Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Nature ; 621(7977): 206-214, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37648856

RESUMEN

Transient receptor potential (TRP) channels are a large, eukaryotic ion channel superfamily that control diverse physiological functions, and therefore are attractive drug targets1-5. More than 210 structures from more than 20 different TRP channels have been determined, and all are tetramers4. Despite this wealth of structures, many aspects concerning TRPV channels remain poorly understood, including the pore-dilation phenomenon, whereby prolonged activation leads to increased conductance, permeability to large ions and loss of rectification6,7. Here, we used high-speed atomic force microscopy (HS-AFM) to analyse membrane-embedded TRPV3 at the single-molecule level and discovered a pentameric state. HS-AFM dynamic imaging revealed transience and reversibility of the pentamer in dynamic equilibrium with the canonical tetramer through membrane diffusive protomer exchange. The pentamer population increased upon diphenylboronic anhydride (DPBA) addition, an agonist that has been shown to induce TRPV3 pore dilation. On the basis of these findings, we designed a protein production and data analysis pipeline that resulted in a cryogenic-electron microscopy structure of the TRPV3 pentamer, showing an enlarged pore compared to the tetramer. The slow kinetics to enter and exit the pentameric state, the increased pentamer formation upon DPBA addition and the enlarged pore indicate that the pentamer represents the structural correlate of pore dilation. We thus show membrane diffusive protomer exchange as an additional mechanism for structural changes and conformational variability. Overall, we provide structural evidence for a non-canonical pentameric TRP-channel assembly, laying the foundation for new directions in TRP channel research.


Asunto(s)
Multimerización de Proteína , Canales Catiónicos TRPV , Anhídridos/química , Anhídridos/farmacología , Análisis de Datos , Difusión , Subunidades de Proteína/química , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/ultraestructura , Microscopía de Fuerza Atómica , Terapia Molecular Dirigida , Microscopía por Crioelectrón , Estructura Cuaternaria de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos
2.
Nature ; 622(7981): 195-201, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37730991

RESUMEN

Type A γ-aminobutyric acid receptors (GABAARs) are the principal inhibitory receptors in the brain and the target of a wide range of clinical agents, including anaesthetics, sedatives, hypnotics and antidepressants1-3. However, our understanding of GABAAR pharmacology has been hindered by the vast number of pentameric assemblies that can be derived from 19 different subunits4 and the lack of structural knowledge of clinically relevant receptors. Here, we isolate native murine GABAAR assemblies containing the widely expressed α1 subunit and elucidate their structures in complex with drugs used to treat insomnia (zolpidem (ZOL) and flurazepam) and postpartum depression (the neurosteroid allopregnanolone (APG)). Using cryo-electron microscopy (cryo-EM) analysis and single-molecule photobleaching experiments, we uncover three major structural populations in the brain: the canonical α1ß2γ2 receptor containing two α1 subunits, and two assemblies containing one α1 and either an α2 or α3 subunit, in which the single α1-containing receptors feature a more compact arrangement between the transmembrane and extracellular domains. Interestingly, APG is bound at the transmembrane α/ß subunit interface, even when not added to the sample, revealing an important role for endogenous neurosteroids in modulating native GABAARs. Together with structurally engaged lipids, neurosteroids produce global conformational changes throughout the receptor that modify the ion channel pore and the binding sites for GABA and insomnia medications. Our data reveal the major α1-containing GABAAR assemblies, bound with endogenous neurosteroid, thus defining a structural landscape from which subtype-specific drugs can be developed.


Asunto(s)
Microscopía por Crioelectrón , Neuroesteroides , Receptores de GABA-A , Ácido gamma-Aminobutírico , Animales , Ratones , Sitios de Unión/efectos de los fármacos , Depresión Posparto/tratamiento farmacológico , Flurazepam/farmacología , Ácido gamma-Aminobutírico/metabolismo , Hipnóticos y Sedantes/farmacología , Activación del Canal Iónico/efectos de los fármacos , Neuroesteroides/metabolismo , Neuroesteroides/farmacología , Fotoblanqueo , Pregnanolona/farmacología , Conformación Proteica/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Receptores de GABA-A/ultraestructura , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Zolpidem/farmacología
3.
Cancer Sci ; 112(10): 4013-4025, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34252226

RESUMEN

Although the role of bromodomain-containing protein 4 (BRD4) in ovarian cancer, pancreatic cancer, lymphoma, and many other diseases is well known, its function in cutaneous melanoma is only partially understood. The results of the present study show that the BRD4 inhibitor JQ1 promotes the apoptosis of B16 melanoma cells by altering mitochondrial dynamics, thereby inducing mitochondrial dysfunction and increasing oxidative stress. We found that treatment of B16 cells with different concentrations of JQ1 (125 nmol/L or 250 nmol/L) significantly downregulated the expression of protein subunits involved in mitochondrial respiratory chain complexes I, III, IV, and V, increased reactive oxygen species, induced energy metabolism dysfunction, significantly enhanced apoptosis, and activated the mitochondrial apoptosis pathway. At the same time, JQ1 inhibited the activation of AMP-activated protein kinase, a metabolic energy sensor. In addition, we found that the mRNA and protein levels of mitochondrial dynamin-related protein 1 increased, whereas the levels of mitochondrial fusion protein 1 and optic atrophy protein 1 decreased. Mechanistically, we determined that JQ1 inhibited the expression of c-Myc and altered mitochondrial dynamics, eventually leading to changes in the mitochondrial function, metabolism, and apoptosis of B16 melanoma cells.


Asunto(s)
Apoptosis/fisiología , Azepinas/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Melanoma/metabolismo , Mitocondrias/efectos de los fármacos , Neoplasias Cutáneas/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Respiración de la Célula/efectos de los fármacos , Dinaminas/efectos de los fármacos , Dinaminas/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Metabolismo Energético/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Proteína-1 Reguladora de Fusión/metabolismo , Humanos , Melanoma/patología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/patología , Factores de Transcripción/metabolismo
4.
Nature ; 526(7572): 224-9, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26344198

RESUMEN

The strychnine-sensitive glycine receptor (GlyR) mediates inhibitory synaptic transmission in the spinal cord and brainstem and is linked to neurological disorders, including autism and hyperekplexia. Understanding of molecular mechanisms and pharmacology of glycine receptors has been hindered by a lack of high-resolution structures. Here we report electron cryo-microscopy structures of the zebrafish α1 GlyR with strychnine, glycine, or glycine and ivermectin (glycine/ivermectin). Strychnine arrests the receptor in an antagonist-bound closed ion channel state, glycine stabilizes the receptor in an agonist-bound open channel state, and the glycine/ivermectin complex adopts a potentially desensitized or partially open state. Relative to the glycine-bound state, strychnine expands the agonist-binding pocket via outward movement of the C loop, promotes rearrangement of the extracellular and transmembrane domain 'wrist' interface, and leads to rotation of the transmembrane domain towards the pore axis, occluding the ion conduction pathway. These structures illuminate the GlyR mechanism and define a rubric to interpret structures of Cys-loop receptors.


Asunto(s)
Microscopía por Crioelectrón , Receptores de Glicina/metabolismo , Receptores de Glicina/ultraestructura , Pez Cebra , Regulación Alostérica , Animales , Sitios de Unión , Glicina/metabolismo , Glicina/farmacología , Activación del Canal Iónico/efectos de los fármacos , Ivermectina/metabolismo , Ivermectina/farmacología , Modelos Moleculares , Neurotransmisores/metabolismo , Neurotransmisores/farmacología , Conformación Proteica/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores de Glicina/agonistas , Receptores de Glicina/antagonistas & inhibidores , Rotación , Transducción de Señal , Estricnina/metabolismo , Estricnina/farmacología
5.
Ecotoxicol Environ Saf ; 166: 336-344, 2018 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-30278395

RESUMEN

Cadmium (Cd) is a pervasive carcinogen and environmental endocrine disruptor. We studied the changes in learning and memory of offspring mice, whose mothers were exposed to 10 mg Cd/L via the drinking water during pregnancy and lactation period, as well as the changes of testosterone and estrogen levels, serum Cd levels, the histopathological changes and the changes in the mRNA and protein levels of different subunits of γ-aminobutyric acid receptor subtype A subunits (GABAARs) in the hippocampus at the prepuberty, puberty, young adult, and adult stages. At birth, Cd had no obvious effect on mice offspring as statistically accessed based on their body weight, body length, and tail length (all p > 0.05). After grouped, the serum Cd levels increased in the three exposed groups more than in the normal control group at stages (all p < 0.05). Only serum estradiol of female offspring at age 7 weeks was significantly decreased compared with other groups (all p < 0.05). Histopathological results showed that the arrangement of the cells in hippocampal CA1 area of mice offspring was significantly sparse in the exposed groups compared with the control group. At 5 and 7 weeks, two Cd-exposed groups showed prolonged escape latency and exploring time for the platform compared with the normal group in the Morris water maze (all p < 0.05). Only increased protein expression of GABAARα5 was found in the Cd group at these two ages. At age 12 weeks, similar impaired learning and memory of female mice, and decreased protein expression of GABAARδ was found in Cd-exposed groups. Collectively, low-dose Cd had no effect on the growth of mice offspring but affected their learning and memory, especially female offspring, at puberty, young adulthood, and adulthood through changed structure in the hippocampal CA1 area and protein expression of GABAARα5 and GABAARδ.


Asunto(s)
Cadmio/toxicidad , Carcinógenos/toxicidad , Contaminantes Ambientales/toxicidad , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Cadmio/metabolismo , Modelos Animales de Enfermedad , Estrógenos/metabolismo , Femenino , Hipocampo/efectos de los fármacos , Masculino , Ratones , Embarazo , Subunidades de Proteína/efectos de los fármacos , Receptores de GABA/metabolismo , Receptores de GABA-A/metabolismo , Maduración Sexual/efectos de los fármacos , Testosterona/metabolismo
6.
J Pharmacol Exp Ther ; 362(1): 177-185, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28483800

RESUMEN

M-channel inhibitors, especially XE991, are being used increasingly in animal experiments; however, insufficient characterization of XE991 at times confounds the interpretation of results when using this compound. Here, we demonstrate that XE991 and linopirdine are state-dependent inhibitors that favor the activated-subunit of neuronal Kv7/KCNQ channels. We performed patch-clamp experiments on homomeric Kv7.2 or heteromeric Kv7.2/3 channels expressed in Chinese hamster ovary cells to characterize XE991 and linopirdine. Neither inhibitor was efficacious around the resting membrane potential of cells in physiologic conditions. Inhibition of Kv7.2 and Kv7.2/3 channels by XE991 was closely related with channel activation. When the voltage dependence of activation was left-shifted by retigabine or right-shifted by the mutation, Kv7.2(R214D), the shift in half-activation voltage proportionally coincided with the shift in the half-effective potential for XE991 inhibition. Inhibition kinetics during XE991 wash-in was facilitated at depolarized potentials. Ten-minute washout of XE991 resulted in ∼30% current recovery, most of which was attributed to surface transport of Kv7.2 channels. Linopirdine also exhibited similar inhibition characteristics, with the exception of near- complete current recovery after washout at depolarized potentials. Inhibition kinetics of both XE991 and linopirdine was not as sensitive to changes in voltage as would be predicted by open- channel inhibition. Instead, they were well explained by binding to a single activated subunit. The characteristics of XE991 and linopirdine should be taken into account when these M-channel inhibitors are used in experiments.


Asunto(s)
Antracenos/farmacología , Indoles/farmacología , Canal de Potasio KCNQ1/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/farmacología , Piridinas/farmacología , Animales , Células CHO , Carbamatos/farmacología , Cricetinae , Cricetulus , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ2/efectos de los fármacos , Cinética , Potenciales de la Membrana/efectos de los fármacos , Mutación , Técnicas de Placa-Clamp , Fenilendiaminas/farmacología , Subunidades de Proteína/efectos de los fármacos , Ratas
7.
Biochim Biophys Acta ; 1844(4): 713-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24530967

RESUMEN

The proteasome, a complex multimeric structure strictly implicated in cell protein degradation, has gained the status of privileged drug target since its functional involvement in relevant pathways ruling the cell life, such as cell cycle, transcription and protein quality control, and the recent marketing of bortezomib as proteasome inhibitor for anti-cancer therapy. The marine γ-hydroxybutenolide terpenoid petrosaspongiolide M has been recently discovered as new proteasome inhibitor through a chemical proteomic approach and in cell biological assays. In this study a deep investigation has been carried out on the molecular mechanism of interaction of petrosaspongiolide M with the immunoproteasome, a proteasomal variant mainly involved in the immune responses. The results define a picture in which petrosaspongiolide M exerts its inhibitory activity by binding the active sites in the inner core of the immunoproteasome and/or covalently linking a Lys residue at the proteasome core/11S activator particle interface. Moreover, petrosaspongiolide M is also able to impair autophagy, a complementary pathway involved in protein degradation and cross-talking with the proteasome system. On this basis, petrosaspongiolide M could represent an interesting molecule for its propensity to modulate intracellular proteolysis through a dual inhibition of the immunoproteasome and autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Ácido Oleanólico/análogos & derivados , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/química , Subunidades de Proteína/química , Proteolisis/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Humanos , Cinética , Linfocitos/citología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Ácido Oleanólico/química , Ácido Oleanólico/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/inmunología , Inhibidores de Proteasoma/farmacología , Unión Proteica , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/inmunología , Espectrometría de Masa por Ionización de Electrospray
8.
Int J Neuropsychopharmacol ; 17(8): 1221-32, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24556017

RESUMEN

Stress exposure during adolescence/early adulthood has been shown to increase the risk for psychiatric disorders such as schizophrenia. Reelin plays an essential role in brain development and its levels are decreased in schizophrenia. However, the relationship between stress exposure and reelin expression remains unclear. We therefore treated adolescent reelin heteroyzogous mice (HRM) and wild-type (WT) littermates with the stress hormone, corticosterone (CORT) in their drinking water (25 mg/l) for 3 wk. In adulthood, we measured levels of full-length (FL) reelin and the N-R6 and N-R2 cleavage fragments in the frontal cortex (FC) and dorsal (DH) and ventral (VH) hippocampus. As expected, levels of all reelin forms were approximately 50% lower in HRMs compared to WT. In male mice, CORT treatment significantly decreased FL and N-R2 expression in the FC and N-R2 and N-R6 levels in the DH. This reelin down-regulation was accompanied by significant reductions in downstream N-methyl-D-aspartate (NMDA) GluN2C subunit levels. There were no effects of CORT treatment in the VH of either of the sexes and only subtle changes in female DH. CORT-induced reelin and GluN2C down-regulation in males was not associated with changes in two GABAergic neuron markers, GAD67 and parvalbumin, or glucocorticoids receptors (GR). These results show that CORT treatment causes long-lasting and selective reductions of reelin form levels in male FC and DH accompanied by changes in NMDAR subunit composition. This sex-specific reelin down-regulation in regions implicated in schizophrenia could be involved in the effects of stress in this disease.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Corticosterona/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Lóbulo Frontal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/metabolismo , Serina Endopeptidasas/metabolismo , Caracteres Sexuales , Animales , Moléculas de Adhesión Celular Neuronal/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Lóbulo Frontal/metabolismo , Glutamato Descarboxilasa/metabolismo , Heterocigoto , Hipocampo/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Parvalbúminas/metabolismo , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteína Reelina , Esquizofrenia/etiología , Serina Endopeptidasas/genética
9.
Nat Cell Biol ; 9(6): 636-45, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17486119

RESUMEN

Stromal interacting molecule 1 (STIM1) is a Ca(2+) sensor that conveys the Ca(2+) load of the endoplasmic reticulum to store-operated channels (SOCs) at the plasma membrane. Here, we report that STIM1 binds TRPC1, TRPC4 and TRPC5 and determines their function as SOCs. Inhibition of STIM1 function inhibits activation of TRPC5 by receptor stimulation, but not by La(3+), suggesting that STIM1 is obligatory for activation of TRPC channels by agonists, but STIM1 is not essential for channel function. Through a distinct mechanism, STIM1 also regulates TRPC3 and TRPC6. STIM1 does not bind TRPC3 and TRPC6, and regulates their function indirectly by mediating the heteromultimerization of TRPC3 with TRPC1 and TRPC6 with TRPC4. TRPC7 is not regulated by STIM1. We propose a new definition of SOCs, as channels that are regulated by STIM1 and require the store depletion-mediated clustering of STIM1. By this definition, all TRPC channels, except TRPC7, function as SOCs.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canales Catiónicos TRPC/metabolismo , Sitios de Unión/fisiología , Señalización del Calcio/efectos de los fármacos , Línea Celular , Agonistas Colinérgicos/farmacología , Gadolinio/farmacología , Humanos , Sustancias Macromoleculares/metabolismo , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Unión Proteica/fisiología , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/efectos de los fármacos , Canales Catiónicos TRPC/genética
10.
Biophys J ; 105(3): 640-7, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23931312

RESUMEN

Improving our understanding of the mechanisms and effects of anesthetics is a critically important part of neuroscience. The currently dominant theory is that anesthetics and similar molecules act by binding to Cys-loop receptors in the postsynaptic terminal of nerve cells and potentiate or inhibit their function. Although structures for some of the most important mammalian channels have still not been determined, a number of important results have been derived from work on homologous cationic channels in bacteria. However, partly due to the lack of a nervous system in bacteria, there are a number of questions about how these results relate to higher organisms. The recent determination of a structure of the eukaryotic chloride channel, GluCl, is an important step toward accurate modeling of mammalian channels, because it is more similar in function to human Cys-loop receptors such as GABAAR or GlyR. One potential issue with using GluCl to model other receptors is the presence of the large ligand ivermectin (IVM) positioned between all five subunits. Here, we have performed a series of microsecond molecular simulations to study how the dynamics and structure of GluCl change in the presence versus absence of IVM. When the ligand is removed, subunits move at least 2 Å closer to each other compared to simulations with IVM bound. In addition, the pore radius shrinks to 1.2 Å, all of which appears to support a model where IVM binding between subunits stabilizes an open state, and that the relaxed nonIVM conformations might be suitable for modeling other channels. Interestingly, the presence of IVM also has an effect on the structure of the important loop C located at the neurotransmitter-binding pocket, which might help shed light on its partial agonist behavior.


Asunto(s)
Canales de Cloruro/química , Ivermectina/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Secuencia de Aminoácidos , Animales , Canales de Cloruro/efectos de los fármacos , Canales de Cloruro/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Ivermectina/química , Ligandos , Macaca mulatta , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo
11.
Neuron ; 55(5): 779-85, 2007 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-17785184

RESUMEN

Synaptic NMDA-type glutamate receptors (NMDARs) play important roles in synaptic plasticity, brain development, and pathology. In the last few years, the view of NMDARs as relatively fixed components of the postsynaptic density has changed. A number of studies have now shown that both the number of receptors and their subunit compositions can be altered. During development, the synaptic NMDARs subunit composition changes, switching from predominance of NR2B-containing to NR2A-containing receptors, but little is known about the mechanisms involved in this developmental process. Here, we report that, depending on the pattern of NMDAR activation, the subunit composition of synaptic NMDARs is under extremely rapid, bidirectional control at neonatal synapses. This switching, which is at least as rapid as that seen with AMPARs, will have immediate and dramatic consequences on the integrative capacity of the synapse.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Envejecimiento/fisiología , Animales , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Técnicas de Cultivo de Órganos , Piperidinas/farmacología , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Factores de Tiempo
12.
Alcohol Alcohol ; 46(6): 672-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21903702

RESUMEN

AIMS: Tolerance to ethanol-induced inhibition of N-methyl-D-aspartate receptors (NMDARs) is thought to underlie the acute adaptive mechanisms against ethanol. To explore these compensatory upregulating mechanisms of NMDARs, we investigated the expression and phosphorylation of NMDAR subunits in vivo following an acute ethanol treatment. METHODS: Male Sprague-Dawley rats were given 4 g/kg ethanol, and the phospho-S896-NR1, NR2A and NR2B subunits of NMDAR were immunoblotted from the cerebral cortex and hippocampus. We also examined the mRNAs and ubiquitinated forms of the NR2A and NR2B subunits. RESULTS: Acute ethanol treatment increased phospho-S896-NR1 at 30 min in the cerebral cortex and hippocampus, and the increase was maintained until 2 h in the hippocampus. Ethanol increased total NR2A and NR2B expression at 30 min in the cortex and hippocampus, and the NR2A increase was maintained until 2 h in the hippocampus. The increased expression of the NR2A and NR2B subunits was not associated with statistically significant alterations in mRNA expression or protein ubiquitination. CONCLUSION: Acute ethanol treatment increased NR1 subunit phosphorylation and NR2A and NR2B subunit expression in the cerebral cortex and hippocampus of rats. These effects of ethanol on the NMDAR subunits may underlie the mechanisms that compensate for ethanol-induced inhibition of NMDARs. However, the regulation of NR2A and NR2B in this paradigm is not dependent on transcriptional changes.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Subunidades de Proteína/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Fosforilación/efectos de los fármacos , Subunidades de Proteína/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Ubiquitinación/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
13.
Pharmacol Rep ; 73(4): 1096-1108, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34426901

RESUMEN

The role of adenosine A2A receptor (A2AR) and striatal-enriched protein tyrosine phosphatase (STEP) interactions in the striatal-pallidal GABA neurons was recently discussed in relation to A2AR overexpression and cocaine-induced increases of brain adenosine levels. As to phosphorylation, combined activation of A2AR and metabotropic glutamate receptor 5 (mGluR5) in the striatal-pallidal GABA neurons appears necessary for phosphorylation of the GluA1 unit of the AMPA receptor to take place. Robert Yasuda (J Neurochem 152: 270-272, 2020) focused on finding a general mechanism by which STEP activation is enhanced by increased A2AR transmission in striatal-pallidal GABA neurons expressing A2AR and dopamine D2 receptor. In his Editorial, he summarized in a clear way the significant effects of A2AR activation on STEP in the dorsal striatal-pallidal GABA neurons which involves a rise of intracellular levels of calcium causing STEP activation through its dephosphorylation. However, the presence of the A2AR in an A2AR-fibroblast growth factor receptor 1 (FGFR1) heteroreceptor complex can be required in the dorsal striatal-pallidal GABA neurons for the STEP activation. Furthermore, Won et al. (Proc Natl Acad Sci USA 116: 8028-8037, 2019) found in mass spectrometry experiments that the STEP splice variant STEP61 can bind to mGluR5 and inactivate it. In addition, A2AR overexpression can lead to increased formation of A2AR-mGluR5 heterocomplexes in ventral striatal-pallidal GABA neurons. It involves enhanced facilitatory allosteric interactions leading to increased Gq-mediated mGluR5 signaling activating STEP. The involvement of both A2AR and STEP in the actions of cocaine on synaptic downregulation was also demonstrated. The enhancement of mGluR5 protomer activity by the A2AR protomer in A2AR-mGluR5 heterocomplexes in the nucleus accumbens shell appears to have a novel significant role in STEP mechanisms by both enhancing the activation of STEP and being a target for STEP61.


Asunto(s)
Neuronas GABAérgicas/fisiología , Fosforilación/genética , Fosforilación/fisiología , Células del Asta Posterior/fisiología , Receptor de Adenosina A2A/metabolismo , Animales , Cocaína/farmacología , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/patología , Neuronas GABAérgicas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Células del Asta Posterior/efectos de los fármacos , Subunidades de Proteína/efectos de los fármacos , Proteínas Tirosina Fosfatasas/genética , Receptor de Adenosina A2A/genética , Receptor del Glutamato Metabotropico 5/genética
14.
Sci Rep ; 11(1): 4257, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33608565

RESUMEN

The worldwide CoVid-19 pandemic has led to an unprecedented push across the whole of the scientific community to develop a potent antiviral drug and vaccine as soon as possible. Existing academic, governmental and industrial institutions and companies have engaged in large-scale screening of existing drugs, in vitro, in vivo and in silico. Here, we are using in silico modelling of possible SARS-CoV-2 drug targets, as deposited on the Protein Databank (PDB), and ascertain their dynamics, flexibility and rigidity. For example, for the SARS-CoV-2 spike protein-using its complete homo-trimer configuration with 2905 residues-our method identifies a large-scale opening and closing of the S1 subunit through movement of the S[Formula: see text] domain. We compute the full structural information of this process, allowing for docking studies with possible drug structures. In a dedicated database, we present similarly detailed results for the further, nearly 300, thus far resolved SARS-CoV-2-related protein structures in the PDB.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Desarrollo de Medicamentos/métodos , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Antivirales/uso terapéutico , Sitios de Unión , COVID-19/epidemiología , COVID-19/virología , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Pandemias/prevención & control , Unión Proteica , Dominios Proteicos/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/ultraestructura
15.
Mol Pharmacol ; 77(5): 874-83, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20159947

RESUMEN

Evidence suggests that the long-term adaptations in the hippocampus after repeated drug treatment may parallel its role during memory formation. The neuroplasticity that subserves learning and memory is also believed to underlie addictive processes. We have reported previously that repeated morphine administration alters local distribution of endocytic proteins at hippocampal synapses, which could in turn affect expression of glutamate receptors. Glutamatergic systems, including alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs), are believed to be involved in opiate-induced neuronal and behavioral plasticity, although the mechanisms underlying these effects are only beginning to be understood. The present study further examines the effects of repeated morphine administration on the expression and composition of AMPARs and the functional ramifications. Twelve hours after the last morphine injection, we observed an increased expression of AMPARs lacking glutamate receptor (GluR) 2 in hippocampal synaptic fractions. Immunoblotting studies show that 12 h after morphine treatment, GluR1 subunits are increased at the postsynaptic density (PSD) and at extrasynaptic sites, whereas GluR3 subunits are only increased at the PSD, and they show how this alters receptor subunit composition. In addition, we provide electrophysiological evidence that AMPARs are switched to Ca(2+)-permeable (GluR2-lacking) at the synapse 12 h after repeated morphine treatment, affecting the magnitude of long-term depression at hippocampal neurons. We propose that morphine-induced changes in glutamatergic synaptic transmission in the hippocampus may play an important role in the neuroadaptations induced by repeated morphine administration.


Asunto(s)
Morfina/farmacología , Receptores AMPA/biosíntesis , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Antagonistas del GABA/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácidos Fosfínicos/farmacología , Fosforilación , Picrotoxina/farmacología , Propanolaminas/farmacología , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/genética , Receptores AMPA/deficiencia , Receptores AMPA/efectos de los fármacos , Receptores AMPA/genética , Receptores AMPA/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
16.
J Cell Physiol ; 223(2): 511-8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20162564

RESUMEN

Chondrocytes possess the capacity to transduce load-induced mechanical stimuli into electrochemical signals. The aim of this study was to functionally characterize an ion channel activated in response to membrane stretch in isolated primary equine chondrocytes. We used patch-clamp electrophysiology to functionally characterize this channel and immunohistochemistry to examine its distribution in articular cartilage. In cell-attached patch experiments, the application of negative pressures to the patch pipette (in the range of 20-200 mmHg) activated ion channel currents in six of seven patches. The mean activated current was 45.9 +/- 1.1 pA (n = 4) at a membrane potential of 33 mV (cell surface area approximately 240 microm(2)). The mean slope conductance of the principal single channels resolved within the total stretch-activated current was 118 +/- 19 pS (n = 6), and reversed near the theoretical potassium equilibrium potential, E(K+), suggesting it was a high-conductance potassium channel. Activation of these high-conductance potassium channels was inhibited by extracellular TEA (K(d) approx. 900 microM) and iberiotoxin (K(d) approx. 40 nM). This suggests that the current was largely carried by BK-like potassium (MaxiK) channels. To further characterize these BK-like channels, we used inside-out patches of chondrocyte membrane: we found these channels to be activated by elevation in bath calcium concentration. Immunohistochemical staining of equine cartilage samples with polyclonal antibodies to the alpha1- and beta1-subunits of the BK channel revealed positive immunoreactivity for both subunits in superficial zone chondrocytes. These experiments support the hypothesis that functional BK channels are present in chondrocytes and may be involved in mechanotransduction and chemotransduction.


Asunto(s)
Cartílago/metabolismo , Condrocitos/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Estrés Mecánico , Animales , Cartílago/citología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Condrocitos/citología , Condrocitos/efectos de los fármacos , Caballos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Mecanotransducción Celular/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Presión/efectos adversos , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Soporte de Peso/fisiología
17.
J Neurochem ; 113(4): 1036-45, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20420581

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) transmit the agonist signal to the channel gate through a number of extracellular domains. We have previously shown that particular details of the process of coupling binding to gating could be quantitative and qualitatively different in muscle and neuronal type nAChRs. We have extended previous studies on homomeric alpha7 nAChRs to heteromeric alpha3beta4 nAChRs, by mutating residues located at loops 2 and 7, and M2-M3 linker of both alpha3 and beta4 subunits which, in order to monitor surface expression, were modified to bind alpha-bungarotoxin, and expressed in Xenopus oocytes. We show that, in general, mutations in these domains of both alpha3 and beta4 subunits affect the gating function, although the effects are slightly larger if they are inserted in the alpha3 subunit. However, the involvement of a previously reported intrasubunit interaction in coupling (Gln48-Ile130) seems to be restricted to the beta4 subunit. We also show that mutations at these domains, particularly loop 2 of the alpha3 subunit, change the pharmacological profile of alpha3beta4 nAChRs, decreasing nicotine's and increasing cytisine's effectiveness relative to acetylcholine. It is concluded that, unlike muscle nAChRs, the non-alpha subunits play a relevant role in the coupling process of neuronal alpha3beta4 nAChRs.


Asunto(s)
Membrana Celular/química , Activación del Canal Iónico/genética , Receptores Nicotínicos/química , Acetilcolina/metabolismo , Acetilcolina/farmacología , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Femenino , Humanos , Activación del Canal Iónico/efectos de los fármacos , Mutación/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Oocitos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Subunidades de Proteína/química , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/genética , Receptores Nicotínicos/efectos de los fármacos , Receptores Nicotínicos/genética , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Xenopus laevis
18.
Mol Pain ; 6: 83, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-21108841

RESUMEN

BACKGROUND: Neuropathic pain is characterized by hypersensitivity to innocuous stimuli (tactile allodynia) that is nearly always resistant to NSAIDs or even opioids. Gabapentin, a GABA analogue, was originally developed to treat epilepsy. Accumulating clinical evidence supports the effectiveness of this drug for diverse neuropathic pain. In this study, we showed that the anti-allodynic effect of gabapentin was changed by the circadian oscillation in the expression of its target molecule, the calcium channel α2δ-1 subunit. RESULTS: Mice were underwent partial sciatic nerve ligation (PSL) to create a model of neuropathic pain. The paw withdrawal threshold (PWT) in PSL mice significantly decreased and fluctuated with a period length about 24 h. The PWT in PSL mice was dose-dependently increased by intraperitoneal injection of gabapentin, but the anti-allodynic effects varied according to its dosing time. The protein levels of α2δ-1 subunit were up-regulated in the DRG of PSL mice, but the protein levels oscillated in a circadian time-dependent manner. The time-dependent oscillation of α2δ-1 subunit protein correlated with fluctuations in the maximal binding capacity of gabapentin. The anti-allodynic effect of gabapentin was attenuated at the times of the day when α2δ-1 subunit protein was abundant. CONCLUSIONS: These findings suggest that the dosing time-dependent difference in the anti-allodynic effects of gabapentin is attributable to the circadian oscillation of α2δ-1 subunit expression in the DRG and indicate that the optimizing its dosing schedule helps to achieve rational pharmacotherapy for neuropathic pain.


Asunto(s)
Aminas/administración & dosificación , Canales de Calcio/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/administración & dosificación , Hiperalgesia/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Ácido gamma-Aminobutírico/administración & dosificación , Aminas/farmacología , Aminas/uso terapéutico , Animales , Ritmo Circadiano/fisiología , Ácidos Ciclohexanocarboxílicos/farmacología , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Gabapentina , Ratones , Subunidades de Proteína/efectos de los fármacos , Factores de Tiempo , Ácido gamma-Aminobutírico/farmacología , Ácido gamma-Aminobutírico/uso terapéutico
19.
Mol Pain ; 6: 5, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-20092646

RESUMEN

The functional properties of alpha-amino-3-hydroxy-5-methy-4-isoxazole propionate (AMPA) receptors in different brain regions, such as hippocampus and cerebellum, have been well studied in vitro and in vivo. The AMPA receptors present a unique characteristic in the mechanisms of subunit regulation during LTP (long-term potentiation) and LTD (long-term depression), which are involved in the trafficking, altered composition and phosphorylation of AMPA receptor subunits. Accumulated data have demonstrated that spinal AMPA receptors play a critical role in the mechanism of both acute and persistent pain. However, less is known about the biochemical regulation of AMPA receptor subunits in the spinal cord in response to painful stimuli. Recent studies have shown that some important regulatory processes, such as the trafficking of AMPA receptor subunit, subunit compositional changes, phosphorylation of AMPA receptor subunits, and their interaction with partner proteins may contribute to spinal nociceptive transmission. Of all these regulation processes, the phosphorylation of AMPA receptor subunits is the most important since it may trigger or affect other cellular processes. Therefore, these study results may suggest an effective strategy in developing novel analgesics targeting AMPA receptor subunit regulation that may be useful in treating persistent and chronic pain without unacceptable side effects in the clinics.


Asunto(s)
Dolor/metabolismo , Receptores AMPA/metabolismo , Médula Espinal/metabolismo , Animales , Ácido Glutámico/metabolismo , Humanos , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Fosforilación/efectos de los fármacos , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Receptores AMPA/efectos de los fármacos , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Médula Espinal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
20.
Artículo en Zh | MEDLINE | ID: mdl-20465948

RESUMEN

OBJECTIVE: To investigate the dynamic changes of neurofilaments (NFs) proteins in spinal cords of hens with phenylmethylsulfonyl fluoride (PMSF) pretreatment for exploring the mechanism of tri-o-cresyl phosphate (TOCP)-induced delayed neuropathy (OPIDN). METHOD: Adult Roman hens were randomly divided into three groups, control, TOCP and PMSF + TOCP. Birds in PMSF + TOCP set were pretreated with PMSF, 24 hours later, hens in both TOCP group and PMSF + TOCP group were administrated with TOCP at a single dosage of 750 mg/kg. Then all animals were sacrificed on the corresponding time-points of 1, 5, 10, and 21 days respectively after dosing of 750 mg/kg TOCP. The spinal cords were dissected, homogenized, and centrifuged at 100,000 x g. The levels of high molecular neurofilament (NF-H), medium molecular neurofilament (NF-M) and low molecular neurofilament (NF-L) in both pellet and supernatant fractions of spinal cords were determined by SDS-PAGE and Western-blotting. RESULTS: The hens in TOCP group showed paralysis gait at the end of 21-day experimental period. The levels of NFs proteins in spinal cords changed obviously. Compared with control, the NFs in pellet showed a dramatic decrease on day 10 and then followed by a recovery. In the supernatant, the NFs proteins showed similar changes, which decreased significantly on day 10 and almost recovered control on day 21. Such as, NF-L, NF-M and NF-H decreased by 51%, 86% and 38% on day 10. The OPIDN signs were not observed in PMSF + TOCP group, and imbalances of NFs were obviously alleviated. Compared with control, only NF-M in pellet increased by 21% (P < 0.05) on day 21, others remained no changes; The levels of NF-H and NF-M in supernatant respectively increased by 19% and 35% on day 21, others were no significant statistical differences. CONCLUSION: TOCP may induce imbalance of NFs levels in progress of OPIDN, and PMSF pretreatment may protect animals from OPIDN by reducing above changes, which may explain that TOCP-induced imbalance of NFs may be connected with the occurrence and development of OPIDN.


Asunto(s)
Proteínas de Neurofilamentos/efectos de los fármacos , Fluoruro de Fenilmetilsulfonilo/farmacología , Médula Espinal/patología , Tritolilfosfatos/toxicidad , Animales , Pollos , Femenino , Subunidades de Proteína/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA