Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Int J Mol Sci ; 22(18)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34576341

RESUMEN

Neurofibromatosis type 1 (NF1) is a common inherited disorder caused by mutations of the NF1 gene that encodes the Ras-GTPase activating protein neurofibromin, leading to overactivation of Ras-dependent signaling pathways such as the mTOR pathway. It is often characterized by a broad range of cognitive symptoms that are currently untreated. The serotonin 5-HT6 receptor is a potentially relevant target in view of its ability to associate with neurofibromin and to engage the mTOR pathway to compromise cognition in several cognitive impairment paradigms. Here, we show that constitutively active 5-HT6 receptors contribute to increased mTOR activity in the brain of Nf1+/- mice, a preclinical model recapitulating some behavioral alterations of NF1. Correspondingly, peripheral administration of SB258585, a 5-HT6 receptor inverse agonist, or rapamycin, abolished deficits in long-term social and associative memories in Nf1+/- mice, whereas administration of CPPQ, a neutral antagonist, did not produce cognitive improvement. These results show a key influence of mTOR activation by constitutively active 5-HT6 receptors in NF1 cognitive symptoms. They provide a proof of concept that 5-HT6 receptor inverse agonists already in clinical development as symptomatic treatments to reduce cognitive decline in dementia and psychoses, might be repurposed as therapies alleviating cognitive deficits in NF1 patients.


Asunto(s)
Neurofibromatosis 1/metabolismo , Receptores de Serotonina/metabolismo , Animales , Humanos , Serotonina/metabolismo , Tiofenos/metabolismo
2.
Sci Signal ; 13(618)2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32047117

RESUMEN

The serotonin (5-hydroxytrypatmine) receptor 5-HT6 (5-HT6R) has emerged as a promising target to alleviate the cognitive symptoms of neurodevelopmental diseases. We previously demonstrated that 5-HT6R finely controls key neurodevelopmental steps, including neuronal migration and the initiation of neurite growth, through its interaction with cyclin-dependent kinase 5 (Cdk5). Here, we showed that 5-HT6R recruited G protein-regulated inducer of neurite outgrowth 1 (GPRIN1) through a Gs-dependent mechanism. Interactions between the receptor and either Cdk5 or GPRIN1 occurred sequentially during neuronal differentiation. The 5-HT6R-GPRIN1 interaction enhanced agonist-independent, receptor-stimulated cAMP production without altering the agonist-dependent response in NG108-15 neuroblastoma cells. This interaction also promoted neurite extension and branching in NG108-15 cells and primary mouse striatal neurons through a cAMP-dependent protein kinase A (PKA)-dependent mechanism. This study highlights the complex allosteric modulation of GPCRs by protein partners and demonstrates how dynamic interactions between GPCRs and their protein partners can control the different steps of highly coordinated cellular processes, such as dendritic tree morphogenesis.


Asunto(s)
AMP Cíclico/metabolismo , Dendritas/metabolismo , Receptores de Serotonina/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Movimiento Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Ratones , Morfogénesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Unión Proteica , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Serotonina/genética
3.
Neuropharmacology ; 126: 128-141, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28844596

RESUMEN

Alzheimer's disease (AD) is the main cause of dementia and a major health issue worldwide. The complexity of the pathology continues to challenge its comprehension and the implementation of effective treatments. In the last decade, a number of possible targets of intervention have been pointed out, among which the stimulation of 5-HT4 receptors (5-HT4Rs) seems very promising. 5-HT4R agonists exert pro-cognitive effects, inhibit amyloid-ß peptide (Aß) production and therefore directly and positively impact AD progression. In the present work, we investigated the effects of RS 67333, a partial 5-HT4R agonist, after chronic administration in the 5xFAD mouse model of AD. 5xFAD male mice and their wild type (WT) male littermates received either RS 67333 or vehicle solution i.p., twice a week, for 2 or 4 months. Cognitive performance was evaluated in a hippocampal-dependent behavioral task, the olfactory tubing maze (OTM). Mice were then sacrificed to evaluate the metabolism of the amyloid precursor protein (APP), amyloidosis and neuroinflammatory processes. No beneficial effects of RS 67333 were observed in 5xFAD mice after 2 months of treatment, while 5xFAD mice treated for 4 months showed better cognitive abilities compared to vehicle-treated 5xFAD mice. The beneficial effects of RS 67333 on learning and memory correlated with the decrease in both amyloid plaque load and neuroinflammation, more specifically in the entorhinal cortex. The most significant improvements in learning and memory and reduction of pathology stigmata were observed after the 4-month administration of RS 67333, demonstrating that treatment duration is important to alleviate amyloidosis and glial reactivity, particularly in the entorhinal cortex. These results confirm the 5-HT4R as a promising target for AD pathogenesis and highlight the need for further investigations to characterize fully the underlying mechanisms of action.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Precursor de Proteína beta-Amiloide/metabolismo , Compuestos de Anilina/administración & dosificación , Corteza Entorrinal/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Piperidinas/administración & dosificación , Agonistas del Receptor de Serotonina 5-HT4/administración & dosificación , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Encefalitis/prevención & control , Corteza Entorrinal/metabolismo , Corteza Entorrinal/patología , Masculino , Ratones Transgénicos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/prevención & control
4.
Biochem J ; 473(13): 1953-65, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27143784

RESUMEN

The activity of serotonergic systems depends on the reuptake of extracellular serotonin via its plasma membrane serotonin [5-HT (5-hydroxytryptamine)] transporter (SERT), a member of the Na(+)/Cl(-)-dependent solute carrier 6 family. SERT is finely regulated by multiple molecular mechanisms including its physical interaction with intracellular proteins. The majority of previously identified SERT partners that control its functional activity are soluble proteins, which bind to its intracellular domains. SERT also interacts with transmembrane proteins, but its association with other plasma membrane transporters remains to be established. Using a proteomics strategy, we show that SERT associates with ASCT2 (alanine-serine-cysteine-threonine 2), a member of the solute carrier 1 family co-expressed with SERT in serotonergic neurons and involved in the transport of small neutral amino acids across the plasma membrane. Co-expression of ASCT2 with SERT in HEK (human embryonic kidney)-293 cells affects glycosylation and cell-surface localization of SERT with a concomitant reduction in its 5-HT uptake activity. Conversely, depletion of cellular ASCT2 by RNAi enhances 5-HT uptake in both HEK-293 cells and primary cultured mesencephalon neurons. Mimicking the effect of ASCT2 down-regulation, treatment of HEK-293 cells and neurons with the ASCT2 inhibitor D-threonine also increases 5-HT uptake. Moreover, D-threonine does not enhance further the maximal velocity of 5-HT uptake in cells depleted of ASCT2. Collectively, these findings provide evidence for a complex assembly involving SERT and a member of another solute carrier family, which strongly influences the subcellular distribution of SERT and the reuptake of 5-HT.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/genética , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Ratones , Antígenos de Histocompatibilidad Menor/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Unión Proteica , Interferencia de ARN , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Treonina/farmacología
5.
Physiol Rev ; 95(4): 1157-87, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26269525

RESUMEN

TOR (target of rapamycin) and its mammalian ortholog mTOR have been discovered in an effort to understand the mechanisms of action of the immunosuppressant drug rapamycin extracted from a bacterium of the Easter Island (Rapa Nui) soil. mTOR is a serine/threonine kinase found in two functionally distinct complexes, mTORC1 and mTORC2, which are differentially regulated by a great number of nutrients such as glucose and amino acids, energy (oxygen and ATP/AMP content), growth factors, hormones, and neurotransmitters. mTOR controls many basic cellular functions such as protein synthesis, energy metabolism, cell size, lipid metabolism, autophagy, mitochondria, and lysosome biogenesis. In addition, mTOR-controlled signaling pathways regulate many integrated physiological functions of the nervous system including neuronal development, synaptic plasticity, memory storage, and cognition. Thus it is not surprising that deregulation of mTOR signaling is associated with many neurological and psychiatric disorders. Preclinical and preliminary clinical studies indicate that inhibition of mTORC1 can be beneficial for some pathological conditions such as epilepsy, cognitive impairment, and brain tumors, whereas stimulation of mTORC1 (direct or indirect) can be beneficial for other pathologies such as depression or axonal growth and regeneration.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Fenómenos Fisiológicos Celulares/fisiología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Encéfalo/fisiología , Humanos
6.
Nat Chem Biol ; 10(7): 590-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24880860

RESUMEN

The serotonin6 receptor (5-HT6R) is a promising target for treating cognitive deficits of schizophrenia often linked to alterations of neuronal development. This receptor controls neurodevelopmental processes, but the signaling mechanisms involved remain poorly understood. Using a proteomic strategy, we show that 5-HT6Rs constitutively interact with cyclin-dependent kinase 5 (Cdk5). Expression of 5-HT6Rs in NG108-15 cells induced neurite growth and expression of voltage-gated Ca(2+) channels, two hallmarks of neuronal differentiation. 5-HT6R-elicited neurite growth was agonist independent and prevented by the 5-HT6R antagonist SB258585, which behaved as an inverse agonist. Moreover, it required receptor phosphorylation at Ser350 by Cdk5 and Cdc42 activity. Supporting a role of native 5-HT6Rs in neuronal differentiation, neurite growth of primary neurons was reduced by SB258585, by silencing 5-HT6R expression or by mutating Ser350 into alanine. These results reveal a functional interplay between Cdk5 and a G protein-coupled receptor to control neuronal differentiation.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/genética , Hipocampo/metabolismo , Neuritas/ultraestructura , Receptores de Serotonina/genética , Animales , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Quinasa 5 Dependiente de la Ciclina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Humanos , Ligandos , Ratones , Mutación , Neuritas/metabolismo , Fosforilación , Piperazinas/farmacología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
7.
ACS Chem Neurosci ; 4(1): 130-40, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23336052

RESUMEN

In addition to the amyloidogenic pathway, amyloid precursor protein (APP) can be cleaved by α-secretases, producing soluble and neuroprotective APP alpha (sAPPα) (nonamyloidogenic pathway) and thus preventing the generation of pathogenic amyloid-ß. However, the mechanisms regulating APP cleavage by α-secretases remain poorly understood. Here, we showed that expression of serotonin type 4 receptors (5-HT(4)Rs) constitutively (without agonist stimulation) induced APP cleavage by the α-secretase ADAM10 and the release of neuroprotective sAPPα in HEK-293 cells and cortical neurons. This effect was independent of cAMP production. Interestingly, we demonstrated that 5-HT(4) receptors physically interacted with the mature form of ADAM10. Stimulation of 5-HT(4) receptors by an agonist further increased sAPPα secretion, and this effect was mediated by cAMP/Epac signaling. These findings describe a new mechanism whereby a GPCR constitutively stimulates the cleavage of APP by α-secretase and promotes the nonamyloidogenic pathway of APP processing.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Serotonina 5-HT4/fisiología , Proteína ADAM10 , Enfermedad de Alzheimer/etiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Benzofuranos/farmacología , Corteza Cerebral/citología , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Fármacos Neuroprotectores/farmacología , Receptores Acoplados a Proteínas G/fisiología , Receptores de Serotonina 5-HT4/metabolismo , Agonistas del Receptor de Serotonina 5-HT4/farmacología
8.
Brain Res ; 1511: 65-72, 2013 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-23148949

RESUMEN

G protein-coupled receptors (GPCRs) can activate simultaneously multiple signaling pathways upon agonist binding. The combined use of engineered GPCRs, such as the receptors activated solely by synthetic ligands (RASSLs), and of biased ligands that activate only one pathway at a time might help deciphering the physiological role of each G protein signaling. In order to find serotonin type 4 receptor (5-HT4R) biased ligands, we analyzed the ability of several compounds to activate the Gs and G(q/11) pathways in COS-7 cells that transiently express wild type 5-HT4R, the 5-HT4R-D(100)A mutant (known also as 5-HT4-RASSL, or Rs1) or the 5-HT4R-T(104)A mutant, which modifies agonist-induced 5-HT4R activation. This analysis allowed completing the pharmacological profile of the two mutant 5-HT4Rs, but we did not find any biased ligand for the mutant receptors. Conversely, we identified the first biased agonists for wild type 5-HT4R. Indeed, RS 67333 and prucalopride acted as partial agonists to induce cAMP accumulation, but as antagonists on inositol phosphate production. Moreover, they showed very different antagonist potencies that could be exploited to study the activation of the G(s) pathway, with or without concomitant block of G(q/11) signaling. This article is part of a Special Issue entitled Optogenetics (7th BRES).


Asunto(s)
Ligandos , Receptores de Serotonina 5-HT4/genética , Receptores de Serotonina 5-HT4/metabolismo , Animales , Chlorocebus aethiops , AMP Cíclico/metabolismo , Fosfatos de Inositol/metabolismo , Mutación/genética , Serotoninérgicos/farmacología , Transfección
9.
J Neurosci ; 31(29): 10677-88, 2011 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-21775611

RESUMEN

Recent studies suggest that blood-brain barrier (BBB) permeability contributes to epileptogenesis in symptomatic epilepsies. We have previously described angiogenesis, aberrant vascularization, and BBB alteration in drug-refractory temporal lobe epilepsy. Here, we investigated the role of vascular endothelial growth factor (VEGF) in an in vitro integrative model of vascular remodeling induced by epileptiform activity in rat organotypic hippocampal cultures. After kainate-induced seizure-like events (SLEs), we observed an overexpression of VEGF and VEGF receptor-2 (VEGFR-2) as well as receptor activation. Vascular density and branching were significantly increased, whereas zonula occludens 1 (ZO-1), a key protein of tight junctions (TJs), was downregulated. These effects were fully prevented by VEGF neutralization. Using selective inhibitors of VEGFR-2 signaling pathways, we found that phosphatidylinositol 3-kinase is involved in cell survival, protein kinase C (PKC) in vascularization, and Src in ZO-1 regulation. Recombinant VEGF reproduced the kainate-induced vascular changes. As in the kainate model, VEGFR-2 and Src were involved in ZO-1 downregulation. These results showed that VEGF/VEGFR-2 initiates the vascular remodeling induced by SLEs and pointed out the roles of PKC in vascularization and Src in TJ dysfunction, respectively. This suggests that Src pathway could be a therapeutic target for BBB protection in epilepsies.


Asunto(s)
Ondas Encefálicas/fisiología , Regulación hacia Abajo/fisiología , Endotelio Vascular/fisiología , Hipocampo/fisiología , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Anestésicos Locales/farmacología , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Ondas Encefálicas/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Interacciones Farmacológicas , Endotelio Vascular/efectos de los fármacos , Hipocampo/efectos de los fármacos , Ácido Kaínico/farmacología , L-Lactato Deshidrogenasa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Propidio , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Teprotido/farmacología , Tetrodotoxina/farmacología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de la Zonula Occludens-1
10.
Genome Med ; 3(7): 43, 2011 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-21745417

RESUMEN

We propose an innovative, integrated, cost-effective health system to combat major non-communicable diseases (NCDs), including cardiovascular, chronic respiratory, metabolic, rheumatologic and neurologic disorders and cancers, which together are the predominant health problem of the 21st century. This proposed holistic strategy involves comprehensive patient-centered integrated care and multi-scale, multi-modal and multi-level systems approaches to tackle NCDs as a common group of diseases. Rather than studying each disease individually, it will take into account their intertwined gene-environment, socio-economic interactions and co-morbidities that lead to individual-specific complex phenotypes. It will implement a road map for predictive, preventive, personalized and participatory (P4) medicine based on a robust and extensive knowledge management infrastructure that contains individual patient information. It will be supported by strategic partnerships involving all stakeholders, including general practitioners associated with patient-centered care. This systems medicine strategy, which will take a holistic approach to disease, is designed to allow the results to be used globally, taking into account the needs and specificities of local economies and health systems.

11.
J Biol Chem ; 285(3): 1989-2002, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19915011

RESUMEN

Strategies based on activating GLP-1 receptor (GLP-1R) are intensively developed for the treatment of type 2 diabetes. The exhaustive knowledge of the signaling pathways linked to activated GLP-1R within the beta-cells is of major importance. In beta-cells, GLP-1 activates the ERK1/2 cascade by diverse pathways dependent on either Galpha(s)/cAMP/cAMP-dependent protein kinase (PKA) or beta-arrestin 1, a scaffold protein. Using pharmacological inhibitors, beta-arrestin 1 small interfering RNA, and islets isolated from beta-arrestin 1 knock-out mice, we demonstrate that GLP-1 stimulates ERK1/2 by two temporally distinct pathways. The PKA-dependent pathway mediates rapid and transient ERK1/2 phosphorylation that leads to nuclear translocation of the activated kinases. In contrast, the beta-arrestin 1-dependent pathway produces a late ERK1/2 activity that is restricted to the beta-cell cytoplasm. We further observe that GLP-1 phosphorylates the cytoplasmic proapoptotic protein Bad at Ser-112 but not at Ser-155. We find that the beta-arrestin 1-dependent ERK1/2 activation engaged by GLP-1 mediates the Ser-112 phosphorylation of Bad, through p90RSK activation, allowing the association of Bad with the scaffold protein 14-3-3, leading to its inactivation. beta-Arrestin 1 is further found to mediate the antiapoptotic effect of GLP-1 in beta-cells through the ERK1/2-p90RSK-phosphorylation of Bad. This new regulatory mechanism engaged by activated GLP-1R involving a beta-arrestin 1-dependent spatiotemporal regulation of the ERK1/2-p90RSK activity is now suspected to participate in the protection of beta-cells against apoptosis. Such signaling mechanism may serve as a prototype to generate new therapeutic GLP-1R ligands.


Asunto(s)
Apoptosis/efectos de los fármacos , Arrestinas/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Células Secretoras de Insulina/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Proteínas 14-3-3/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteína Letal Asociada a bcl/química , beta-Arrestina 1 , beta-Arrestinas
12.
EMBO J ; 28(18): 2706-18, 2009 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-19661922

RESUMEN

G protein-coupled receptors (GPCRs) have been found to trigger G protein-independent signalling. However, the regulation of G protein-independent pathways, especially their desensitization, is poorly characterized. Here, we show that the G protein-independent 5-HT(4) receptor (5-HT(4)R)-operated Src/ERK (extracellular signal-regulated kinase) pathway, but not the G(s) pathway, is inhibited by GPCR kinase 5 (GRK5), physically associated with the proximal region of receptor' C-terminus in both human embryonic kidney (HEK)-293 cells and colliculi neurons. This inhibition required two sequences of events: the association of beta-arrestin1 to a phosphorylated serine/threonine cluster located within the receptor C-t domain and the phosphorylation, by GRK5, of beta-arrestin1 (at Ser(412)) bound to the receptor. Phosphorylated beta-arrestin1 in turn prevented activation of Src constitutively bound to 5-HT(4)Rs, a necessary step in receptor-stimulated ERK signalling. This is the first demonstration that beta-arrestin1 phosphorylation by GRK5 regulates G protein-independent signalling.


Asunto(s)
Arrestinas/biosíntesis , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Receptores de Serotonina 5-HT4/metabolismo , Línea Celular , Proteínas de Unión al GTP/metabolismo , Humanos , Mutación , Neuronas/metabolismo , Péptidos/química , Fosforilación , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Serina/química , beta-Arrestinas , Familia-src Quinasas/metabolismo
13.
J Physiol ; 587(1): 101-13, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19001039

RESUMEN

Extensive work has shown that activation of the cAMP-dependent protein kinase A (PKA) is crucial for long-term depression (LTD) of synaptic transmission in the hippocampus, a phenomenon that is thought to be involved in memory formation. Here we studied the role of an alternative target of cAMP, the exchange protein factor directly activated by cyclic AMP (Epac). We show that pharmacological activation of Epac by the selective agonist 8-(4-chlorophenylthio)-2'-O-methyl-cAMP (8-pCPT) induces LTD in the CA1 region. Paired-pulse facilitation of synaptic responses remained unchanged after induction of this LTD, suggesting that it depended on postsynaptic mechanisms. The 8-pCPT-induced LTD was blocked by the Epac signalling inhibitor brefeldin-A (BFA), Rap-1 antagonist geranylgeranyltransferase inhibitor (GGTI) and p38 mitogen activated protein kinase (P38-MAPK) inhibitor SB203580. This indicated a direct involvement of Epac in this form of LTD. As for other forms of LTD, a mimetic peptide of the PSD-95/Disc-large/ZO-1 homology (PDZ) ligand motif of the AMPA receptor subunit GluR2 blocked the Epac-LTD, suggesting involvement of PDZ protein interaction. The Epac-LTD also depended on mobilization of intracellular Ca(2+), proteasome activity and mRNA translation, but not transcription, as it was inhibited by thapsigargin, lactacystin and anisomycin, but not actinomycin-D, respectively. Finally, we found that the pituitary adenylate cyclase activating polypeptide (PACAP) can induce an LTD that was mutually occluded by the Epac-LTD and blocked by BFA or SB203580, suggesting that the Epac-LTD could be mobilized by stimulation of PACAP receptors. Altogether these results provided evidence for a new form of hippocampal LTD.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Hipocampo/fisiología , Depresión Sináptica a Largo Plazo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Animales , Brefeldino A/farmacología , Calcio/metabolismo , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Glucógeno Sintasa Quinasa 3/fisiología , Glucógeno Sintasa Quinasa 3 beta , Factores de Intercambio de Guanina Nucleótido/agonistas , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Complejo de la Endopetidasa Proteasomal/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Receptores AMPA/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
14.
J Biol Chem ; 284(7): 4332-42, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19074139

RESUMEN

In pancreatic beta-cells, the pituitary adenylate cyclase-activating polypeptide (PACAP) exerts a potent insulin secretory effect via PAC(1) and VPAC receptors (Rs) through the Galpha(s)/cAMP/protein kinase A pathway. Here, we investigated the mechanisms linking PAC(1)R to ERK1/2 activation in INS-1E beta-cells and pancreatic islets. PACAP caused a transient (5 min) increase in ERK1/2 phosphorylation via PAC(1)Rs and promoted nuclear translocation of a fraction of cytosolic p-ERK1/2. Both protein kinase A- and Src-dependent pathways mediated this transient ERK1/2 activation. Moreover, PACAP potentiated glucose-induced long-lasting ERK1/2 activation. Blocking Ca(2+) influx abolished glucose-induced ERK1/2 activation and PACAP potentiating effect. Glucose stimulation during KCl depolarization showed that, in addition to the triggering signal (rise in cytosolic [Ca(2+)]), the amplifying pathway was also involved in glucose-induced sustained ERK1/2 activation and was required for PACAP potentiation. The finding that at 30 min glucose-induced p-ERK1/2 was detected in both cytosol and nucleus while the potentiating effect of PACAP was only observed in the cytosol, suggested the involvement of the scaffold protein beta-arrestin. Indeed, beta-arrestin 1 (beta-arr1) depletion (in beta-arr1 knockout mouse islets or in INS-1E cells by siRNA) completely abolished PACAP potentiation of long-lasting ERK1/2 activation by glucose. Finally, PACAP potentiated glucose-induced CREB transcriptional activity and IRS-2 mRNA expression mainly via the ERK1/2 signaling pathway, and likewise, beta-arr1 depletion reduced the PACAP potentiating effect on IRS-2 expression. These results establish for the first time that PACAP potentiates glucose-induced long-lasting ERK1/2 activation via a beta-arr1-dependent pathway and thus provide new insights concerning the mechanisms of PACAP and glucose actions in pancreatic beta-cells.


Asunto(s)
Arrestinas/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Edulcorantes/farmacología , Animales , Arrestinas/genética , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glucosa/metabolismo , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Células Secretoras de Insulina/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Edulcorantes/metabolismo , Factores de Tiempo , beta-Arrestina 1 , beta-Arrestinas
15.
Mol Biol Cell ; 19(11): 4640-50, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18768750

RESUMEN

The serotonin (5-hydroxytryptamine; 5-HT)(2C) receptor is a G protein-coupled receptor (GPCR) exclusively expressed in CNS that has been implicated in numerous brain disorders, including anxio-depressive states. Like many GPCRs, 5-HT(2C) receptors physically interact with a variety of intracellular proteins in addition to G proteins. Here, we show that calmodulin (CaM) binds to a prototypic Ca(2+)-dependent "1-10" CaM-binding motif located in the proximal region of the 5-HT(2C) receptor C-terminus upon receptor activation by 5-HT. Mutation of this motif inhibited both beta-arrestin recruitment by 5-HT(2C) receptor and receptor-operated extracellular signal-regulated kinase (ERK) 1,2 signaling in human embryonic kidney-293 cells, which was independent of G proteins and dependent on beta-arrestins. A similar inhibition was observed in cells expressing a dominant-negative CaM or depleted of CaM by RNA interference. Expression of the CaM mutant also prevented receptor-mediated ERK1,2 phosphorylation in cultured cortical neurons and choroid plexus epithelial cells that endogenously express 5-HT(2C) receptors. Collectively, these findings demonstrate that physical interaction of CaM with recombinant and native 5-HT(2C) receptors is critical for G protein-independent, arrestin-dependent receptor signaling. This signaling pathway might be involved in neurogenesis induced by chronic treatment with 5-HT(2C) receptor agonists and their antidepressant-like activity.


Asunto(s)
Arrestinas/metabolismo , Calmodulina/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Calcio/farmacología , Línea Celular , Plexo Coroideo/citología , Activación Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Receptor de Serotonina 5-HT2C/química , Transducción de Señal/efectos de los fármacos , beta-Arrestinas , Proteínas ras/metabolismo
16.
J Neurosci ; 28(34): 8604-14, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18716219

RESUMEN

The metabotropic glutamate receptor 7 (mGluR7) is widely expressed throughout the brain and primarily localized at presynaptic active zones, where it is thought to regulate neurotransmitter release. Protein interacting with C kinase 1 (PICK1), a postsynaptic density protein-95/disc-large tumor suppressor protein/zonula occludens-1 (PDZ)-domain protein, binds to the three C-terminal amino acids (-LVI) of the predominant mGluR7 splice variant, mGluR7a, and has been implicated in the synaptic clustering of this receptor. Here, we generated knock-in mice in which the C-terminal LVI coding sequence of exon 10 of the mGluR7 gene was replaced by three alanine codons (-AAA). Immunoprecipitation showed that the PICK1-mGluR7a interaction is disrupted in mGluR7a(AAA/AAA) mice. However, the synaptic localization of mGluR7a was not altered in cultured hippocampal neurons and brain sections prepared from the knock-in animals. In cerebellar granule cell cultures, the group III mGluR agonist l-AP-4 decreased the frequency of spontaneous excitatory currents in neurons derived from wild-type but not mGluR7a(AAA/AAA) mice, consistent with the interaction between mGluR7a and PICK1 being required for protein kinase C-mediated inhibition of glutamate release. At the behavioral level, the mGluR7a(AAA/AAA) mice showed no deficits in motor coordination, pain sensitivity, and anxiety but exhibited significant defects in hippocampus-dependent spatial working memory. In addition, they displayed a high susceptibility to the convulsant drug pentylenetetrazole. Together, these results indicate that PICK1 binding to the C-terminal region of mGluR7a is crucial for agonist-triggered presynaptic signaling in vivo.


Asunto(s)
Proteínas Portadoras/metabolismo , Convulsivantes , Antagonistas de Aminoácidos Excitadores , Trastornos de la Memoria/genética , Proteínas Nucleares/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Glutamato Metabotrópico/genética , Convulsiones/genética , Secuencias de Aminoácidos/genética , Animales , Conducta Animal , Encéfalo/patología , Proteínas de Ciclo Celular , Células Cultivadas , Cerebelo/patología , Cerebelo/fisiopatología , Predisposición Genética a la Enfermedad , Ácido Glutámico , Ligandos , Ratones , Ratones Transgénicos , Mutación , Inhibición Neural , Oocitos , Pentilenotetrazol , Terminales Presinápticos , Isoformas de Proteínas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Convulsiones/inducido químicamente , Transducción de Señal , Percepción Espacial , Sinapsis , Xenopus laevis
17.
Neuropharmacology ; 55(6): 922-31, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18603269

RESUMEN

Twenty years ago, we started the characterization of a 5-HT receptor coupled to cAMP production in neurons. This receptor obviously had a different pharmacology to the other 5-HT receptors described at that time, i.e. the 5-HT(1), 5-HT(2), 5-HT(3) receptors. We proposed to name it the 5-HT(4) receptor. Nowadays, 5-HT(4) receptors are one of the most studied GPCRs belonging to the "rhodopsin" family. Thanks to the existence of a great variety of ligands with inverse agonist, partial agonist, agonist and antagonist profiles, the pharmacological and physiological properties of this receptor are beginning to emerge. Although some 5-HT(4) partial agonists have been on the market for gastro-intestinal pathologies, 5-HT(4) receptor drugs have still to be commercialized for brain disorders. However, since 5-HT(4) receptors have recognized effects on memory, depression and feeding in animal models, there is still hope for a therapeutic destiny of this interesting target in brain disorders.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Receptores de Serotonina , Serotoninérgicos/farmacología , Animales , AMP Cíclico/metabolismo , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Modelos Moleculares , Receptores de Serotonina/genética , Receptores de Serotonina/historia , Receptores de Serotonina/metabolismo
18.
Nat Neurosci ; 11(8): 940-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18641645

RESUMEN

Absence epilepsy is a neurological disorder that causes a recurrent loss of consciousness and generalized spike-and-wave discharges on an electroencephalogram (EEG). The role of metabotropic glutamate receptors (mGluRs) and associated scaffolding proteins in absence epilepsy has been unclear to date. We investigated a possible role for these proteins in absence epilepsy, focusing on the mGluR7a receptor and its PDZ-interacting protein, protein interacting with C kinase 1 (PICK1), in rats and mice. Injection of a cell-permeant dominant-negative peptide or targeted mutation of the mGluR7a C terminus, both of which disrupt the interaction between the receptor and PDZ proteins, caused behavioral symptoms and EEG discharges that are characteristic of absence epilepsy. Inactivation of the Pick1 gene also facilitated pharmacological induction of the absence epilepsy phenotype. The cortex and thalamus, which are known to participate in absence epilepsy, were involved, but the hippocampus was not. Our results indicate that disruption of the mGluR7a-PICK1 complex is sufficient to induce absence epilepsy-like seizures in rats and mice, thus providing, to the best of our knowledge, the first animal model of metabotropic glutamate receptor-PDZ protein interaction in absence epilepsy.


Asunto(s)
Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Epilepsia Tipo Ausencia/fisiopatología , Proteínas Nucleares/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Células Cultivadas , Electrodos Implantados , Electroencefalografía/efectos de los fármacos , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Proteínas Nucleares/genética , Péptidos/farmacología , Fenotipo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología
19.
Trends Pharmacol Sci ; 28(12): 602-7, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18001849

RESUMEN

A large body of evidence indicates that agonists of some G protein-coupled receptors (GPCRs) can activate growth factor receptor tyrosine kinases (RTKs) in the absence of added growth factor. This phenomenon, called transactivation, is an important pathway that contributes to growth-promoting activity of many GPCR ligands. Reciprocally, recent advances indicate that RTKs utilize GPCR signalling molecules to transduce signals and that RTK ligands themselves can transactivate GPCRs. This novel transactivation process, which places GPCR signalling downstream of RTKs, either requires the production of a GPCR ligand of the transactivated GPCR or occurs in a ligand independent manner within an integrated signalling network. Here, we provide an overview of the molecular mechanisms involved in this novel cross-communication between GPCRs and RTKs and discuss its relevance in the specification of growth factor signalling and functions.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/fisiología , Animales , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Receptores de Lisoesfingolípidos/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología
20.
Brain ; 130(Pt 7): 1942-56, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17533168

RESUMEN

Previous studies from our group, focusing on neuro-glial remodelling in human temporal lobe epilepsy (TLE), have shown the presence of immature vascular cells in various areas of the hippocampus. Here, we investigated angiogenic processes in hippocampi surgically removed from adult patients suffering from chronic intractable TLE, with various aetiologies. We compared hippocampi from TLE patients to hippocampi obtained after surgery or autopsy from non-epileptic patients (NE). We quantified the vascular density, checked for the expression of angiogenic factors and their receptors and looked for any blood-brain barrier (BBB) leakage. We used a relevant model of rat limbic epilepsy, induced by lithium-pilocarpine treatment, to understand the sequence of events. In humans, the vessel density was significantly higher in TLE than in NE patients. This was neither dependent on the aetiology nor on the degree of neuronal loss, but was positively correlated with seizure frequency. In the whole hippocampus, we observed many complex, tortuous microvessels. In the dentate gyrus, when the granular layer was dispersed, long microvessels appeared radially orientated. Vascular endothelial factor (VEGF) and tyrosine kinase receptors were detected in different types of cells. An impairment of the BBB was demonstrated by the loss of tight junctions and by Immunoglobulines G (IgG) leakage and accumulation in neurons. In the rat model of TLE, VEGF over-expression and BBB impairment occurred early after status epilepticus, followed by a progressive increase in vascularization. In humans and rodents, angiogenic processes and BBB disruption were still obvious in the chronic focus, probably activated by recurrent seizures. We suggest that the persistent leakage of serum IgG in the interstitial space and their uptake by neurons may participate in hypoperfusion and in neuronal dysfunction occurring in TLE.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Epilepsia del Lóbulo Temporal/complicaciones , Neovascularización Patológica/etiología , Adolescente , Adulto , Inductores de la Angiogénesis/metabolismo , Animales , Encéfalo/patología , Muerte Celular , Niño , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/fisiopatología , Femenino , Hipocampo/irrigación sanguínea , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Neuronas/patología , Permeabilidad , Ratas , Ratas Sprague-Dawley , Receptor TIE-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA