Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
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
2.
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
4.
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
5.
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
6.
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
7.
Cell Chem Biol ; 27(11): 1359-1370.e8, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32649904

RESUMEN

Multidrug resistance (MDR) in cancer remains a major challenge for the success of chemotherapy. Natural products have been a rich source for the discovery of drugs against MDR cancers. Here, we applied high-throughput cytotoxicity screening of an in-house natural product library against MDR SGC7901/VCR cells and identified that the cyclodepsipeptide verucopeptin demonstrated notable antitumor potency. Cytological profiling combined with click chemistry-based proteomics revealed that ATP6V1G directly interacted with verucopeptin. ATP6V1G, a subunit of the vacuolar H+-ATPase (v-ATPase) that has not been previously targeted, was essential for SGC7901/VCR cell growth. Verucopeptin exhibited strong inhibition of both v-ATPase activity and mTORC1 signaling, leading to substantial pharmacological efficacy against SGC7901/VCR cell proliferation and tumor growth in vivo. Our results demonstrate that targeting v-ATPase via its V1G subunit constitutes a unique approach for modulating v-ATPase and mTORC1 signaling with great potential for the development of therapeutics against MDR cancers.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Depsipéptidos/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Productos Biológicos/síntesis química , Productos Biológicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Depsipéptidos/síntesis química , Depsipéptidos/química , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Subunidades de Proteína/efectos de los fármacos , Proteómica , ATPasas de Translocación de Protón Vacuolares/metabolismo
8.
Oncogene ; 38(13): 2364-2379, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30518875

RESUMEN

DNA replication machinery is responsible for accurate and efficient duplication of the chromosome. Since inhibition of DNA replication can lead to replication fork stalling, resulting in DNA damage and apoptotic death, inhibitors of DNA replication are commonly used in cancer chemotherapy. Ribonucleotide reductase (RNR) is the rate-limiting enzyme in the biosynthesis of deoxyribonucleoside triphosphates (dNTPs) that are essential for DNA replication and DNA damage repair. Gemcitabine, a nucleotide analog that inhibits RNR, has been used to treat various cancers. However, patients often develop resistance to this drug during treatment. Thus, new drugs that inhibit RNR are needed to be developed. In this study, we identified a synthetic analog of resveratrol (3,5,4'-trihydroxy-trans-stilbene), termed DHS (trans-4,4'-dihydroxystilbene), that acts as a potent inhibitor of DNA replication. Molecular docking analysis identified the RRM2 (ribonucleotide reductase regulatory subunit M2) of RNR as a direct target of DHS. At the molecular level, DHS induced cyclin F-mediated down-regulation of RRM2 by the proteasome. Thus, treatment of cells with DHS reduced RNR activity and consequently decreased synthesis of dNTPs with concomitant inhibition of DNA replication, arrest of cells at S-phase, DNA damage, and finally apoptosis. In mouse models of tumor xenografts, DHS was efficacious against pancreatic, ovarian, and colorectal cancer cells. Moreover, DHS overcame both gemcitabine resistance in pancreatic cancer and cisplatin resistance in ovarian cancer. Thus, DHS is a novel anti-cancer agent that targets RRM2 with therapeutic potential either alone or in combination with other agents to arrest cancer development.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Neoplasias/patología , Ribonucleótido Reductasas/antagonistas & inhibidores , Estilbenos/farmacología , Animales , Línea Celular Tumoral , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Femenino , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Subunidades de Proteína/efectos de los fármacos , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/metabolismo , Estilbenos/química , Ensayos Antitumor por Modelo de Xenoinjerto
9.
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
10.
Physiol Behav ; 189: 26-31, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29476874

RESUMEN

Compared to younger individuals, older human subjects have significantly lower food intakes and an increased satiety response. N-methyl-d-aspartate (NMDA) receptors expressed by vagal afferent neurons originating from nodose ganglia (NG) are involved in modulating the satiety response. The present study investigated how NMDA receptor subunit phenotypes in NG neurons change with age and how these age-related alterations in food intake are modulated by presynaptic NMDA receptors in the NG of male Sprague Dawley rats (six week-old and sixty week-old). Food intake was measured at 30-, 60-, and 120-min following intraperitoneal administration of cholecystokinin (CCK) or the non-competitive NMDA receptor antagonist MK-801. Immunofluorescence was used to determine NMDA receptor subunit expression (NR1, NR2B, NR2C, and NR2D) in the NG. The results showed that, CCK reduced food intake at 30-, 60-, and 120-min post injection in both young and the middle-age animals, with no statistical difference between the groups at 30- and 60-min. In contrast, MK-801 produced an increase in food intake that was significantly higher in middle-age rats compared to young animals at all time points studied. NR1 subunit was expressed by almost all NG neurons in both age groups. In young rats, NR2B, NR2C, and NR2D subunits were expressed in 56.1%, 49.3%, and 13.9% of NG neurons, respectively. In contrast, only 30.3% of the neuronal population in middle-aged rats expressed NR2B subunit immunoreactivity, NR2C was present in 34.1%, and only 10.6% of total neurons expressed the NR2D subunit. In conclusion, glutamate-dependent regulation of food intake is altered with age and one of the potential mechanisms through which this age-related changes in intake occur is changes in NMDA receptor phenotypes on vagal afferent neurons located in NG.


Asunto(s)
Envejecimiento/fisiología , Ingestión de Alimentos/fisiología , Neuronas Aferentes/fisiología , Ganglio Nudoso/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Respuesta de Saciedad/fisiología , Animales , Colecistoquinina/farmacología , Maleato de Dizocilpina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Masculino , Neuronas Aferentes/efectos de los fármacos , Ganglio Nudoso/efectos de los fármacos , Fenotipo , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/efectos de los fármacos , Ratas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/biosíntesis
11.
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
12.
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
13.
J Med Chem ; 57(14): 6083-91, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-24950374

RESUMEN

Inhibition of the catalytic subunit of the heterodimeric methionine S-adenosyl transferase-2 (MAT2A) with fluorinated N,N-dialkylaminostilbenes (FIDAS agents) offers a potential avenue for the treatment of liver and colorectal cancers where upregulation of this enzyme occurs. A study of structure-activity relationships led to the identification of the most active compounds as those with (1) either a 2,6-difluorostyryl or 2-chloro-6-fluorostyryl subunit, (2) either an N-methylamino or N,N-dimethylamino group attached in a para orientation relative to the 2,6-dihalostyryl subunit, and (3) either an N-methylaniline or a 2-(N,N-dimethylamino)pyridine ring. These modifications led to FIDAS agents that were active in the low nanomolar range, that formed water-soluble hydrochloride salts, and that possessed the desired property of not inhibiting the human hERG potassium ion channel at concentrations at which the FIDAS agents inhibit MAT2A. The active FIDAS agents may inhibit cancer cells through alterations of methylation reactions essential for cancer cell survival and growth.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Dominio Catalítico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Metionina Adenosiltransferasa/antagonistas & inhibidores , Piridinas/farmacología , Pirimidinas/farmacología , Estilbenos/farmacología , Compuestos de Anilina/síntesis química , Compuestos de Anilina/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Biocatálisis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Metionina Adenosiltransferasa/metabolismo , Estructura Molecular , Subunidades de Proteína/efectos de los fármacos , Piridinas/síntesis química , Piridinas/química , Pirimidinas/síntesis química , Pirimidinas/química , Estilbenos/síntesis química , Estilbenos/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
14.
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
15.
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
16.
J Neuroendocrinol ; 26(1): 26-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24313753

RESUMEN

In supraoptic nucleus (SON) magnocellular neurosecretory cells (MNCs), γ-GABA, via activation of GABAA receptors (GABAA Rs), mediates persistent tonic inhibitory currents (Itonic ), as well as conventional inhibitory postsynaptic currents (IPSCs, Iphasic ). In the present study, we examined the functional significance of Itonic in SON MNCs challenged by 24-h water deprivation (24WD). Although the main characteristics of spontaneous IPSCs were similar in 24WD compared to euhydrated (EU) rats, Itonic , measured by bicuculline (BIC)-induced Iholding shifts, was significantly smaller in 24WD compared to EU rats (P < 0.05). Propofol and diazepam prolonged IPSC decay time to a similar extent in both groups but induced less Itonic in 24WD compared to EU rats, suggesting a selective decrease in GABAA receptors mediating Itonic over Iphasic in 24WD rats. THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol), a preferential δ subunit agonist, and L-655,708, a GABAA receptor α5 subunit selective imidazobenzodiazepine, caused a significantly smaller inward and outward shift in Iholding , respectively, in 24WD compared to EU rats (P < 0.05 in both cases), suggesting an overall decrease in the α5 subunit-containing GABAA Rs and the δ subunit-containing receptors mediating Itonic in 24WD animals. Consistent with a decrease in 24WD Itonic , bath application of GABA induced significantly less inhibition of the neuronal firing activity in 24WD compared to EU SON MNCs (P < 0.05). Taken together, the results of the present study indicate a selective decrease in GABAA Rs functions mediating Itonic as opposed to those mediating Iphasic in SON MNCs, demonstrating the functional significance of Itonic with respect to increasing neuronal excitability and hormone secretion in 24WD rats.


Asunto(s)
Potenciales Postsinápticos Inhibidores/fisiología , Células Neuroendocrinas/fisiología , Receptores de GABA-A/fisiología , Núcleo Supraóptico/citología , Núcleo Supraóptico/fisiología , Privación de Agua/fisiología , Animales , Bicuculina/farmacología , Diazepam/farmacología , Agonistas del GABA/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Hipnóticos y Sedantes/farmacología , Imidazoles/farmacología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Isoxazoles/farmacología , Masculino , Células Neuroendocrinas/efectos de los fármacos , Propofol/farmacología , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/fisiología , Ratas , Núcleo Supraóptico/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología
17.
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
18.
Neuropharmacology ; 73: 398-403, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23810831

RESUMEN

The 5-HT3 receptor is a cation selective member of the pentameric Cys-loop ligand-gated ion channels. While five subunits are known to exist, only two receptor subtypes have been significantly characterized: the homomeric receptor consisting of five A subunits and the heteromeric receptor containing both A and B subunits. The agonist recognition and activation of these receptors is orchestrated by six recognition loops three, A-C, on the principal subunit, and three, D-F, on the complementary subunit. In this study we have focused on the B loop of the principal subunit and loop D of the complementary subunit where aligned amino acids differ between the two subunits. A mutational analysis has been carried out using both 5-HT and m-chlorophenylbiguanide (mCPBG) to characterize receptor activation in the mutant receptors using two-electrode voltage clamp in Xenopus oocytes. The results show that the B loop W178I mutation of the 5-HT3A subunit markedly reduces the efficacy of mCPBG in both the homomeric and heteromeric receptors, while activation by 5-HT remains intact. Replacement of the D loop amino acid triplet RQY of the 5-HT3A subunit, with the aligned residues from the 5-HT3B subunit, QEV, converts 5-HT to a weak partial agonist in both the homomer and heteromer, but does not compromise activation by mCPBG. Exchange of the RQY triplet for the 5-HT3B subunit homologue, QEV, increases the Hill coefficient and decreases the EC50 of this mutant when expressed with the wild type 5-HT3A subunit.


Asunto(s)
Biguanidas/farmacología , Subunidades de Proteína/fisiología , Receptores de Serotonina 5-HT3/química , Receptores de Serotonina 5-HT3/metabolismo , Agonistas del Receptor de Serotonina 5-HT3/farmacología , Serotonina/farmacología , Animales , Análisis Mutacional de ADN , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Oocitos/efectos de los fármacos , Oocitos/fisiología , Subunidades de Proteína/efectos de los fármacos , Receptores de Serotonina 5-HT3/genética , Xenopus
19.
J Rheumatol ; 40(5): 663-73, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23504381

RESUMEN

OBJECTIVE: Dysregulation of proteasome subunit ß1i expression has been shown in total blood mononuclear cells (PBMC) from patients with primary Sjögren syndrome (pSS), a B cell-driven systemic autoimmune disorder. METHODS: Proteasome activation was investigated in sorted blood cells from patients with pSS and controls by measuring transcript levels of constitutive (ß1/ß2/ß5) and corresponding immunoproteasome catalytic subunits (ß1i/ß2i/ß5i) using real-time PCR. At protein level, ß1i protein expression was analyzed by immunoblotting. Functional effects of proteasome inhibition on proteolytic activity and induction of apoptosis were also evaluated in cellular subsets. RESULTS: The proteasome was found to be activated in pSS, with upregulation of gene expression of catalytic proteasome subunits. Western blot analysis revealed decreased ß1i protein expression in pSS B lymphocytes, with decreased protein despite increased messenger RNA (mRNA) levels. After proteasome inhibition in vitro, proteolytic activity was less reduced and resistance to apoptosis was increased in B lymphocytes compared to other cells. CONCLUSION: In pSS, catalytic subunits of the proteasome are upregulated at the mRNA level, while dysregulation of subunit ß1i is attributed to B lymphocytes. B cell resistance after proteasome inhibition differs from the classical concept of increased susceptibility toward inhibition in activated cells, supporting the novel notion that susceptibility depends on cellular intrinsic factors and on proteasome activation.


Asunto(s)
Linfocitos B/enzimología , Regulación de la Expresión Génica , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma/metabolismo , Síndrome de Sjögren/genética , Adulto , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Ácidos Borónicos/farmacología , Bortezomib , Dominio Catalítico/efectos de los fármacos , Dominio Catalítico/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Pacientes Ambulatorios , Complejo de la Endopetidasa Proteasomal/biosíntesis , Inhibidores de Proteasoma/farmacología , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/genética , Pirazinas/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Síndrome de Sjögren/sangre , Síndrome de Sjögren/patología
20.
CNS Drugs ; 26(3): 229-44, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22145708

RESUMEN

Clobazam was initially developed in the early 1970s as a nonsedative anxiolytic agent, and is currently available as adjunctive therapy for epilepsy and anxiety disorders in more than 100 countries. In October 2011, clobazam (Onfi™; Lundbeck Inc., Deerfield, IL, USA) was approved by the US FDA for use as adjunctive therapy for the treatment of seizures associated with Lennox-Gastaut syndrome in patients aged 2 years and older. It is a long-acting 1,5-benzodiazepine whose structure distinguishes it from the classic 1,4-benzodiazepines, such as diazepam, lorazepam and clonazepam. Clobazam is well absorbed, with peak concentrations occurring linearly 1-4 hours after administration. Both clobazam and its active metabolite, N-desmethylclobazam, are metabolized in the liver via the cytochrome P450 pathway. The mean half-life of N-desmethylclobazam (67.5 hours) is nearly double the mean half-life of clobazam (37.5 hours). Clobazam was synthesized with the anticipation that its distinct chemical structure would provide greater efficacy with fewer benzodiazepine-associated adverse effects. Frequently reported adverse effects of clobazam therapy include dizziness, sedation, drowsiness and ataxia. Evidence gathered from approximately 50 epilepsy clinical trials in adults and children indicated that the sedative effects observed with clobazam treatment were less severe than those reported with 1,4-benzodiazepines. In several studies of healthy volunteers and patients with anxiety, clobazam appeared to enhance participants' performance in cognitive tests, further distinguishing it from the 1,4-benzodiazepines. The anxiolytic and anticonvulsant effects of clobazam are associated with allosteric activation of the ligand-gated GABA(A) receptor. GABA(A) receptors are found extensively throughout the CNS, occurring synaptically and extrasynaptically. GABA(A) receptors are composed of five protein subunits, two copies of a single type of α subunit, two copies of one type of ß subunit and a γ subunit. This arrangement results in a diverse assortment of receptor subtypes. As benzodiazepine pharmacology is influenced by differences in affinity for particular GABA(A) subtypes, characterizing the selectivity of different benzodiazepines is a promising avenue for establishing appropriate use of these agents in neurological disorders. Molecular techniques have significantly advanced since the inception of clobazam as a clinical agent, adding to the understanding of the GABA(A) receptor, its subunits and benzodiazepine pharmacology. Transgenic mouse models have been particularly useful in this regard. Comparative studies between transgenic and wild-type mice have further defined relationships between GABA(A) receptor composition and drug effects. From such studies, we have learned that sedating and amnesic effects are mediated by the GABA(A) α(1) subunit, α(2) receptors mediate anxiolytic effects, α subunits are involved with anticonvulsant activity, α(5) may be implicated in learning and memory, and ß(3) subunit deficiency decreases GABA inhibition. Despite progress in determining the role of various subunits to specific benzodiazepine pharmacological actions, the precise mechanism of action of clobazam, and more importantly, how that mechanism of action translates into clinical consequences (i.e. efficacy, tolerability and safety) remain unknown. Testing clobazam and 1,4-benzodiazepines using a range of recombinant GABA(A) receptor subtypes would hopefully elucidate the subunits involved and strengthen our understanding of clobazam and its mechanism of action.


Asunto(s)
Benzodiazepinas/farmacología , Agonistas de Receptores de GABA-A/farmacología , Receptores de GABA-A/fisiología , Animales , Benzodiazepinas/uso terapéutico , Clobazam , Agonistas de Receptores de GABA-A/uso terapéutico , Antagonistas de Receptores de GABA-A/farmacología , Humanos , Modelos Neurológicos , Subunidades de Proteína/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA