Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Bioorg Chem ; 128: 105905, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35710525

RESUMEN

We identified, via high-throughput screening using a FLIPR® calcium assay, compound 1, which incorporated a dihydroquinolinyl-2-oxoethylsulfanyl-(1H,5H)-pyrimidinedione core and activated the µ-opioid receptor (MOR) in the presence of naloxone or naltrexone. A structure-activity relationship study of the analogs of 1 led to the design of compound 21, which activated MOR in the presence of naloxone with an EC50 of 3.3 ± 0.2 µM. MOR activation by the compound 21-antagonist pair was antagonist-dependent. Compound 21 did not affect the potency of the orthosteric agonist, morphine, toward MOR, indicating that it affected the function of MOR antagonists rather than that of the agonists. Computer modeling of the compound 21-MOR-naloxone complex revealed major interactions between compound 21 and MOR, including hydrogen bonding with Ser196, π-π stacking with Tyr149, and sulfur-aromatic interaction with Trp192. This study may pave the way for developing agents capable of safe and effective MOR modulation.


Asunto(s)
Naloxona , Naltrexona , Analgésicos Opioides , Imidazoles , Naloxona/farmacología , Naltrexona/farmacología , Receptores Opioides , Sulfonamidas , Tiofenos
2.
Anesthesiology ; 126(5): 952-966, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28212204

RESUMEN

BACKGROUND: The authors investigated the pharmacology and signaling pathways of the opioid receptors modulated by compound 1, 1-(2,4-dibromophenyl)-3,6,6-trimethyl-1,5,6,7-tetrahydro-4H-indazol-4-one. METHODS: In vitro studies of compound 1 were assessed by using a radioligand-binding assay (n = 3), a cyclic adenosine monophosphate assay (n = 3), a ß-arrestin assay (n = 3), an internalization assay (n = 3), and an immunohistochemistry (n = 8). In vivo studies of compound 1 were characterized using a tail-flick test (n = 5 to 6), tail-clip test (n = 7), von Frey hair test (n = 5), and charcoal meal test (n = 5). RESULTS: Compound 1 elicited robust effects in µ-opioid (mean ± SD; binding affinity: 15 ± 2 nM; cyclic adenosine monophosphate assay: 24 ± 6 nM), δ-opioid (82 ± 7 nM; 1.9 ± 0.1 µM), and κ-opioid (76 ± 9 nM; 1.4 ± 0.5 µM) receptor-expressing cells. Compound 1 acts as a full agonist of ß-arrestin-2 recruitment in µ-opioid (1.1 ± 0.3 µM) and δ-opioid (9.7 ± 1.9 µM) receptor-expressing cells. Compound 1 caused less gastrointestinal dysfunction (charcoal meal test: morphine: 82 ± 5%; compound 1: 42 ± 5%) as well as better antinociception in mechanical pain hypersensitivity (tail-clip test: morphine: 10 ± 3 s; compound 1: 19 ± 1 s) and in cancer-induced pain (von Frey hair test: morphine: 0.1 ± 0.1 g; compound 1: 0.3 ± 0.1 g) than morphine at equi-antinociceptive doses. CONCLUSIONS: Compound 1 produced antinociception with less gastrointestinal dysfunction than morphine.


Asunto(s)
Enfermedades Gastrointestinales/inducido químicamente , Indazoles/farmacología , Morfina , Receptores Opioides/agonistas , Analgésicos Opioides/farmacología , Animales , Modelos Animales de Enfermedad , Enfermedades Gastrointestinales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Nucleic Acids Res ; 42(21): 13012-25, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25361975

RESUMEN

Heterogeneous nuclear ribonucleoprotein K (hnRNP K) binds to the promoter region of mu-opioid receptor (MOR) to regulate its transcriptional activity. How hnRNP K contributes to the analgesic effects of morphine, however, is largely unknown. We provide evidence that morphine increases hnRNP K protein expression via MOR activation in rat primary cortical neurons and HEK-293 cells expressing MORs, without increasing mRNA levels. Using the bicistronic reporter assay, we examined whether morphine-mediated accumulation of hnRNP K resulted from translational control. We identified potential internal ribosome entry site elements located in the 5' untranslated regions of hnRNP K transcripts that were regulated by morphine. This finding suggests that internal translation contributes to the morphine-induced accumulation of hnRNP K protein in regions of the central nervous system correlated with nociceptive and antinociceptive modulatory systems in mice. Finally, we found that down-regulation of hnRNP K mediated by siRNA attenuated morphine-induced hyperpolarization of membrane potential in AtT20 cells. Silencing hnRNP K expression in the spinal cord increased nociceptive sensitivity in wild-type mice, but not in MOR-knockout mice. Thus, our findings identify the role of translational control of hnRNP K in morphine-induced analgesia through activation of MOR.


Asunto(s)
Regiones no Traducidas 5'/efectos de los fármacos , Analgésicos Opioides/farmacología , Ribonucleoproteína Heterogénea-Nuclear Grupo K/biosíntesis , Morfina/farmacología , Neuronas/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Receptores Opioides mu/metabolismo , Animales , Secuencia de Bases , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Secuencia Conservada , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Ratones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Nocicepción , Ratas , Ribosomas/metabolismo , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Regulación hacia Arriba
4.
Bioorg Med Chem ; 22(17): 4694-703, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25087049

RESUMEN

The µ-opioid receptor (MOR) is the major opioid receptor targeted by most analgesics in clinical use. However, the use of all known MOR agonists is associated with severe adverse effects. We reported that the 1-phenyl-3,6,6-trimethyl-1,5,6,7-tetrahydro-4H-indazol-4-ones are novel opioid receptor agonists. Subsequent structural modification resulted in the potent MOR/KOR (κ-opioid receptor) agonists 19, 20, and 21. Testing the analgesic effect of these in WT B6 mice (tail-flick test) gave ED50 values of 8.4, 10.9, and 26.6mg/kg, respectively. The 1-phenyl-3,6,6-trimethyl-1,5,6,7-tetrahydro-4H-indazol-4-one core could be addressed in 1 or 2 synthetic steps with moderate to high percent of yield. In the adenylyl cyclase assay, compound 19 displayed a MOR/KOR agonist profile, with IC50 values of 0.73 and 0.41µM, respectively. Current results suggest that compound 19 is a promising lead to go further development and in vitro/in vivo adverse effects studies.


Asunto(s)
Analgésicos/farmacología , Descubrimiento de Drogas , Indazoles/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Analgésicos/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Indazoles/síntesis química , Indazoles/química , Ratones , Ratones Congénicos , Estructura Molecular , Dolor/tratamiento farmacológico , Dimensión del Dolor , Relación Estructura-Actividad , Cola (estructura animal)/efectos de los fármacos
5.
Cell Chem Biol ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39025070

RESUMEN

Morphinan antagonists, which block opioid effects at mu-opioid receptors, have been studied for their analgesic potential. Previous studies have suggested that these antagonists elicit analgesia with fewer adverse effects in the presence of the mutant mu-opioid receptor (MOR; S196A). However, introducing a mutant receptor for medical applications represents significant challenges. We hypothesize that binding a chemical compound to the MOR may elicit a comparable effect to the S196A mutation. Through high-throughput screening and structure-activity relationship studies, we identified a modulator, 4-(2-(4-fluorophenyl)-4-oxothiazolidin-3-yl)-3-methylbenzoic acid (BPRMU191), which confers agonistic properties to small-molecule morphinan antagonists, which induce G protein-dependent MOR activation. Co-application of BPRMU191 and morphinan antagonists resulted in MOR-dependent analgesia with diminished side effects, including gastrointestinal dysfunction, antinociceptive tolerance, and physical and psychological dependence. Combining BPRMU191 and morphinan antagonists could serve as a potential therapeutic strategy for severe pain with reduced adverse effects and provide an avenue for studying G protein-coupled receptor modulation.

6.
Neurobiol Learn Mem ; 93(3): 372-82, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19995614

RESUMEN

In many instances, increase in neuronal activity can induce biphasic secretion of a modulator. The initial release of the modulator triggers the induction of synaptic plasticity, whereas the second-phase release reinforces the efficacy of synaptic transmission and growth of dendrites and axons. In this study, we showed that fear conditioning not only induced the first but also a second peak of brain-derived neurotrophic factor (BDNF) expression. Fluorescent immunohistostaining confirmed that BDNF expression increased at 1 and 12 h after conditioning and returned to baseline at 30 h after conditioning. Mature BDNF expression increased in a similar manner. TrkB-IgG or K252a infusion before training impaired fear memory on days 1 and 7 after training. In contrast, TrkB-IgG or K252a infusion 9 h after fear conditioning did not affect memory retention on day 1 after training but impaired fear memory on day 7 after training. Fear conditioning significantly enhanced Zif268 expression in the amygdala at 12 h after training; this enhanced expression was completely inhibited by TrkB-IgG infusion 9 h after training. The level of growth-associated protein 43 (GAP-43), a marker of newly formed synapses, in the amygdala increased 7 days after fear conditioning. Moreover, conditioned rats had higher AMPA/NMDA ratio than unpaired rats. These results suggest that consolidated memory could be continuously modulated by previous molecular changes produced during memory acquisition.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Miedo , Expresión Génica/genética , Memoria/fisiología , Estimulación Acústica , Animales , Carbazoles/metabolismo , Técnica del Anticuerpo Fluorescente , Hipocampo/metabolismo , Alcaloides Indólicos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptor trkB/genética , Receptor trkB/metabolismo , Reflejo de Sobresalto , Transducción de Señal/fisiología , Factores de Tiempo
7.
Neuropharmacology ; 166: 107678, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31278929

RESUMEN

There is unmet need to design an analgesic with fewer side effects for severe pain management. Although traditional opioids are the most effective painkillers, they are accompanied by severe adverse responses, such as respiratory depression, constipation symptoms, tolerance, withdrawal, and addiction. We indicated BPR1M97 as a dual mu opioid receptor (MOP)/nociceptin-orphanin FQ peptide (NOP) receptor full agonist and investigated the pharmacology of BPR1M97 in multiple animal models. In vitro studies on BPR1M97 were assessed using cyclic-adenosine monophosphate production, ß-arrestin, internalization, and membrane potential assays. In vivo studies were characterized using the tail-flick, tail-clip, lung functional, heart functional, acetone drop, von Frey hair, charcoal meal, glass bead, locomotor activity, conditioned place preference (CPP) and naloxone precipitation tests. BPR1M97 elicited full agonist properties for all cell-based assays tested in MOP-expressing cells. However, it acted as a G protein-biased agonist for NOP. BPR1M97 initiated faster antinociceptive effects at 10 min after subcutaneous injection and elicited better analgesia in cancer-induced pain than morphine. Unlike morphine, BPR1M97 caused less respiratory, cardiovascular, and gastrointestinal dysfunction. In addition, BPR1M97 decreased global activity and induced less withdrawal jumping precipitated by naloxone. Thus, BPR1M97 could serve as a novel small molecule dual receptor agonist for antinociception with fewer side effects than morphine. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Analgésicos/uso terapéutico , Morfina/uso terapéutico , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Animales , Células CHO , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/patología , Cricetulus , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Dimensión del Dolor/métodos , Resultado del Tratamiento , Receptor de Nociceptina
8.
Sci Rep ; 9(1): 2405, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30787373

RESUMEN

Morphine is a unique opioid analgesic that activates the mu-opioid receptor (MOR) without efficiently promoting its endocytosis that may underlie side effects. Our objective was to discover a novel enhancer of ligand-induced MOR endocytosis and determine its effects on analgesia, tolerance and dependence. We used high-throughput screening to identify convallatoxin as an enhancer of ligand-induced MOR endocytosis with high potency and efficacy. Treatment of cells with convallatoxin enhanced morphine-induced MOR endocytosis through an adaptor protein 2 (AP2)/clathrin-dependent mechanism, attenuated morphine-induced phosphorylation of MOR, and diminished desensitization of membrane hyperpolarization. Furthermore, co-treatment with chronic convallatoxin reduced morphine tolerance in animal models of acute thermal pain and chronic inflammatory pain. Acute convallatoxin administration reversed morphine tolerance and dependence in morphine-tolerant mice. These findings suggest convallatoxin are potentially therapeutic for morphine side effects and open a new avenue to study MOR trafficking.


Asunto(s)
Analgésicos/farmacología , Morfina/farmacología , Receptores Opioides mu/genética , Estrofantinas/farmacología , Analgesia/métodos , Analgésicos/química , Animales , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Humanos , Ligandos , Ratones , Receptores Opioides mu/efectos de los fármacos
9.
Eur J Med Chem ; 167: 312-323, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30776693

RESUMEN

Morphine is widely used for the treatment of severe pain. This analgesic effect is mediated principally by the activation of µ-opioid receptors (MOR). However, prolonged activation of MOR also results in tolerance, dependence, addiction, constipation, nausea, sedation, and respiratory depression. To address this problem, we sought alternative ways to activate MOR - either by use of novel ligands, or via a novel activation mechanism. To this end, a series of compounds were screened using a sensitive CHO-K1/MOR/Gα15 cell-based FLIPR® calcium high-throughput screening (HTS) assay, and the bithiazole compound 5a was identified as being able activate MOR in combination with naloxone. Structural modifications of 5a resulted in the discovery of lead compound 5j, which could effectively activate MOR in combination with the MOR antagonist naloxone or naltrexone. In vivo, naloxone in combination with 100 mg/kg of compound 5j elicited antinociception in a mouse tail-flick model with an ED50 of 17.5 ±â€¯4 mg/kg. These results strongly suggest that the mechanism by which the 5j/naloxone combination activates MOR is worthy of further study, as its discovery has the potential to yield an entirely novel class of analgesics.


Asunto(s)
Analgésicos/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/uso terapéutico , Receptores Opioides mu/agonistas , Tiazoles/farmacología , Aminas , Animales , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Muridae , Antagonistas de Narcóticos/farmacología , Relación Estructura-Actividad
10.
Eur J Med Chem ; 126: 202-217, 2017 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-27776274

RESUMEN

µ-Opioid receptor (MOR) agonists are analgesics used clinically for the treatment of moderate to severe pain, but their use is associated with severe adverse effects such as respiratory depression, constipation, tolerance, dependence, and rewarding effects. In this study, we identified N-({2-[(4-bromo-2-trifluoromethoxyphenyl)sulfonyl]-1,2,3,4-tetrahydro-1-isoquinolinyl}methyl)cyclohexanecarboxamide (1) as a novel opioid receptor agonist by high-throughput screening. Structural modifications made to 1 to improve potency and blood-brain-barrier (BBB) penetration resulted in compounds 45 and 46. Compound 45 was a potent MOR/KOR (κ-opioid receptor) agonist, and compound 46 was a potent MOR and medium KOR agonist. Both 45 and 46 demonstrated a significant anti-nociceptive effect in a tail-flick test performed in wild type (WT) B6 mice. The ED50 value of 46 was 1.059 mg/kg, and the brain concentrations of 45 and 46 were 7424 and 11696 ng/g, respectively. Accordingly, compounds 45 and 46 are proposed for lead optimization and in vivo disease-related pain studies.


Asunto(s)
Analgésicos/química , Analgésicos/farmacología , Benzamidas/química , Benzamidas/farmacología , Receptores Opioides mu/metabolismo , Adenilil Ciclasas/metabolismo , Analgésicos/síntesis química , Analgésicos/metabolismo , Animales , Benzamidas/síntesis química , Benzamidas/metabolismo , Barrera Hematoencefálica/metabolismo , Línea Celular , Evaluación Preclínica de Medicamentos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Masculino , Ratones , Simulación de Dinámica Molecular , Conformación Proteica , Receptores Opioides mu/química , Relación Estructura-Actividad
11.
Neuropsychopharmacology ; 31(2): 287-96, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16034442

RESUMEN

This study is designed to characterize the signal cascades by which brain-derived neurotrophic factor (BDNF) modulates long-term memory of fear conditioning. Enzyme-linked immunosorbent assay (ELISA) and Western blot analysis of tissue homogenates taken from fear-conditioned rats showed an increase in the amygdala of BDNF protein levels and its receptor TrkB phosphorylation. Bilateral administration of a TrkB ligand scavenger TrkB IgG and a Trk-specific tyrosine kinase inhibitor K252a to the amygdala impaired fear memory, as measured with fear-potentiated startle. Fear conditioning resulted in the association of Shc and TrkB, Shc and Ras, the increase in active Ras and phosphorylation of mitogen-activated protein kinase (MAPK). Treatment of amygdala slices with BDNF for 15 min increased the levels of active Ras, and MAPK and Akt phosphorylation. BDNF-induced MAPK phosphorylation was completely abolished by MEK inhibitors, and was partially inhibited by farnesyltransferase or phosphatidylinositol-3 kinase (PI-3 kinase) inhibitors. On the other hand, BDNF-induced Akt phosphorylation was unaffected by farnesyltransferase or MEK inhibitors, but could be blocked by PI-3 kinase inhibitors. Together, these data suggest a requirement of BDNF for fear learning. The memory-enhancing effect of BDNF involves the activation of MAPK and PI-3 kinase. BDNF-induced MAPK phosphorylation in the amygdala is mediated via TrkB and the Shc-binding site. Shc binding to TrkB leads to activation of Ras, Raf, and MEK. In addition, BDNF could induce phosphorylation of MAPK via activation of PI-3 kinase.


Asunto(s)
Amígdala del Cerebelo/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Condicionamiento Psicológico/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Animales , Anticuerpos/farmacología , Conducta Animal , Western Blotting/métodos , Factor Neurotrófico Derivado del Encéfalo/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Electrochoque/métodos , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Miedo , Expresión Génica/efectos de los fármacos , Inmunoprecipitación/métodos , Técnicas In Vitro , Masculino , Proteínas de Unión al GTP Monoméricas/farmacología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Factores de Tiempo
12.
Brain Pathol ; 21(3): 249-62, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21029239

RESUMEN

The expression of hypoxia-inducible factor-1-alpha (HIF-1α) is upregulated in ischemic stroke, but its function is still unclear. In the present study, biphasic expression of HIF-1α was observed during 1-12 h and after 48 h in neurons exposed to ischemic stress in vitro and in vivo. Treating neurons with 2-methoxyestradiol (2ME2) 0.5 h after ischemic stress or pre-silencing HIF-1α with small interfering RNA (siRNA) decreased brain injury, brain edema and number of apoptotic cell, and downregulates Nip-like protein X (Nix) expression. Conversely, applying 2ME2 to neurons 8 h after ischemic stress or silencing the HIF-1α with siRNA 12 h after oxygen-glucose deprivation (OGD) increased neuron damage and decreased vascular endothelial growth factor (VEGF) expression. Taken together, these results demonstrate that HIF-1α induced by ischemia in early and late times leads cellular apoptosis and survival, respectively, and provides a new insight into the divergent roles of HIF-1α expression in neurons after ischemic stroke.


Asunto(s)
Apoptosis/fisiología , Isquemia Encefálica/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , 2-Metoxiestradiol , Animales , Antimetabolitos/farmacología , Isquemia Encefálica/complicaciones , Supervivencia Celular/fisiología , Células Cultivadas , Estradiol/análogos & derivados , Estradiol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Silenciador del Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Neuronas/patología , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/patología , Factores de Tiempo
13.
Mol Pharmacol ; 72(2): 350-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17456785

RESUMEN

We have demonstrated previously that brain-derived neurotrophic factor (BDNF) signaling in the amygdala is required for the consolidation of fear memory. This study is designed to characterize the signal cascades by which fear conditioning modulates transcriptional and translational expression of BDNF. Real-time reverse transcription-coupled polymerase chain reaction showed a significant increase in BDNF exon I- and III-containing mRNA in the amygdala of fear-conditioned rats, indicating that fear conditioning was capable of up-regulating BDNF mRNA. Bilateral administration of actinomycin D or anisomycin to the amygdala attenuated conditioning-induced increase in BDNF protein. Inhibitors for N-methyl-d-aspartate (NMDA) receptor, L-type voltage-dependent calcium channel (L-VDCC), adenylyl cyclase, cAMP-dependent protein kinase (PKA), and calcium/calmodulin-dependent kinase IV (CaMKIV) significantly reduced the increase. Moreover, DNA affinity precipitation and chromatin immunoprecipitation assays showed that phosphorylated cAMP response element-binding protein (p-CREB) binding activity in the proximal region of BDNF promoter I and III was significantly increased after fear conditioning. Intra-amygdala administration of cAMP response element decoy DNA before training impaired fear learning. Taken together, these results suggest that calcium influx through NMDA receptors and L-VDCCs during fear conditioning activates PKA and CaMKIV resulting in CREB phosphorylation. The phosphorylated CREB binds to BDNF promoter and up-regulates the expression of BDNF in the amygdala, which helps the consolidation of fear memory.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Miedo , Memoria , Transcripción Genética , Animales , Canales de Calcio Tipo L/fisiología , Condicionamiento Psicológico , Masculino , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA