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Proteína Desacopladora 1/genética , Adipocitos Beige/metabolismo , Animales , Temperatura Corporal/genética , Frío , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Noqueados , Termogénesis/genética
5.
J Neurosci ; 39(28): 5452-5465, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31085607

RESUMEN

Sleep deprivation (SD) interferes with hippocampal structural and functional plasticity, formation of long-term memory and cognitive function. The molecular mechanisms underlying these effects are incompletely understood. Here, we show that SD impaired synaptic tagging and capture and behavioral tagging, two major mechanisms of associative learning and memory. Strikingly, mutant male mice lacking the p75 neurotrophin receptor (p75NTR) were resistant to the detrimental effects of SD on hippocampal plasticity at both cellular and behavioral levels. Mechanistically, SD increased p75NTR expression and its interaction with phosphodiesterase. p75NTR deletion preserved hippocampal structural and functional plasticity by preventing SD-mediated effects on hippocampal cAMP-CREB-BDNF, cAMP-PKA-LIMK1-cofilin, and RhoA-ROCK2 pathways. Our study identifies p75NTR as an important mediator of hippocampal structural and functional changes associated with SD, and suggests that targeting p75NTR could be a promising strategy to limit the memory and cognitive deficits that accompany sleep loss.SIGNIFICANCE STATEMENT The lack of sufficient sleep is a major health concern in today's world. Sleep deprivation (SD) affects cognitive functions such as memory. We have investigated how associative memory mechanisms, synaptic tagging and capture (STC), was impaired in SD mice at cellular and behavioral level. Interestingly, mutant male mice that lacked the p75 neurotrophin receptor (p75NTR) were seen to be resistant to the SD-induced impairments in hippocampal synaptic plasticity and STC. Additionally, we elucidated the molecular pathways responsible for this rescue of plasticity in the mutant mice. Our study has thus identified p75NTR as a promising target to limit the cognitive deficits associated with SD.


Asunto(s)
Hipocampo/metabolismo , Memoria , Plasticidad Neuronal , Receptores de Factor de Crecimiento Nervioso/metabolismo , Privación de Sueño/genética , Factores Despolimerizantes de la Actina/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasas Lim/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
6.
Biol Rev Camb Philos Soc ; 94(4): 1282-1293, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30762293

RESUMEN

The death domain (DD) is a globular protein motif with a signature feature of an all-helical Greek-key motif. It is a primary mediator of a variety of biological activities, including apoptosis, cell survival and cytoskeletal changes, which are related to many neurodegenerative diseases, neurotrauma, and cancers. DDs exist in a wide range of signalling proteins including p75 neurotrophin receptor (p75NTR ), a member of the tumour necrosis factor receptor superfamily. The specific signalling mediated by p75NTR in a given cell depends on the type of ligand engaging the extracellular domain and the recruitment of cytosolic interactors to the intracellular domain, especially the DD, of the receptor. In solution, the p75NTR -DDs mainly form a symmetric non-covalent homodimer. In response to extracellular signals, conformational changes in the p75NTR extracellular domain (ECD) propagate to the p75NTR -DD through the disulfide-bonded transmembrane domain (TMD) and destabilize the p75NTR -DD homodimer, leading to protomer separation and exposure of binding sites on the DD surface. In this review, we focus on recent advances in the study of the structural mechanism of p75NTR -DD signalling through recruitment of diverse intracellular interactors for the regulation and control of diverse functional outputs.


Asunto(s)
Dominio de Muerte , Receptor de Factor de Crecimiento Nervioso/química , Receptor de Factor de Crecimiento Nervioso/metabolismo , Secuencia de Aminoácidos , Animales , Regulación de la Expresión Génica , Modelos Moleculares , Conformación Proteica , Receptor de Factor de Crecimiento Nervioso/genética
8.
Cell Chem Biol ; 25(12): 1485-1494.e5, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30293939

RESUMEN

Small molecules offer powerful ways to alter protein function. However, most proteins in the human proteome lack small-molecule probes, including the large class of non-catalytic transmembrane receptors, such as death receptors. We hypothesized that small molecules targeting the interfaces between transmembrane domains (TMDs) in receptor complexes may induce conformational changes that alter receptor function. Applying this concept in a screening assay, we identified a compound targeting the TMD of death receptor p75NTR that induced profound conformational changes and receptor activity. The compound triggered apoptotic cell death dependent on p75NTR and JNK activity in neurons and melanoma cells, and inhibited tumor growth in a melanoma mouse model. Due to their small size and crucial role in receptor activation, TMDs represent attractive targets for small-molecule manipulation of receptor function.


Asunto(s)
Antineoplásicos/farmacología , Melanoma/tratamiento farmacológico , Melanoma/patología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/química , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Melanoma/metabolismo , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
9.
Cell Rep ; 19(10): 1977-1986, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28591570

RESUMEN

The role of neurotrophic factors as endogenous survival proteins for brain neurons remains contentious. In the cerebellum, the signals controlling survival of molecular layer interneurons (MLIs) are unknown, and direct evidence for the requirement of a full complement of MLIs for normal cerebellar function and motor learning has been lacking. Here, we show that Purkinje cells (PCs), the target of MLIs, express the neurotrophic factor GDNF during MLI development and survival of MLIs depends on GDNF receptors GFRα1 and RET. Conditional mutant mice lacking either receptor lose a quarter of their MLIs, resulting in compromised synaptic inhibition of PCs, increased PC firing frequency, and abnormal acquisition of eyeblink conditioning and vestibulo-ocular reflex performance, but not overall motor activity or coordination. These results identify an endogenous survival mechanism for MLIs and reveal the unexpected vulnerability and selective requirement of MLIs in the control of cerebellar-dependent motor learning.


Asunto(s)
Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Aprendizaje/fisiología , Actividad Motora/fisiología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Células de Purkinje/metabolismo , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-ret/genética , Células de Purkinje/citología
10.
Cell Rep ; 18(2): 367-379, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28076782

RESUMEN

During embryonic development of the cerebellum, Purkinje cells (PCs) migrate away from the ventricular zone to form the PC plate. The mechanisms that regulate PC migration are incompletely understood. Here, we report that the neurotrophic receptor GFRα1 is transiently expressed in developing PCs and loss of GFRα1 delays PC migration. Neither GDNF nor RET, the canonical GFRα1 ligand and co-receptor, respectively, contribute to this process. Instead, we found that the neural cell adhesion molecule NCAM is co-expressed and directly interacts with GFRα1 in embryonic PCs. Genetic reduction of NCAM expression enhances wild-type PC migration and restores migration in Gfra1 mutants, indicating that NCAM restricts PC migration in the embryonic cerebellum. In vitro experiments indicated that GFRα1 can function both in cis and trans to counteract NCAM and promote PC migration. Collectively, our studies show that GFRα1 contributes to PC migration by limiting NCAM function.


Asunto(s)
Movimiento Celular , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Células de Purkinje/citología , Células de Purkinje/metabolismo , Animales , Cerebelo/citología , Embrión de Mamíferos/metabolismo , Femenino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-ret/metabolismo , Células Madre/citología , Células Madre/metabolismo
11.
J Neurosci ; 36(20): 5587-95, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27194337

RESUMEN

UNLABELLED: The p75 neurotrophin receptor (p75(NTR)) mediates neuronal death in response to neural insults by activating a caspase apoptotic pathway. The oligomeric state and activation mechanism that enable p75(NTR) to mediate these effects have recently been called into question. Here, we have investigated mutant mice lacking the p75(NTR) death domain (DD) or a highly conserved transmembrane (TM) cysteine residue (Cys(259)) implicated in receptor dimerization and activation. Neuronal death induced by proneurotrophins or epileptic seizures was assessed and compared with responses in p75(NTR) knock-out mice and wild-type animals. Proneurotrophins induced apoptosis of cultured hippocampal and cortical neurons from wild-type mice, but mutant neurons lacking p75(NTR), only the p75(NTR) DD, or just Cys(259) were all equally resistant to proneurotrophin-induced neuronal death. Homo-FRET anisotropy experiments demonstrated that both NGF and proNGF induce conformational changes in p75(NTR) that are dependent on the TM cysteine. In vivo, neuronal death induced by pilocarpine-mediated seizures was significantly reduced in the hippocampus and somatosensory, piriform, and entorhinal cortices of all three strains of p75(NTR) mutant mice. Interestingly, the levels of protection observed in mice lacking the DD or only Cys(259) were identical to those of p75(NTR) knock-out mice even though the Cys(259) mutant differed from the wild-type receptor in only one amino acid residue. We conclude that, both in vitro and in vivo, neuronal death induced by p75(NTR) requires the DD and TM Cys(259), supporting the physiological relevance of DD signaling by disulfide-linked dimers of p75(NTR) in the CNS. SIGNIFICANCE STATEMENT: A detailed understanding of the physiological significance of distinct structural determinants in the p75 neurotrophin receptor (p75(NTR)) is crucial for the identification of suitable drug targets in this receptor. We have tested the relevance of the p75(NTR) death domain (DD) and the highly conserved transmembrane residue Cys(259) for the ability of p75(NTR) to induce apoptosis in neurons of the CNS using gene-targeted mutant mice. The physiological importance of these determinants had been contested in some recent in vitro studies. Our results indicate a requirement for DD signaling by disulfide-linked dimers of p75(NTR) for neuronal death induced by proneurotrophins and epileptic seizures. These new mouse models will be useful for clarifying different aspects of p75(NTR) physiology.


Asunto(s)
Apoptosis , Corteza Cerebral/metabolismo , Mutación , Multimerización de Proteína , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Animales , Células COS , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Chlorocebus aethiops , Cisteína/genética , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Pilocarpina/toxicidad , Dominios Proteicos , Receptores de Factor de Crecimiento Nervioso/química , Receptores de Factor de Crecimiento Nervioso/genética , Convulsiones/etiología , Convulsiones/genética
12.
Elife ; 3: e03245, 2014 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-25161195

RESUMEN

Obesity is associated with blunted ß-adrenoreceptor (ß-AR)-mediated lipolysis and lipid oxidation in adipose tissue, but the mechanisms linking nutrient overload to catecholamine resistance are poorly understood. We report that targeted disruption of TGF-ß superfamily receptor ALK7 alleviates diet-induced catecholamine resistance in adipose tissue, thereby reducing obesity in mice. Global and fat-specific Alk7 knock-out enhanced adipose ß-AR expression, ß-adrenergic signaling, mitochondrial biogenesis, lipid oxidation, and lipolysis under a high fat diet, leading to elevated energy expenditure, decreased fat mass, and resistance to diet-induced obesity. Conversely, activation of ALK7 reduced ß-AR-mediated signaling and lipolysis cell-autonomously in both mouse and human adipocytes. Acute inhibition of ALK7 in adult mice by a chemical-genetic approach reduced diet-induced weight gain, fat accumulation, and adipocyte size, and enhanced adipocyte lipolysis and ß-adrenergic signaling. We propose that ALK7 signaling contributes to diet-induced catecholamine resistance in adipose tissue, and suggest that ALK7 inhibitors may have therapeutic value in human obesity.


Asunto(s)
Receptores de Activinas Tipo I/genética , Tejido Adiposo/metabolismo , Obesidad/genética , Receptores Adrenérgicos beta/genética , Receptores de Activinas Tipo I/antagonistas & inhibidores , Receptores de Activinas Tipo I/deficiencia , Adenosina Trifosfato/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/patología , Animales , Catecolaminas/metabolismo , Dieta Alta en Grasa , Grasas de la Dieta/efectos adversos , Regulación de la Expresión Génica , Humanos , Peroxidación de Lípido/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Ratones Noqueados , Recambio Mitocondrial/efectos de los fármacos , Obesidad/etiología , Obesidad/patología , Obesidad/prevención & control , Cultivo Primario de Células , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
13.
Diabetologia ; 57(1): 148-56, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24132781

RESUMEN

AIMS/HYPOTHESIS: Glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells is regulated by paracrine factors, the identity and mechanisms of action of which are incompletely understood. Activins are expressed in pancreatic islets and have been implicated in the regulation of GSIS. Activins A and B signal through a common set of intracellular components, but it is unclear whether they display similar or distinct functions in glucose homeostasis. METHODS: We examined glucose homeostatic responses in mice lacking activin B and in pancreatic islets derived from these mutants. We compared the ability of activins A and B to regulate downstream signalling, ATP production and GSIS in islets and beta cells. RESULTS: Mice lacking activin B displayed elevated serum insulin levels and GSIS. Injection of a soluble activin B antagonist phenocopied these changes in wild-type mice. Isolated pancreatic islets from mutant mice showed enhanced GSIS, which could be rescued by exogenous activin B. Activin B negatively regulated GSIS and ATP production in wild-type islets, while activin A displayed the opposite effects. The downstream mediator Smad3 responded preferentially to activin B in pancreatic islets and beta cells, while Smad2 showed a preference for activin A, indicating distinct signalling effects of the two activins. In line with this, overexpression of Smad3, but not Smad2, decreased GSIS in pancreatic islets. CONCLUSIONS/INTERPRETATION: These results reveal a tug-of-war between activin ligands in the regulation of insulin secretion by beta cells, and suggest that manipulation of activin signalling could be a useful strategy for the control of glucose homeostasis in diabetes and metabolic disease.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas Smad/metabolismo , Animales , Prueba de Tolerancia a la Glucosa , Subunidades beta de Inhibinas/genética , Subunidades beta de Inhibinas/metabolismo , Secreción de Insulina , Masculino , Ratones , Ratones Mutantes , Microscopía Electrónica , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Smad/genética
14.
Artículo en Inglés | MEDLINE | ID: mdl-23378586

RESUMEN

The identification of the ret oncogene by Masahide Takahashi and Geoffrey Cooper in 1985 was both serendipitous and paradigmatic ( Takahashi et al. 1985). By transfecting total DNA from a human lymphoma into mouse NIH3T3 cells, they obtained one clone, which in secondary transformants yielded more than 100-fold improvement in transformation efficiency. Subsequent investigations revealed that the ret oncogene was not present as such in the primary lymphoma, but was derived by DNA rearrangement during transfection from normal human sequences of the ret locus. At the time, activation by DNA rearrangement had not been previously described for a transforming gene with the NIH3T3 transfection assay. The discovery of ret opened a field of study that has had a profound impact in cancer research, developmental biology, and neuroscience, and that continues to yield surprises and important insights to this day.


Asunto(s)
Proteínas Proto-Oncogénicas c-ret/fisiología , Animales , Transformación Celular Neoplásica , Evolución Molecular , Reordenamiento Génico , Humanos , Riñón/embriología , Ligandos , Ratones , Células 3T3 NIH , Sistema Nervioso/embriología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-ret/química , Transducción de Señal
15.
Cell Rep ; 2(6): 1563-70, 2012 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-23260665

RESUMEN

Structural determinants underlying signaling specificity in the tumor necrosis factor receptor superfamily (TNFRSF) are poorly characterized, and it is unclear whether different signaling outputs can be genetically dissociated. The p75 neurotrophin receptor (p75(NTR)), also known as TNFRSF16, is a key regulator of trophic and injury responses in the nervous system. Here, we describe a genetic approach for dissecting p75(NTR) signaling and deciphering its underlying logic. Structural determinants important for regulation of cell death, NF-κB, and RhoA pathways were identified in the p75(NTR) death domain (DD). Proapoptotic and prosurvival pathways mapped onto nonoverlapping epitopes, demonstrating that different signaling outputs can be genetically separated in p75(NTR). Dissociation of c-Jun kinase (JNK) and caspase-3 activities indicated that JNK is necessary but not sufficient for p75(NTR)-mediated cell death. RIP2 recruitment and RhoGDI release were mechanistically linked, indicating that competition for DD binding underlies crosstalk between NF-κB and RhoA pathways in p75(NTR) signaling. These results provide insights into the logic of p75(NTR) signaling and pave the way for a genetic dissection of p75(NTR) function and physiology.


Asunto(s)
MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Células HEK293 , Humanos , MAP Quinasa Quinasa 4/genética , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Células 3T3 NIH , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Estructura Terciaria de Proteína , Ratas , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Inhibidor alfa de Disociación del Nucleótido Guanina rho/genética , Inhibidor alfa de Disociación del Nucleótido Guanina rho/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
16.
J Cell Sci ; 124(Pt 16): 2797-805, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21807944

RESUMEN

GDNF (glial cell line-derived neurotrophic factor) promotes the differentiation and migration of GABAergic neuronal precursors of the medial ganglionic eminence (MGE). These functions are dependent on the GPI-anchored receptor GFRα1, but independent of its two known transmembrane receptor partners RET and NCAM. Here we show that soluble GFRα1 is also able to promote differentiation and migration of GABAergic MGE neurons. These activities require endogenous production of GDNF. Although GDNF responsiveness is abolished in Gfra1(-/-) neurons, it can be restored upon addition of soluble GFRα1, a result that is only compatible with the existence of a previously unknown transmembrane signaling partner for the GDNF-GFRα1 complex in GABAergic neurons. The roles of two candidate transmembrane receptors previously implicated in GABAergic interneuron development--MET, a receptor for hepatocyte growth factor (HGF), and ErbB4, the neuregulin receptor--were examined. GDNF did not induce the activation of either receptor, nor did inhibition of MET or ErbB4 impair GDNF activity in GABAergic MGE neurons. Unexpectedly, however, inhibition of MET or HGF per se promoted neuronal differentiation and migration and enhanced the activity of GDNF on MGE neurons. These effects were dependent on endogenous GDNF and GFRα1, suggesting that MET signaling negatively regulates GDNF activity in the MGE. In agreement with this, Met mutant MGE neurons showed enhanced responses to GDNF and inhibition of MET or HGF increased Gfra1 mRNA expression in MGE cells. In vivo, expression of MET and GFRα1 overlapped in the MGE, and a loss-of-function mutation in Met increased Gfra1 expression in this region. Together, these observations demonstrate the existence of a novel transmembrane receptor partner for the GDNF-GFRα1 complex and uncover an unexpected interplay between GDNF-GFRα1 and HGF-MET signaling in the early diversification of cortical GABAergic interneuron subtypes.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Eminencia Media/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Células COS , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Chlorocebus aethiops , Receptores ErbB/antagonistas & inhibidores , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/patología , Ganglios/patología , Factor Neurotrófico Derivado de la Línea Celular Glial/inmunología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Indoles/farmacología , Eminencia Media/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Quinazolinas , Receptor ErbB-4 , Transducción de Señal/genética , Sulfonamidas/farmacología , Tirfostinos/farmacología
17.
J Biol Chem ; 285(41): 31867-75, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20682772

RESUMEN

The signaling mechanisms by which neurotrophic receptors regulate neuronal survival and axonal growth are still incompletely understood. In the receptor tyrosine kinase RET, a receptor for GDNF (glial cell line-derived neurotrophic factor), the functions of the majority of tyrosine residues that become phosphorylated are still unknown. Here we have identified the protein-tyrosine phosphatase SHP2 as a novel direct interactor of RET and the first effector known to bind to phosphorylated Tyr(687) in the juxtamembrane region of the receptor. We show that SHP2 is recruited to RET upon ligand binding in a cooperative fashion, such that both interaction with Tyr(687) and association with components of the Tyr(1062) signaling complex are required for stable recruitment of SHP2 to the receptor. SHP2 recruitment contributes to the ability of RET to activate the PI3K/AKT pathway and promote survival and neurite outgrowth in primary neurons. Furthermore, we find that activation of protein kinase A (PKA) by forskolin reduces the recruitment of SHP2 to RET and negatively affects ligand-mediated neurite outgrowth. In agreement with this, mutation of Ser(696), a known PKA phosphorylation site in RET, enhances SHP2 binding to the receptor and eliminates the effect of forskolin on ligand-induced outgrowth. Together, these findings establish SHP2 as a novel positive regulator of the neurotrophic activities of RET and reveal Tyr(687) as a critical platform for integration of RET and PKA signals. We anticipate that several other phosphotyrosines of unknown function in neuronal receptor tyrosine kinases will also support similar regulatory functions.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal/fisiología , Animales , Células COS , Chlorocebus aethiops , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Ratones , Mutación , Células 3T3 NIH , Células PC12 , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Ratas , Transducción de Señal/efectos de los fármacos
18.
J Cell Sci ; 122(Pt 18): 3351-7, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19706676

RESUMEN

Dimerization is recognized as a crucial step in the activation of many plasma membrane receptors. However, a growing number of receptors pre-exist as dimers in the absence of ligand, indicating that, although necessary, dimerization is not always sufficient for signaling. The p75 neurotrophin receptor (p75(NTR)) forms disulfide-linked dimers at the cell surface independently of ligand binding through Cys257 in its transmembrane domain. Here, we show that crosslinking of p75(NTR) dimers by cysteine-scanning mutagenesis results in constitutive, ligand-independent activity in several pathways that are normally engaged upon neurotrophin stimulation of native receptors. The activity profiles of different disulfide-crosslinked p75(NTR) mutants were similar but not identical, suggesting that different configurations of p75(NTR) dimers might be endowed with different functions. Interestingly, crosslinked p75(NTR) mutants did not mimic the effects of the myelin inhibitors Nogo or MAG, suggesting the existence of ligand-specific activation mechanisms. Together, these results support a conformational model of p75(NTR) activation by neurotrophins, and reveal a genetic approach to generate gain-of-function receptor variants with distinct functional profiles.


Asunto(s)
Reactivos de Enlaces Cruzados/metabolismo , Disulfuros/metabolismo , Multimerización de Proteína , Receptor de Factor de Crecimiento Nervioso/química , Receptor de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Células COS , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Chlorocebus aethiops , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ligandos , Datos de Secuencia Molecular , Proteínas Mutantes/efectos de los fármacos , Proteínas Mutantes/metabolismo , FN-kappa B/metabolismo , Factores de Crecimiento Nervioso/farmacología , Multimerización de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Ratas , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Transducción de Señal/efectos de los fármacos , Factor 6 Asociado a Receptor de TNF/metabolismo
19.
Neuron ; 62(1): 72-83, 2009 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-19376068

RESUMEN

Ligand-mediated dimerization has emerged as a universal mechanism of growth factor receptor activation. Neurotrophins interact with dimers of the p75 neurotrophin receptor (p75(NTR)), but the mechanism of receptor activation has remained elusive. Here, we show that p75(NTR) forms disulphide-linked dimers independently of neurotrophin binding through the highly conserved Cys(257) in its transmembrane domain. Mutation of Cys(257) abolished neurotrophin-dependent receptor activity but did not affect downstream signaling by the p75(NTR)/NgR/Lingo-1 complex in response to MAG, indicating the existence of distinct, ligand-specific activation mechanisms for p75(NTR). FRET experiments revealed a close association of p75(NTR) intracellular domains that was transiently disrupted by conformational changes induced upon NGF binding. Although mutation of Cys(257) did not alter the oligomeric state of p75(NTR), the mutant receptor was no longer able to propagate conformational changes to the cytoplasmic domain upon ligand binding. We propose that neurotrophins activate p75(NTR) by a mechanism involving rearrangement of disulphide-linked receptor subunits.


Asunto(s)
Multimerización de Proteína/fisiología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Animales Recién Nacidos , Sitios de Unión/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Células Cultivadas , Chlorocebus aethiops , Cisteína/metabolismo , Proteínas Fluorescentes Verdes/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Mutación/genética , FN-kappa B/metabolismo , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oligopéptidos/genética , Unión Proteica/genética , Conformación Proteica , Multimerización de Proteína/genética , ARN Interferente Pequeño/metabolismo , Ratas , Receptor de Factor de Crecimiento Nervioso/genética , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/fisiología , Ganglio Cervical Superior/citología , Transfección/métodos , Proteína de Unión al GTP rhoA/metabolismo
20.
Stem Cells Dev ; 17(2): 233-43, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18447639

RESUMEN

Mouse embryonic stem (ES) cells grown in feeder-free suspension cultures in the presence of leukemia inhibitory factor (LIF) and basic fibroblast growth factor (bFGF) form spheres that retain pluripotency after multiple passages. ES cell-derived spheres of any passage acquired increased competence to differentiate into neurons over time in culture. Eight-day-old spheres produced many neurons upon plating in differentiation conditions whereas 3-day-old spheres produce none, even after monolayer expansion or treatment with blockers of inhibitory signals, indicating the acquisition of a reversible, proto-neurogenic state during sphere development. Gene expression profiling with oligonucleotide microarrays was used to identify the transcriptional changes accompanying this process. Sphere growth was characterized by down-regulation of a subset of ES cell-expressed genes during the first few days of sphere formation, and progressive up-regulation of novel genes over the course of 1 week in culture. Differential gene expression between 3-day-old and 8 day-old spheres was verified by quantitative real-time PCR experiments. Gene Set Enrichment Analysis (GSEA) of microarray data indicated that neurogenic potential in the late stages of sphere development correlated predominantly with up-regulation of pathways related to mitochondrial function, cell metabolism, oxidative stress, hypoxia, and down-regulation of RNA transcription and proteasome machineries, as well as pathways induced by myc and repressed by retinoic acid. We propose that differences in cellular metabolic state brought about by cell-cell contact and paracrine interactions in the sphere niche may play crucial roles in biasing the early stages of ES cell differentiation toward a neuronal phenotype.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Neuronas/fisiología , Esferoides Celulares/fisiología , Algoritmos , Animales , Diferenciación Celular/genética , Células Cultivadas , Análisis por Conglomerados , Interpretación Estadística de Datos , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Ratones , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proyectos de Investigación , Esferoides Celulares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA