Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Ann Oncol ; 27(8): 1579-85, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27457310

RESUMEN

BACKGROUND: Anti-angiogenic therapies are effective in metastatic renal cell carcinoma (mRCC), but resistance is inevitable. A dual-inhibition strategy focused on hypoxia-inducible factor (HIF) is hypothesized to be active in this refractory setting. CRLX101 is an investigational camptothecin-containing nanoparticle-drug conjugate (NDC), which durably inhibits HIF1α and HIF2α in preclinical models and in gastric cancer patients. Synergy was observed in the preclinical setting when combining this NDC and anti-angiogenic agents, including bevacizumab. PATIENTS AND METHODS: Patients with refractory mRCC were treated every 2 weeks with bevacizumab (10 mg/kg) and escalating doses of CRLX101 (12, 15 mg/m(2)) in a 3 + 3 phase I design. An expansion cohort of 10 patients was treated at the recommended phase II dose (RP2D). Patients were treated until progressive disease or prohibitive toxicity. Adverse events (AEs) were assessed using CTCAE v4.0 and clinical outcome using RECIST v1.1. RESULTS: Twenty-two patients were response-evaluable in an investigator-initiated trial at two academic medical centers. RCC histologies included clear cell (n = 12), papillary (n = 5), chromophobe (n = 2), and unclassified (n = 3). Patients received a median of two prior therapies, with at least one prior vascular endothelial tyrosine kinase inhibitor therapy (VEGF-TKI). No dose-limiting toxicities were observed. Grade ≥3 AEs related to CRLX101 included non-infectious cystitis (5 events), fatigue (3 events), anemia (2 events), diarrhea (2 events), dizziness (2 events), and 7 other individual events. Five of 22 patients (23%) achieved partial responses, including 3 of 12 patients with clear cell histology and 2 of 10 patients (20%) with non-clear cell histology. Twelve of 22 patients (55%) achieved progression-free survival (PFS) of >4 months. CONCLUSIONS: CRLX101 combined with bevacizumab is safe in mRCC. This combination fulfilled the protocol's predefined threshold for further examination with responses and prolonged PFS in a heavily pretreated population. A randomized phase II clinical trial in mRCC of this combination is ongoing.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Bevacizumab/administración & dosificación , Camptotecina/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Ciclodextrinas/administración & dosificación , Adulto , Anciano , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/efectos adversos , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/química , Bevacizumab/efectos adversos , Camptotecina/efectos adversos , Carcinoma de Células Renales/patología , Ciclodextrinas/efectos adversos , Supervivencia sin Enfermedad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/clasificación , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nanopartículas/administración & dosificación , Nanopartículas/química , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos
2.
Vision Res ; 38(10): 1443-54, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9667010

RESUMEN

Glutamate is the major excitatory neurotransmitter in the vertebrate retina. Native glutamate transporters have been well characterized in several retinal neurons, particularly from the salamander retina. We have cloned five distinct glutamate transporters from the salamander retina and examined their localization and functional properties: sEAAT1, sEEAAT2A, sEAAT2B, sEAAT5A and sEAAT5B. sEAAT1 is a homologue of the glutamate transporter EAAT1 (GLAST), sEAAT2A and sEAAT2B are homologues of EAAT2 (GLT-1) and sEAAT5A and sEAAT5B are homologues of the recently cloned human retinal glutamate transporter EAAT5. Localization was determined by immunocytochemical techniques using antibodies directed at portions of the highly divergent carboxy terminal. Glutamate transporters were found in glial, photoreceptor, bipolar, amacrine and ganglion cells. The pharmacology and ionic dependence were determined by two-electrode voltage clamp recordings from Xenopus laevis oocytes which had previously been injected with one of the glutamate transporter mRNAs. Each of the transporters behaved in a manner consistent with a glutamate transporter and there were some distinguishing characteristics which make it possible to link the function in native cells with the behavior of the cloned transporters in this study.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/análisis , Glutamatos/análisis , Retina/química , Secuencia de Aminoácidos , Sistema de Transporte de Aminoácidos X-AG , Animales , Transporte Biológico Activo , Proteínas Portadoras/análisis , Proteínas Portadoras/química , Clonación Molecular , Aminoácidos Excitadores/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Glutamatos/metabolismo , Potenciales de la Membrana , Oocitos/fisiología , Urodelos , Xenopus
3.
J Neurosci ; 13(1): 402-11, 1993 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8093715

RESUMEN

L-Glutamate elicits an inwardly rectifying current at hyperpolarized potentials in isolated retinal cones of the tiger salamander, as measured under whole-cell patch clamp. Evidence presented in this article supports the notion that cones possess a high-affinity glutamate transporter. This glutamate-elicited current shows no desensitization over a period of several minutes, and has an affinity (Km) of 10 microM. The inward current is mimicked by the amino acids L-aspartate, D-aspartate, L-cysteate, and to a lesser extent D-glutamate. It is neither blocked by the glutamate receptor antagonists kynurenic acid (1 mM), 6-cyano-7-nitroquinoxaline-2,3-dione (100 microM), or 2-amino-5-phosphonovalerate (100 microM), nor elicited by the glutamate receptor agonists (100 microM each) kainate, quisqualate, NMDA, or 2-amino-4-phosphonobutyrate. The glutamate-elicited current was reduced by the glutamate transport blockers dihydrokainate (DHKA), DL-threo-beta-hydroxyaspartate (beta HA), and L-trans-pyrrolidine-2,4-dicarboxylic acid. When glutamate was present on both sides of the membrane, the blockers reduced both uptake and release; the blocker-sensitive current as a function of membrane potential represents the transport current-voltage relation (I-V), and the reversal potential of the I-V represents the transporter equilibrium potential. This potential was a function of the equilibrium potential for glutamate. DHKA and beta HA depolarized horizontal cells in a retinal slice, and abolished their light responses, suggesting that in the absence of glutamate transport, glutamate concentrations in the cleft rise to a level that saturates the postsynaptic receptors. The high capacity of the cone glutamate transporter is well suited for the rapid removal of glutamate from the synaptic cleft required for the signaling of a light onset to postsynaptic cells.


Asunto(s)
Ambystoma/metabolismo , Glicoproteínas/metabolismo , Células Fotorreceptoras/metabolismo , Sistema de Transporte de Aminoácidos X-AG , Aminoácidos/farmacología , Animales , Transporte Biológico , Electrofisiología , Antagonistas de Aminoácidos Excitadores , Glutamatos/farmacología , Ácido Glutámico , Glicoproteínas/antagonistas & inhibidores , Técnicas In Vitro , Concentración Osmolar , Células Fotorreceptoras/efectos de los fármacos , Células Fotorreceptoras/fisiología , Receptores de Glutamato/fisiología , Retina/citología , Retina/metabolismo , Retina/fisiología , Transmisión Sináptica/efectos de los fármacos
4.
Vis Neurosci ; 14(1): 13-8, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9057264

RESUMEN

AMPA and NMDA type glutamate receptors were studied in isolated catfish cone horizontal cells using the whole-cell and outside-out patch-recording techniques. In whole-cell recordings, cyclothiazide (CTZ) enhanced the peak current in response to glutamate (in the presence of NMDA receptor antagonists). In patch recordings, currents evoked by rapid and maintained applications of glutamate desensitized with a time constant of one millisecond. CTZ blocked this rapid desensitization. Recovery from desensitization of the AMPA receptors was rapid, having a time constant of 8.65 ms. In contrast, the whole-cell and patch responses to applications of NMDA were much smaller than the AMPA receptor responses and did not desensitize. The relative contribution of these two receptor subtypes depends critically on the condition of the synapse; if glutamate levels are tonically present, the NMDA receptors contribute significantly to the postsynaptic response. If glutamate levels fall rapidly following the release of a single quantum of glutamate, then AMPA receptor currents will dominate the postsynaptic response.


Asunto(s)
Receptores AMPA/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Transmisión Sináptica/fisiología , Animales , Antihipertensivos/farmacología , Benzotiadiazinas/farmacología , Bagres , Relación Dosis-Respuesta a Droga , Ácido Glutámico/farmacología , Técnicas In Vitro , Potenciales de la Membrana , Técnicas de Placa-Clamp , Receptores AMPA/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
5.
Proc Natl Acad Sci U S A ; 93(9): 4153-8, 1996 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-8633032

RESUMEN

Application of L-glutamate to retinal glial (Müller) cells results in an inwardly rectifying current due to the net influx of one positive charge per molecule of glutamate transported into the cell. However, at positive potentials an outward current can be elicited by glutamate. This outward current is eliminated by removal of external chloride ions. Substitution of external chloride with the anions thiocyanate, perchlorate, nitrate, and iodide, which are known to be more permeant at other chloride channels, results in a considerably larger glutamate-elicited outward current at positive potentials. The large outward current in external nitrate has the same ionic dependence, apparent affinity for L-glutamate, and pharmacology as the glutamate transporter previously reported to exist in these cells. Varying the concentration of external nitrate shifts the reversal potential in a manner consistent with a conductance permeable to nitrate. Together, these results suggest that the glutamate transporter in retinal glial cells is associated with an anionic conductance. This anionic conductance may be important for preventing a reduction in the rate of transport due the depolarization that would otherwise occur as a result of electrogenic glutamate uptake.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Canales de Cloruro/fisiología , Cloruros/metabolismo , Ácido Glutámico/farmacología , Neuroglía/fisiología , Retina/citología , Ambystoma , Sistema de Transporte de Aminoácidos X-AG , Animales , Aniones , Canales de Cloruro/efectos de los fármacos , Gluconatos/farmacología , Ácido Glutámico/metabolismo , Técnicas In Vitro , Yoduros/farmacología , Potenciales de la Membrana/efectos de los fármacos , Neuroglía/efectos de los fármacos , Nitratos/farmacología , Técnicas de Placa-Clamp , Percloratos/farmacología , Tiocianatos/farmacología
6.
J Neurosci ; 7(11): 3512-24, 1987 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3681403

RESUMEN

We investigated the ionic interactions responsible for the characteristic nonrepetitive spike activity of amacrine cells. First we measured 4 pharmacologically separable ionic components: a voltage-gated, transient inward sodium current, a voltage-gated, sustained inward calcium current, a calcium-gated, sustained outward potassium current, and a voltage-gated, transient outward potassium current. The measurements provided the time course and magnitudes of the underlying conductances as functions of voltage. Each current was simulated following conventional Hodgkin-Huxley theory. A composite of the simulated currents was analytically reassembled to generate an approximation of the voltage response to a current step. By artificially varying the magnitude and kinetics of the different conductances in the simulation, we determined the range of values that supported the nonrepetitive spike-like response. Amacrine cells tend to remain refractory following an initial spike because (1) the entire activation range for potassium is located at positive potentials with respect to sodium inactivation, so sodium inactivation is never fully extinguished, and (2) the fully activated sodium conductance is of insufficient magnitude to subsequently reach threshold, given this residual inactivation. Shifting the sodium inactivation range by 10 mV, or increasing sodium conductance by 5 times, leads to a more repetitive form of activity. Changes in the magnitude, time course, or activation range of the potassium conductance cannot alter these conditions.


Asunto(s)
Retina/fisiología , Urodelos/fisiología , Animales , Calcio/metabolismo , Conductividad Eléctrica , Electrofisiología , Técnicas In Vitro , Matemática , Potasio/metabolismo , Valores de Referencia
7.
Proc Natl Acad Sci U S A ; 98(26): 15324-9, 2001 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-11752470

RESUMEN

Excitatory amino acid transporters (EAATs) buffer and remove synaptically released L-glutamate and maintain its concentrations below neurotoxic levels. EAATs also mediate a thermodynamically uncoupled substrate-gated anion conductance that may modulate cell excitability. Here, we demonstrate that modification of a cysteine substituted within a C-terminal domain of EAAT1 abolishes transport in both the forward and reverse directions without affecting activation of the anion conductance. EC(50)s for L-glutamate and sodium are significantly lower after modification, consistent with kinetic models of the transport cycle that link anion channel gating to an early step in substrate translocation. Also, decreasing the pH from 7.5 to 6.5 decreases the EC(50) for L-glutamate to activate the anion conductance, without affecting the EC(50) for the entire transport cycle. These findings demonstrate for the first time a structural separation of transport and the uncoupled anion flux. Moreover, they shed light on some controversial aspects of the EAAT transport cycle, including the kinetics of proton binding and anion conductance activation.


Asunto(s)
Transportador 1 de Aminoácidos Excitadores/metabolismo , Compuestos de Sulfhidrilo/química , Aniones , Transporte Biológico , Transportador 1 de Aminoácidos Excitadores/química , Humanos , Activación del Canal Iónico , Cinética , Conformación Proteica
8.
J Neurochem ; 77(2): 550-7, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11299317

RESUMEN

The glutamate analog (+/-) threo-3-methylglutamate (T3MG) has recently been reported to inhibit the EAAT2 but not EAAT1 subtype of high-affinity, Na(+)-dependent excitatory amino acid transporter (EAAT). We have examined the effects of T3MG on glutamate-elicited currents mediated by EAATs 1-4 expressed in Xenopus oocytes and on the transport of radiolabeled substrate in mammalian cell lines expressing EAATs 1-3. T3MG was found to be an inhibitor of EAAT2 and EAAT4 but a weak inhibitor of EAAT1 and EAAT3. T3MG competitively inhibited uptake of D-[(3)H]-aspartate into both cortical and cerebellar synaptosomes with a similar potency, consistent with its inhibitory activity on the cloned EAAT2 and EAAT4 subtypes. In addition, T3MG produced substrate-like currents in oocytes expressing EAAT4 but not EAAT2. However, T3MG was unable to elicit heteroexchange of preloaded D-[(3)H]-aspartate in cerebellar synaptosomes, inconsistent with the behavior of a substrate inhibitor. Finally, T3MG acts as a poor ionotropic glutamate receptor agonist in cultured hippocampal neurons: concentrations greater than 100 microM T3MG were required to elicit significant NMDA receptor-mediated currents. Thus, T3MG represents a pharmacological tool for the study of not only the predominant EAAT2 subtype but also the EAAT4 subtype highly expressed in cerebellum.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Proteínas Portadoras/antagonistas & inhibidores , Ácido Glutámico/farmacología , Inhibidores de la Captación de Neurotransmisores/farmacología , Receptores de Neurotransmisores/antagonistas & inhibidores , Simportadores , 2-Amino-5-fosfonovalerato/farmacología , Transportadoras de Casetes de Unión a ATP/genética , Potenciales de Acción/efectos de los fármacos , Sistema de Transporte de Aminoácidos X-AG , Animales , Ácido Aspártico/metabolismo , Proteínas Portadoras/genética , Línea Celular , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Perros , Transportador 1 de Aminoácidos Excitadores , Transportador 2 de Aminoácidos Excitadores , Transportador 3 de Aminoácidos Excitadores , Transportador 4 de Aminoácidos Excitadores , Proteínas de Transporte de Glutamato en la Membrana Plasmática , Ácido Glutámico/análogos & derivados , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Riñón/citología , Cinética , Oocitos , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/genética , Receptores de Neurotransmisores/genética , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Sinaptosomas/metabolismo , Xenopus laevis
9.
J Neurosci ; 18(2): 698-712, 1998 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9425012

RESUMEN

The rapid re-uptake of extracellular glutamate mediated by a family of high-affinity glutamate transporter proteins is essential to continued glutamatergic signaling and neuronal viability, but the contributions of individual transporter subtypes toward cellular physiology are poorly understood. Because the physiology of glutamate transport in the salamander retina has been well described, we have examined the expression and function of glutamate transporter subtypes in this preparation. cDNAs encoding five distinct salamander excitatory amino acid transporter (sEAAT) subtypes were isolated, and their molecular properties and distributions of expression were compared. We report evidence that at least four distinct sEAAT subtypes are expressed in glial (Müller) cells. In addition, four of the five transporter subtypes are localized in neurons throughout the retina. The brightest immunostaining was seen in the synaptic regions of the inner and outer plexiform layers and in the outer nuclear layer. Using electrophysiological measurements in the Xenopus oocyte expression system, we also examined the pharmacology and ionic dependence of the four expressing transporter subtypes that make it possible to distinguish, on the basis of functional behavior, among the various subtypes. Although no simple correlation between transporter subtype and retinal cell physiology can be made, the diverse population of sEAAT transporter subtypes with unique localization and functional properties indicates that glutamate transporters play a wide variety of roles in retinal function and are likely to underlie both the uptake of glutamate by Müller cells and the glutamate-elicited chloride conductance involved in signal transduction by photoreceptors and bipolar cells.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Proteínas Portadoras/fisiología , Proteínas del Ojo/fisiología , Receptores de Glutamato/fisiología , Receptores de Neurotransmisores/fisiología , Retina/fisiología , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Ambystoma , Secuencia de Aminoácidos , Sistema de Transporte de Aminoácidos X-AG , Animales , Secuencia de Bases , Proteínas Portadoras/química , Proteínas Portadoras/genética , Canales de Cloruro/metabolismo , Transportador 2 de Aminoácidos Excitadores , Proteínas del Ojo/química , Proteínas del Ojo/genética , Expresión Génica , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/metabolismo , Receptores de Glutamato/química , Receptores de Glutamato/genética , Receptores de Neurotransmisores/química , Receptores de Neurotransmisores/genética , Retina/química , Xenopus
10.
Proc Natl Acad Sci U S A ; 94(8): 4155-60, 1997 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9108121

RESUMEN

Although a glutamate-gated chloride conductance with the properties of a sodium-dependent glutamate transporter has been described in vertebrate retinal photoreceptors and bipolar cells, the molecular species underlying this conductance has not yet been identified. We now report the cloning and functional characterization of a human excitatory amino acid transporter, EAAT5, expressed primarily in retina. Although EAAT5 shares the structural homologies of the EAAT gene family, one novel feature of the EAAT5 sequence is a carboxy-terminal motif identified previously in N-methyl-D-aspartate receptors and potassium channels and shown to confer interactions with a family of synaptic proteins that promote ion channel clustering. Functional properties of EAAT5 were examined in the Xenopus oocyte expression system by measuring radiolabeled glutamate flux and two-electrode voltage clamp recording. EAAT5-mediated L-glutamate uptake is sodium- and voltage-dependent and chloride-independent. Transporter currents elicited by glutamate are also sodium- and voltage-dependent, but ion substitution experiments suggest that this current is largely carried by chloride ions. These properties of EAAT5 are similar to the glutamate-elicited chloride conductances previously described in retinal neurons, suggesting that the EAAT5-associated chloride conductance may participate in visual processing.


Asunto(s)
Sistemas de Transporte de Aminoácidos , Proteínas Portadoras/genética , Cloruros/metabolismo , Células Fotorreceptoras , Retina/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/aislamiento & purificación , Proteínas Portadoras/metabolismo , Clonación Molecular , Transportador 5 de Aminoácidos Excitadores , Humanos , Transporte Iónico , Datos de Secuencia Molecular , Urodelos
11.
Vis Neurosci ; 1(3): 317-29, 1988.
Artículo en Inglés | MEDLINE | ID: mdl-2856477

RESUMEN

The neural circuitry underlying movement detection was inferred from studies of amacrine cells under whole-cell patch clamp in retinal slices. Cells were identified by Lucifer yellow staining. Synaptic inputs were driven by "puffing" transmitter substances at the dendrites of presynaptic cells. Spatial sensitivity profiles for amacrine cells were measured by puffing transmitter substances along the lateral spread of their processes. Synaptic pathways were separated and identified with appropriate pre- and postsynaptic pharmacological blocking agents. Two distinct amacrine cell types were found: one with narrow spread of processes that received sustained excitatory synaptic current, the other with very wide spread of processes that received transient excitatory synaptic currents. The transient currents found only in the wide-field amacrine cell were formed presynaptically at GABAB receptors. They could be blocked with baclofen, a GABAB agonist, and their time course was extended by AVA, a GABAB antagonist. Baclofen and AVA had no direct affect upon the wide-field amacrine cell, but picrotoxin blocked a separate, direct GABA input to this cell. The narrow-field amacrine cell was shown to be GABAergic by counterstaining with anti-GABA antiserum after it was filled with Lucifer yellow. Its narrow, spatial profile and sustained synaptic input are properties that closely match those of the GABAergic antagonistic signal that forms transient activity (described above), suggesting that the narrow-field amacrine cell itself is the source of the GABAergic interaction mediating transient activity in the inner plexiform layer (IPL). Other work has shown a GABAB sensitivity at some bipolar terminals, suggesting a population of bipolars as the probable site of interaction mediating transient action. The results suggest that two local populations of amacrine cell types (sustained and transient) interact with the two populations of bipolar cell types (transient forming and nontransient forming). These interactions underlie the formation of the change-detecting subunits. We suggest that local populations of these subunits converge to form the receptive fields of movement-detecting ganglion cells.


Asunto(s)
Percepción de Movimiento/fisiología , Retina/fisiología , Vías Visuales/fisiología , Animales , Baclofeno , Dendritas/fisiología , Electrofisiología , Isoquinolinas , Neuronas/fisiología , Picrotoxina , Receptores de GABA-A/fisiología , Retina/citología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Urodelos
12.
J Biol Chem ; 275(41): 32174-81, 2000 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-10906333

RESUMEN

We have identified a splice variant encoding only the extracellular ligand-binding domain of the gamma-aminobutyric acid B (GABA(B)) receptor subunit GABA(B(1a)). This isoform, which we have named GABA(B(1e)), is detected in both rats and humans. While GABA(B(1e)) is a minor component of the total pool of GABA(B(1)) transcripts detected in the central nervous system, it is the primary isoform found in all peripheral tissues examined. When expressed in a heterologous system, the truncated receptor is both secreted and membrane associated. However, GABA(B(1e)) lacks the ability to bind the radiolabeled antagonist [(3)H]CGP 54626A, activate G-protein coupled inwardly rectifying potassium channels, or inhibit forskolin-induced cAMP production when it is expressed alone or together with GABA(B(2)). Interestingly, when co-expressed with GABA(B(2)), not only does the truncated receptor heterodimerize with GABA(B(2)), the association is of sufficient avidity to disrupt the normal GABA(B(1a))/GABA(B(2)) association. Despite this strong interaction, GABA(B(1e)) fails to disrupt G-protein coupled inwardly rectifying potassium activation by the full-length heterodimer pair of GABA(B(1a))/GABA(B(2)).


Asunto(s)
Empalme Alternativo/genética , Canales de Potasio de Rectificación Interna , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Eliminación de Secuencia/genética , Animales , Línea Celular , Colforsina/farmacología , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Dimerización , Antagonistas del GABA/metabolismo , Antagonistas del GABA/farmacología , Antagonistas de Receptores de GABA-B , Proteínas de Unión al GTP/metabolismo , Humanos , Datos de Secuencia Molecular , Oocitos , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/farmacología , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Pruebas de Precipitina , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Receptores de GABA-B/química , Proteínas Recombinantes/metabolismo , Transfección , Xenopus laevis
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda