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










Base de datos
Intervalo de año de publicación
1.
J Ethnopharmacol ; 329: 118133, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38580187

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Angelica roots are a significant source of traditional medicines for various cultures around the northern hemisphere, from indigenous communities in North America to Japan. Among its many applications, the roots are used to treat type 2 diabetes mellitus; however, this application is not mentioned often. Ethnopharmacological studies have reported the use of A. japonica var. hirsutiflora, A. furcijuga, A. shikokiana, and A. keiskei to treat diabetes symptoms, and further reports have demonstrated the three angelica roots, i.e., A. japonica var. hirsutiflora, A. reflexa, and A. dahurica, exhibit insulin secretagogue activity. AIM OF THE STUDY: This study aimed to phytochemically characterize and compare angelica roots monographed in the European Pharmacopeia 11th, isolate major plant metabolites, and assess extracts and isolates' capability to modulate pancreatic ß-cell function. MATERIALS AND METHODS: Root extracts of Angelica archangelica, Angelica dahurica, Angelica biserrata, and Angelica sinensis were phytochemically profiled using liquid chromatography method coupled with mass spectrometry. Based on this analysis, simple and furanocoumarins were isolated using chromatography techniques. Extracts (1.6-50 µg/mL) and isolated compounds (5-40 µmol/L) were studied for their ability to modulate insulin secretion in the rat insulinoma INS-1 pancreatic ß-cell model. Insulin was quantified by the homogeneous time-resolved fluorescence method. RESULTS: Forty-one secondary metabolites, mostly coumarins, were identified in angelica root extracts. A. archangelica, A. dahurica, and A. biserrata root extracts at concentration of 12.5-50 µg/mL potentiated glucose-induced insulin secretion, which correlated with their high coumarin content. Subsequently, 23 coumarins were isolated from these roots and screened using the same protocol. Coumarins substituted with the isoprenyl group were found to be responsible for the extracts' insulinotropic effect. CONCLUSIONS: Insulinotropic effects of three pharmacopeial angelica roots were found, the metabolite profiles and pharmacological activities of the roots were correlated, and key structures responsible for the modulation of pancreatic ß-cell function were identified. These findings may have implications for the traditional use of angelica roots in treating diabetes. Active plant metabolites may also become lead structures in the search for new antidiabetic treatments.


Asunto(s)
Angelica , Células Secretoras de Insulina , Fitoquímicos , Extractos Vegetales , Raíces de Plantas , Angelica/química , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/química , Animales , Fitoquímicos/farmacología , Fitoquímicos/aislamiento & purificación , Fitoquímicos/análisis , Ratas , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Cumarinas/farmacología , Cumarinas/aislamiento & purificación , Hipoglucemiantes/farmacología , Hipoglucemiantes/aislamiento & purificación , Hipoglucemiantes/química
2.
Biochem Pharmacol ; 202: 115114, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35659880

RESUMEN

The growth hormone secretagogue receptor (GHSR) is a G protein-coupled receptor that regulates essential physiological functions. In particular, activation of GHSR in response to its endogenous agonist ghrelin promotes food intake and blood glucose increase. Therefore, compounds aimed at blocking GHSR signaling constitute potential options against obesity-related metabolic disorders. We have previously developed potent ligands of GHSR based on a triazole scaffold. Here, we report a new 3,4,5-trisubstituted 1,2,4-triazole compound, named JMV 6616, that potently blocks GHSR activity in vitro and in vivo. Specifically, in HEK293T cells JMV 6616 behaves as an inverse agonist since it binds to GHSR and inhibits its ghrelin-independent signaling. Accordingly, using purified labeled GHSR assembled into lipid nanodiscs we found that JMV 6616 decreases GHSR-catalyzed G protein activation and stabilizes an inactive receptor conformation. Importantly, JMV 6616 also acts on native GHSR since it blocks the insulinostatic effect of ghrelin in pancreatic islets. In mice, JMV 6616 inhibits blood glucose-raising effects of ghrelin treatment and the orexigenic actions of acute ghrelin administration. Together, our data suggest that this triazole-derived modulator of GHSR holds promise to mitigate several pathological features associated with eating and metabolic disorders.


Asunto(s)
Ghrelina , Receptores de Ghrelina , Animales , Glucemia , Ghrelina/metabolismo , Ghrelina/farmacología , Células HEK293 , Humanos , Ratones , Triazoles/farmacología
3.
Fundam Clin Pharmacol ; 36(2): 375-377, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34449915

RESUMEN

The hormone ghrelin is the endogenous agonist of the G protein-coupled receptor (GPCR) termed growth hormone secretagogue receptor (GHSR). Ghrelin inhibits glucose-stimulated insulin secretion by activating pancreatic GHSR. Recently, Liver-Expressed Antimicrobial Peptide 2 (LEAP2) was recognized as an endogenous GHSR ligand that blocks ghrelin-induced actions. Nonetheless, the effect of LEAP2 on glucose-stimulated insulin secretion from pancreatic islets is unknown. We aimed at exploring the activity of LEAP2 on glucose-stimulated insulin secretion. Islets of Langerhans isolated from rat pancreas were exposed to glucose in the presence or in the absence of LEAP2 and ghrelin and then insulin secretion was assayed. LEAP2 did not modulate glucose-stimulated insulin secretion. However, LEAP2 blocked the insulinostatic action of ghrelin. Our data show that LEAP2 behaves as an antagonist of pancreatic GHSR.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Ghrelina , Insulina , Islotes Pancreáticos , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Ghrelina/metabolismo , Ghrelina/farmacología , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Hígado , Ratas , Receptores de Ghrelina/metabolismo
4.
J Med Chem ; 63(19): 10796-10815, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32882134

RESUMEN

GHSR controls, among others, growth hormone and insulin secretion, adiposity, feeding, and glucose metabolism. Therefore, an inverse agonist ligand capable of selectively targeting GHSR and reducing its high constitutive activity appears to be a good candidate for the treatment of obesity-related metabolic diseases. In this context, we present a study that led to the development of several highly potent and selective inverse agonists of GHSR based on the 1,2,4-triazole scaffold. We demonstrate that, depending on the nature of the substituents on positions 3, 4, and 5, this scaffold leads to ligands that exert an intrinsic inverse agonist activity on GHSR-catalyzed G protein activation through the stabilization of a specific inactive receptor conformation. Thanks to an in vivo evaluation, we also show that one of the most promising ligands not only exerts an effect on insulin secretion in rat pancreatic islets but also affects the orexigenic effects of ghrelin in mice.


Asunto(s)
Receptores de Ghrelina/agonistas , Triazoles/farmacología , Animales , Agonismo Inverso de Drogas , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ligandos , Ratas , Triazoles/química
5.
Fundam Clin Pharmacol ; 34(5): 571-580, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32083757

RESUMEN

Polyphenols exert pharmacological actions through protein-mediated mechanisms and by modulating intracellular signalling pathways. We recently showed that a gut-microbial metabolite of ellagic acid named urolithin C is a glucose-dependent activator of insulin secretion acting by facilitating L-type Ca2+ channel opening and Ca2+ influx into pancreatic ß-cells. However, it is still unknown whether urolithin C regulates key intracellular signalling proteins in ß-cells. Here, we report that urolithin C enhanced glucose-induced extracellular signal-regulated kinases 1/2 (ERK1/2) activation as shown by higher phosphorylation levels in INS-1 ß-cells. Interestingly, inhibition of ERK1/2 with two structurally distinct inhibitors led to a reduction in urolithin C effect on insulin secretion. Finally, we provide data to suggest that urolithin C-mediated ERK1/2 phosphorylation involved insulin signalling in INS-1 cells. Together, these data indicate that the pharmacological action of urolithin C on insulin secretion relies, in part, on its capacity to enhance glucose-induced ERK1/2 activation. Therefore, our study extends our understanding of the pharmacological action of urolithin C in ß-cells. More generally, our findings revealed that urolithin C modulated the activation of key multifunctional intracellular signalling kinases which participate in the regulation of numerous biological processes.


Asunto(s)
Glucosa/metabolismo , Taninos Hidrolizables/farmacología , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Animales , Línea Celular/metabolismo , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratas
6.
Neurotherapeutics ; 17(1): 17-42, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31919661

RESUMEN

G protein-coupled receptors (GPCRs) constitute the largest class of cell surface signaling receptors and regulate major neurobiological processes. Accordingly, GPCRs represent primary targets for the treatment of brain disorders. Several human genetic polymorphisms affecting GPCRs have been associated to different components of alcohol use disorder (AUD). Moreover, GPCRs have been reported to contribute to several features of alcohol-related behaviors in animal models. Besides traditional pharmacological tools, genetic-based approaches mostly aimed at deleting GPCR genes provided substantial information on how key GPCRs drive alcohol-related behaviors. In this review, we summarize the alcohol phenotypes that ensue from genetic manipulation, in particular gene deletion, of key GPCRs in rodents. We focused on GPCRs that belong to fundamental neuronal systems that have been shown as potential targets for the development of AUD treatment. Data are reviewed with particular emphasis on alcohol reward, seeking, and consumption which are behaviors that capture essential aspects of AUD. Literature survey indicates that in most cases, there is still a gap in defining the intracellular transducers and the functional crosstalk of GPCRs as well as the neuronal populations in which their signaling regulates alcohol actions. Further, the implication of only a few orphan GPCRs has been so far investigated in animal models. Combining advanced pharmacological technologies with more specific genetically modified animals and behavioral preclinical models is likely necessary to deepen our understanding in how GPCR signaling contributes to AUD and for drug discovery.


Asunto(s)
Alcoholismo/genética , Encéfalo/fisiopatología , Receptores Acoplados a Proteínas G/genética , Consumo de Bebidas Alcohólicas/genética , Animales , Humanos , Ratones , Ratones Transgénicos , Neuronas/fisiología , Recompensa , Transducción de Señal
7.
Br J Pharmacol ; 176(20): 4065-4078, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31378934

RESUMEN

BACKGROUND AND PURPOSE: The pharmacology of polyphenol metabolites on beta-cell function is largely undetermined. We sought to identify polyphenol metabolites that enhance the insulin-secreting function of beta-cells and to explore the underlying mechanisms. EXPERIMENTAL APPROACH: INS-1 beta-cells and rat isolated islets of Langerhans or perfused pancreas preparations were used for insulin secretion experiments. Molecular modelling, intracellular Ca2+ monitoring, and whole-cell patch-clamp recordings were used for mechanistic studies. KEY RESULTS: Among a set of polyphenol metabolites, we found that exposure of INS-1 beta-cells to urolithins A and C enhanced glucose-stimulated insulin secretion. We further characterized the activity of urolithin C and its pharmacological mechanism. Urolithin C glucose-dependently enhanced insulin secretion in isolated islets of Langerhans and perfused pancreas preparations. In the latter, enhancement was reversible when glucose was lowered from a stimulating to a non-stimulating concentration. Molecular modelling suggested that urolithin C could dock into the Cav 1.2 L-type Ca2+ channel. Calcium monitoring indicated that urolithin C had no effect on basal intracellular Ca2+ but enhanced depolarization-induced increase in intracellular Ca2+ in INS-1 cells and dispersed cells isolated from islets. Electrophysiology studies indicated that urolithin C dose-dependently enhanced the L-type Ca2+ current for levels of depolarization above threshold and shifted its voltage-dependent activation towards more negative potentials in INS-1 cells. CONCLUSION AND IMPLICATIONS: Urolithin C is a glucose-dependent activator of insulin secretion acting by facilitating L-type Ca2+ channel opening and Ca2+ influx into pancreatic beta-cells. Our work paves the way for the design of polyphenol metabolite-inspired compounds aimed at ameliorating beta-cell function.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Glucosa/metabolismo , Taninos Hidrolizables/metabolismo , Insulina/metabolismo , Animales , Línea Celular , Islotes Pancreáticos/metabolismo , Masculino , Ratas , Ratas Wistar
8.
J Med Chem ; 62(2): 965-973, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30543423

RESUMEN

The ghrelin receptor or growth hormone secretagogue receptor (GHSR) is a G-protein-coupled receptor that controls growth hormone and insulin secretion, food intake, and reward-seeking behaviors. Liver-expressed antimicrobial peptide 2 (LEAP2) was recently described as an endogenous antagonist of GHSR. Here, we present a study aimed at delineating the structural determinants required for LEAP2 activity toward GHSR. We demonstrate that the entire sequence of LEAP2 is not necessary for its actions. Indeed, the N-terminal part alone confers receptor binding and activity to LEAP2. We found that both LEAP2 and its N-terminal part behave as inverse agonists of GHSR and as competitive antagonists of ghrelin-induced inositol phosphate production and calcium mobilization. Accordingly, the N-terminal region of LEAP2 is able to inhibit ghrelin-induced food intake in mice. These data demonstrate an unexpected pharmacological activity for LEAP2 that is likely to have an important role in the control of ghrelin response under normal and pathological conditions.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/química , Receptores de Ghrelina/agonistas , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Unión Competitiva , Agonismo Inverso de Drogas , Células HEK293 , Humanos , Fosfatos de Inositol/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Ratas , Receptores de Ghrelina/antagonistas & inhibidores , Receptores de Ghrelina/metabolismo
9.
PLoS One ; 11(8): e0160948, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27505161

RESUMEN

RACK1 is a scaffolding protein that contributes to the specificity and propagation of several signaling cascades including the cAMP pathway. As such, RACK1 participates in numerous cellular functions ranging from cell migration and morphology to gene transcription. To obtain further insights on the mechanisms whereby RACK1 regulates cAMP-dependent processes, we set out to identify new binding partners of RACK1 during activation of the cAMP signaling using a proteomics strategy. We identified ß-actin as a direct RACK1 binding partner and found that the association between ß-actin and RACK1 is increased in response to the activation of the cAMP pathway. Furthermore, we show that cAMP-dependent increase in BDNF expression requires filamentous actin. We further report that ß-actin associates with the BDNF promoter IV upon the activation of the cAMP pathway and present data to suggest that the association of ß-actin with BDNF promoter IV is RACK1-dependent. Taken together, our data suggest that ß-actin is a new RACK1 binding partner and that the RACK1 and ß-actin association participate in the cAMP-dependent regulation of BDNF transcription.


Asunto(s)
Actinas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Regiones Promotoras Genéticas/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Citoesqueleto de Actina/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Unión Proteica , Transporte de Proteínas , Receptores de Cinasa C Activada , Transcripción Genética
10.
Br J Pharmacol ; 173(8): 1314-28, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27027724

RESUMEN

BACKGROUND AND PURPOSE: Using an in-house bioinformatics programme, we identified and synthesized a novel nonapeptide, H-Pro-Pro-Thr-Thr-Thr-Lys-Phe-Ala-Ala-OH. Here, we have studied its biological activity, in vitro and in vivo, and have identified its target in the brain. EXPERIMENTAL APPROACH: The affinity of the peptide was characterized using purified whole brain and striatal membranes from guinea pigs and rats . Its effect on behaviour in rats following intra-striatal injection of the peptide was investigated. A photoaffinity UV cross-linking approach combined with subsequent affinity purification of the ligand covalently bound to its receptor allowed identification of its target. KEY RESULTS: The peptide bound with high affinity to a single class of binding sites, specifically localized in the striatum and substantia nigra of brains from guinea pigs and rats. When injected within the striatum of rats, the peptide stimulated in vitro and in vivo dopamine release and induced dopamine-like motor effects. We purified the target of the peptide, a ~151 kDa protein that was identified by MS/MS as angiotensin converting enzyme (ACE I). Therefore, we decided to name the peptide acein. CONCLUSION AND IMPLICATIONS: The synthetic nonapeptide acein interacted with high affinity with brain membrane-bound ACE. This interaction occurs at a different site from the active site involved in the well-known peptidase activity, without modifying the peptidase activity. Acein, in vitro and in vivo, significantly increased stimulated release of dopamine from the brain. These results suggest a more important role for brain ACE than initially suspected.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Dopamina/metabolismo , Oligopéptidos/farmacología , Peptidil-Dipeptidasa A/metabolismo , Animales , Encéfalo/enzimología , Dominio Catalítico/efectos de los fármacos , Biología Computacional , Cobayas , Masculino , Oligopéptidos/administración & dosificación , Oligopéptidos/síntesis química , Ratas , Ratas Sprague-Dawley
11.
J Neurochem ; 130(2): 172-84, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24666346

RESUMEN

The mammalian (or mechanistic) target of rapamycin (mTOR) complex 1 (mTORC1) is a serine and threonine kinase that regulates cell growth, survival, and proliferation. mTORC1 is a master controller of the translation of a subset of mRNAs. In the central nervous system mTORC1 plays a crucial role in mechanisms underlying learning and memory by controlling synaptic protein synthesis. Here, we review recent evidence suggesting that the mTORC1 signaling pathway promotes neuroadaptations following exposure to a diverse group of drugs of abuse including stimulants, cannabinoids, opiates, and alcohol. We further describe potential molecular mechanisms by which drug-induced mTORC1 activation may alter brain functions. Finally, we propose that mTORC1 is a focal point shared by drugs of abuse to mediate drug-related behaviors such as reward seeking and excessive drug intake, and offer future directions to decipher the contribution of the kinase to mechanisms underlying addiction. Recent studies suggesting that exposure to diverse classes of drugs of abuse as well as exposure to drug-associated memories lead to mTORC1 kinase activation in the limbic system. In turn, mTORC1 controls the onset and the maintenance of pathological neuroadaptions that underlie several features of drug addiction such as drug seeking and relapse. Therefore, we propose that targeting mTORC1 and its effectors is a promising strategy to treat drug disorders.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Drogas Ilícitas/farmacología , Complejos Multiproteicos/efectos de los fármacos , Complejos Multiproteicos/fisiología , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/fisiología , Animales , Autofagia/efectos de los fármacos , Conducta/efectos de los fármacos , Humanos , Inmunosupresores , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Neuronas/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Sirolimus , Trastornos Relacionados con Sustancias/genética , Trastornos Relacionados con Sustancias/fisiopatología , Trastornos Relacionados con Sustancias/psicología , Transmisión Sináptica/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
12.
Nat Neurosci ; 16(8): 1111-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23792945

RESUMEN

Relapse to alcohol abuse is an important clinical issue that is frequently caused by cue-induced drug craving. Therefore, disruption of the memory for the cue-alcohol association is expected to prevent relapse. It is increasingly accepted that memories become labile and erasable soon after their reactivation through retrieval during a memory reconsolidation process that depends on protein synthesis. Here we show that reconsolidation of alcohol-related memories triggered by the sensory properties of alcohol itself (odor and taste) activates mammalian target of rapamycin complex 1 (mTORC1) in select amygdalar and cortical regions in rats, resulting in increased levels of several synaptic proteins. Furthermore, systemic or central amygdalar inhibition of mTORC1 during reconsolidation disrupts alcohol-associated memories, leading to a long-lasting suppression of relapse. Our findings provide evidence that the mTORC1 pathway and its downstream substrates are crucial in alcohol-related memory reconsolidation and highlight this pathway as a therapeutic target to prevent relapse.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Anisomicina/farmacología , Consumo Excesivo de Bebidas Alcohólicas/psicología , Memoria/efectos de los fármacos , Complejos Multiproteicos/antagonistas & inhibidores , Corteza Prefrontal/fisiopatología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Amígdala del Cerebelo/efectos de los fármacos , Animales , Anisomicina/uso terapéutico , Consumo Excesivo de Bebidas Alcohólicas/prevención & control , Condicionamiento Operante/fisiología , Señales (Psicología) , Etanol/sangre , Etanol/química , Etanol/farmacología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Memoria/fisiología , Complejos Multiproteicos/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal , Odorantes , Fosforilación/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirolimus/uso terapéutico , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología , Serina-Treonina Quinasas TOR/fisiología , Gusto
13.
J Neurosci ; 32(45): 15849-58, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23136424

RESUMEN

Uncontrolled consumption of alcohol is a hallmark of alcohol abuse disorders; however, the central molecular mechanisms underlying excessive alcohol consumption are still unclear. Here, we report that the GTP binding protein, H-Ras in the nucleus accumbens (NAc) plays a key role in neuroadaptations that underlie excessive alcohol-drinking behaviors. Specifically, acute (15 min) systemic administration of alcohol (2.5 g/kg) leads to the activation of H-Ras in the NAc of mice, which is observed even 24 h later. Similarly, rat operant self-administration of alcohol (20%) also results in the activation of H-Ras in the NAc. Using the same procedures, we provide evidence suggesting that the exchange factor GRF1 is upstream of H-Ras activation by alcohol. Importantly, we show that infection of mice NAc with lentivirus expressing a short hairpin RNA that targets the H-Ras gene produces a significant reduction of voluntary consumption of 20% alcohol. In contrast, knockdown of H-Ras in the NAc of mice did not alter water, quinine, and saccharin intake. Furthermore, using two-bottle choice and operant self-administration procedures, we show that inhibiting H-Ras activity by intra-NAc infusion of the farnesyltransferase inhibitor, FTI-276, produced a robust decrease of rats' alcohol drinking; however, sucrose consumption was unaltered. Finally, intra-NAc infusion of FTI-276 also resulted in an attenuation of seeking for alcohol. Together, these results position H-Ras as a central molecular mediator of alcohol's actions within the mesolimbic system and put forward the potential value of the enzyme as a novel target to treat alcohol use disorders.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Etanol/farmacología , Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Consumo de Bebidas Alcohólicas/genética , Animales , Consumo Excesivo de Bebidas Alcohólicas/genética , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Farnesiltransferasa/antagonistas & inhibidores , Masculino , Metionina/análogos & derivados , Metionina/farmacología , Ratones , Ratones Transgénicos , Núcleo Accumbens/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas p21(ras)/genética , Ratas , Ratas Long-Evans , Sacarosa/farmacología , ras-GRF1/genética , ras-GRF1/metabolismo
14.
J Biol Chem ; 287(1): 322-336, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22069327

RESUMEN

RACK1 is a scaffolding protein that spatially and temporally regulates numerous signaling cascades. We previously found that activation of the cAMP signaling pathway induces the translocation of RACK1 to the nucleus. We further showed that nuclear RACK1 is required to promote the transcription of the brain-derived neurotrophic factor (BDNF). Here, we set out to elucidate the mechanism underlying cAMP-dependent RACK1 nuclear translocation and BDNF transcription. We identified the scaffolding protein 14-3-3ζ as a direct binding partner of RACK1. Moreover, we found that 14-3-3ζ was necessary for the cAMP-dependent translocation of RACK1 to the nucleus. We further observed that the disruption of RACK1/14-3-3ζ interaction with a peptide derived from the RACK1/14-3-3ζ binding site or shRNA-mediated 14-3-3ζ knockdown inhibited cAMP induction of BDNF transcription. Together, these data reveal that the function of nuclear RACK1 is mediated through its interaction with 14-3-3ζ. As RACK1 and 14-3-3ζ are two multifunctional scaffolding proteins that coordinate a wide variety of signaling events, their interaction is likely to regulate other essential cellular functions.


Asunto(s)
Proteínas 14-3-3/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al GTP/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Neuronas/citología , Neuronas/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Ratas , Receptores de Cinasa C Activada , Receptores de Superficie Celular/química , Reproducibilidad de los Resultados , Transducción de Señal
15.
Biol Psychiatry ; 70(6): 575-82, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21549353

RESUMEN

BACKGROUND: Neuroadaptations within the nucleus accumbens (NAc) have been implicated in molecular mechanisms underlying the development and/or maintenance of alcohol abuse disorders. We recently reported that the activation of mammalian target of rapamycin complex 1 (mTORC1) signaling pathway in the NAc of rodents, after exposure to alcohol, contributes to alcohol drinking behaviors. The kinase AKT is a main upstream activator of the mTORC1 pathway. We therefore hypothesized that the activation of AKT in the NAc in response to alcohol exposure plays an important role in mechanisms that underlie excessive alcohol consumption. METHODS: Western blot analysis was used to assess the phosphorylation levels of enzymes. Acute exposure of mice to alcohol was achieved by the administration of 2 g/kg alcohol intraperitoneally (i.p.). Two-bottle choice and operant self-administration procedures were used to assess drinking behaviors in rats. RESULTS: We found that acute systemic administration of alcohol and recurring cycles of excessive voluntary consumption of alcohol and withdrawal result in the activation of AKT signaling in the NAc of rodents. We show that inhibition of AKT or its upstream activator, phosphatidylinositol-3-kinase (PI3K), within the NAc of rats attenuates binge drinking as well as alcohol but not sucrose operant self-administration. CONCLUSIONS: Our results suggest that the activation of the AKT pathway in the NAc in response to alcohol exposure is an important contributor to the molecular mechanisms underlying alcohol-drinking behaviors. AKT signaling pathway inhibitors are therefore potential candidates for drug development for the treatment of alcohol use and abuse disorders.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Conducta Adictiva/metabolismo , Conducta Adictiva/fisiopatología , Condicionamiento Operante/efectos de los fármacos , Núcleo Accumbens/fisiopatología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Consumo de Bebidas Alcohólicas/fisiopatología , Androstadienos/administración & dosificación , Androstadienos/farmacología , Animales , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Condicionamiento Operante/fisiología , Etanol/administración & dosificación , Etanol/antagonistas & inhibidores , Etanol/envenenamiento , Masculino , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Núcleo Accumbens/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Long-Evans , Ribonucleósidos/administración & dosificación , Ribonucleósidos/farmacología , Autoadministración/métodos , Autoadministración/psicología , Transducción de Señal/efectos de los fármacos , Sacarosa/administración & dosificación , Wortmanina
16.
Proc Natl Acad Sci U S A ; 107(46): 20093-8, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21041654

RESUMEN

Alcohol addiction is a chronically relapsing disorder that includes certain maladaptive learning and memory. The serine and threonine kinase complex, mammalian target of rapamycin complex 1 (mTORC1), has been implicated in synaptic plasticity, learning, and memory by controlling protein translation. Here we show that administration of alcohol and excessive voluntary consumption of alcohol induce the activation of the mTORC1-mediated signaling pathway in the nucleus accumbens (NAc) of rodents. We further show that the protein expression levels of GluR1 and Homer, two synaptic proteins whose translation has been shown to be modulated by mTORC1, are up-regulated in the NAc of rodents with a history of excessive alcohol consumption. In addition, our results document that the Food and Drug Administration-approved inhibitor of mTORC1, rapamycin, decreases expression of alcohol-induced locomotor sensitization and place preference, as well as excessive alcohol intake and seeking in preclinical rodent models of alcohol abuse. Together, our results suggest that mTORC1 within the NAc is a contributor to molecular mechanisms underlying alcohol-drinking behaviors. Furthermore, despite its massive health and socioeconomic impact worldwide, pharmacotherapies for alcohol abuse and addiction remain limited. Our data therefore put forward the possibility that targeting the mTORC1 signaling cascade is an innovative and valuable strategy for the treatment of alcohol use and abuse disorders.


Asunto(s)
Adaptación Fisiológica , Trastornos Relacionados con Alcohol/fisiopatología , Sistema Nervioso/fisiopatología , Proteínas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Adaptación Fisiológica/efectos de los fármacos , Alcoholes/administración & dosificación , Alcoholes/farmacología , Animales , Proteínas Portadoras/metabolismo , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Proteínas de Andamiaje Homer , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Actividad Motora/efectos de los fármacos , Complejos Multiproteicos , Sistema Nervioso/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ratas , Receptores AMPA/metabolismo , Autoadministración , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/farmacología , Sacarosa/administración & dosificación , Sacarosa/farmacología , Serina-Treonina Quinasas TOR , Regulación hacia Arriba/efectos de los fármacos
17.
J Biol Chem ; 285(25): 19043-50, 2010 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-20410295

RESUMEN

Scaffolding proteins are major contributors to the spatial and temporal orchestration of signaling cascades and hence cellular functions. RACK1 is a scaffolding protein that plays an important role in the regulation of, and cross-talk between, various signaling pathways. Here we report that RACK1 is a mediator of chromatin remodeling, resulting in an exon-specific expression of the brain-derived neurotrophic factor (BDNF) gene. Specifically, we found that following the activation of the cAMP pathway, nuclear RACK1 localizes at the promoter IV region of the BDNF gene by its association with histones H3 and H4, leading to the dissociation of the transcription repressor methyl-CpG-binding protein 2 (MeCP2) from the promoter, resulting in the acetylation of histone H4. These chromatin modifications lead to the activation of the promoter and to the subsequent promoter-controlled transcription of BDNF exon IV. Our findings expand our knowledge regarding the function of scaffolding proteins such as RACK1. Furthermore, this novel mechanism for the regulation of exon-specific expression of the BDNF gene by RACK1 could have implications on the neuronal functions of the growth factor including synaptic plasticity, learning, and memory.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Epigénesis Genética , Proteínas de Unión al GTP/biosíntesis , Proteínas de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Islas de CpG , Histonas/química , Humanos , Aprendizaje , Memoria , Proteína 2 de Unión a Metil-CpG/genética , Ratas , Ratas Sprague-Dawley , Receptores de Cinasa C Activada , Transducción de Señal
18.
Biochem Biophys Res Commun ; 373(1): 80-4, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18544342

RESUMEN

Mu opioid (MOP) receptor activation can be functionally modulated by stimulation of Neuropeptide FF 2 (NPFF(2)) G protein-coupled receptors. Fluorescence recovery after photobleaching experiments have shown that activation of the NPFF(2) receptor dramatically reduces the fraction of MOP receptors confined in microdomains of the plasma membrane of SH-SY5Y neuroblastoma cells. The aim of the present work was to assess if the direct observation of receptor compartmentation by fluorescence techniques in living cells could be related to indirect estimation of receptor partitioning in lipid rafts after biochemical fractionation of the cell. Our results show that MOP receptor distribution in lipid rafts is highly dependent upon the method of purification, questioning the interpretation of previous data regarding MOP receptor compartmentation. Moreover, the NPFF analogue 1DMe does not modify the distribution profile of MOP receptors, clearly demonstrating that membrane fractionation data do not correlate with direct measurement of receptor compartmentation in living cells.


Asunto(s)
Microdominios de Membrana/metabolismo , Receptores de Neuropéptido/metabolismo , Receptores Opioides mu/metabolismo , Fraccionamiento Celular , Línea Celular , Detergentes/química , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Oligopéptidos/farmacología , Receptores de Neuropéptido/agonistas , Receptores de Neuropéptido/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
19.
Proteome Sci ; 4: 23, 2006 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17184524

RESUMEN

BACKGROUND: Opiate addiction reflects plastic changes that endurably alter synaptic transmission within relevant neuronal circuits. The biochemical mechanisms of these adaptations remain largely unknown and proteomics-based approaches could lead to a broad characterization of the molecular events underlying adaptations to chronic drug exposure. RESULTS: Thus, we have started proteomic analyses of the effects of chronic morphine exposure in a recombinant human neuroblastoma SH-SY5Y clone that stably overexpresses the mu-opioid receptor. Cells were treated with morphine for 6, 24 and 72 hours, the proteins were separated by 2-D gel electrophoresis and stained with Coomassie blue, and the protein map was compared with that obtained from untreated cells. Spots showing a statistically significant variation were selected for identification using mass spectrometric analyses. CONCLUSION: A total of 45 proteins were identified, including proteins involved in cellular metabolism, cytoskeleton organization, vesicular trafficking, transcriptional and translational regulation, and cell signaling.

20.
Mol Pharmacol ; 68(2): 467-76, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15901846

RESUMEN

The initial aim of this study was to identify protein changes associated with long-term morphine treatment in a recombinant human neuroblastoma SH-SY5Y clone (sc2) stably overexpressing the human mu-opioid (MOP) receptor. In MOP receptor-overexpressing sc2 cells, short-term morphine exposure was found to be much more potent and efficacious in inhibiting forskolin-elicited production of cAMP, and long-term morphine exposure was shown to induce a substantially higher degree of opiate dependence, as reflected by adenylate cyclase sensitization, than it did in wild-type neuroblastoma cells. Differential proteomic analysis of detergent-resistant membrane rafts isolated from untreated and chronically morphine-treated sc2 cells revealed long-term morphine exposure to have reliably induced a 30 to 40% decrease in the abundance of five proteins, subsequently identified by mass spectrometry as G protein subunits alphai(2), alphai(3), beta(1), and beta(2), and prohibitin. Quantitative Western blot analyses of whole-cell extracts showed that long-term morphine treatment-induced down-regulation of Gbeta but not of the other proteins is highly correlated (r(2) = 0.96) with sensitization of adenylate cyclase. Down-regulation of Gbeta and adenylate cyclase sensitization elicited by long-term morphine treatment were suppressed in the presence of carbobenzoxy-l-leucyl-l-leucyl-l-norvalinal (MG-115) or lactacystin. Thus, sustained activation of the MOP receptor by morphine in sc2 cells seems to promote proteasomal degradation of Gbeta to sensitize adenylate cyclase. Together, our data suggest that the long-term administration of opiates may elicit dependence by altering the neuronal balance of heterotrimeric G proteins and adenylate cyclases, with the ubiquitin-proteasome pathway playing a pivotal role.


Asunto(s)
Adenilil Ciclasas/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Morfina/administración & dosificación , Neuroblastoma/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Adenilil Ciclasas/genética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Subunidades beta de la Proteína de Unión al GTP/genética , Humanos , Neuroblastoma/genética , Complejo de la Endopetidasa Proteasomal/genética , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...