Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Mol Biol ; 433(4): 166790, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33387531

RESUMEN

G protein coupled receptors signal through G proteins or arrestins. A long-standing mystery in the field is why vertebrates have two non-visual arrestins, arrestin-2 and arrestin-3. These isoforms are ~75% identical and 85% similar; each binds numerous receptors, and appear to have many redundant functions, as demonstrated by studies of knockout mice. We previously showed that arrestin-3 can be activated by inositol-hexakisphosphate (IP6). IP6 interacts with the receptor-binding surface of arrestin-3, induces arrestin-3 oligomerization, and this oligomer stabilizes the active conformation of arrestin-3. Here, we compared the impact of IP6 on oligomerization and conformational equilibrium of the highly homologous arrestin-2 and arrestin-3 and found that these two isoforms are regulated differently. In the presence of IP6, arrestin-2 forms "infinite" chains, where each promoter remains in the basal conformation. In contrast, full length and truncated arrestin-3 form trimers and higher-order oligomers in the presence of IP6; we showed previously that trimeric state induces arrestin-3 activation (Chen et al., 2017). Thus, in response to IP6, the two non-visual arrestins oligomerize in different ways in distinct conformations. We identified an insertion of eight residues that is conserved across arrestin-2 homologs, but absent in arrestin-3 that likely accounts for the differences in the IP6 effect. Because IP6 is ubiquitously present in cells, this suggests physiological consequences, including differences in arrestin-2/3 trafficking and JNK3 activation. The functional differences between two non-visual arrestins are in part determined by distinct modes of their oligomerization. The mode of oligomerization might regulate the function of other signaling proteins.


Asunto(s)
Aminoácidos/química , Arrestinas/química , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Arrestinas/metabolismo , Sitios de Unión , Humanos , Ácido Fítico/química , Unión Proteica , Isoformas de Proteínas , Soluciones , Análisis Espectral
2.
Handb Exp Pharmacol ; 219: 205-23, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24292832

RESUMEN

Mammals express four arrestin subtypes, three of which have been shown to self-associate. Cone photoreceptor-specific arrestin-4 is the only one that is a constitutive monomer. Visual arrestin-1 forms tetramers both in crystal and in solution, but the shape of its physiologically relevant solution tetramer is very different from that in the crystal. The biological role of the self-association of arrestin-1, expressed at very high levels in rod and cone photoreceptors, appears to be protective, reducing the concentration of cytotoxic monomers. The two nonvisual arrestin subtypes are highly homologous, and self-association of both is facilitated by IP6, yet they form dramatically different oligomers. Arrestin-2 apparently self-associates into "infinite" chains, very similar to those observed in IP6-soaked crystals, where IP6 connects the concave sides of the N- and C-domains of adjacent protomers. In contrast, arrestin-3 only forms dimers, in which IP6 likely connects the C-domains of two arrestin-3 molecules. Thus, each of the three self-associating arrestins does it in its own way, forming three different types of oligomers. The physiological role of the oligomerization of arrestin-1 and both nonvisual arrestins might be quite different, and in each case it remains to be definitively elucidated.


Asunto(s)
Arrestinas/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Cristalización , Humanos , Ácido Fítico/metabolismo , Multimerización de Proteína
3.
PLoS Biol ; 11(11): e1001714, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24260024

RESUMEN

Pentameric ligand-gated ion channels (pLGICs) are neurotransmitter-activated receptors that mediate fast synaptic transmission. In pLGICs, binding of agonist to the extracellular domain triggers a structural rearrangement that leads to the opening of an ion-conducting pore in the transmembrane domain and, in the continued presence of neurotransmitter, the channels desensitize (close). The flexible loops in each subunit that connect the extracellular binding domain (loops 2, 7, and 9) to the transmembrane channel domain (M2-M3 loop) are essential for coupling ligand binding to channel gating. Comparing the crystal structures of two bacterial pLGIC homologues, ELIC and the proton-activated GLIC, suggests channel gating is associated with rearrangements in these loops, but whether these motions accurately predict the motions in functional lipid-embedded pLGICs is unknown. Here, using site-directed spin labeling (SDSL) electron paramagnetic resonance (EPR) spectroscopy and functional GLIC channels reconstituted into liposomes, we examined if, and how far, the loops at the ECD/TMD gating interface move during proton-dependent gating transitions from the resting to desensitized state. Loop 9 moves ∼9 Šinward toward the channel lumen in response to proton-induced desensitization. Loop 9 motions were not observed when GLIC was in detergent micelles, suggesting detergent solubilization traps the protein in a nonactivatable state and lipids are required for functional gating transitions. Proton-induced desensitization immobilizes loop 2 with little change in position. Proton-induced motion of the M2-M3 loop was not observed, suggesting its conformation is nearly identical in closed and desensitized states. Our experimentally derived distance measurements of spin-labeled GLIC suggest ELIC is not a good model for the functional resting state of GLIC, and that the crystal structure of GLIC does not correspond to a desensitized state. These findings advance our understanding of the molecular mechanisms underlying pLGIC gating.


Asunto(s)
Proteínas Bacterianas/fisiología , Cianobacterias , Activación del Canal Iónico , Canales Iónicos Activados por Ligandos/fisiología , Sustitución de Aminoácidos , Animales , Proteínas Bacterianas/química , Células Cultivadas , Espectroscopía de Resonancia por Spin del Electrón , Canales Iónicos Activados por Ligandos/química , Liposomas/química , Potenciales de la Membrana , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Técnicas de Placa-Clamp , Estructura Cuaternaria de Proteína , Marcadores de Spin , Xenopus laevis
4.
Cell Signal ; 25(12): 2613-24, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24012956

RESUMEN

Arrestin-1 binds light-activated phosphorhodopsin and ensures timely signal shutoff. We show that high transgenic expression of an arrestin-1 mutant with enhanced rhodopsin binding and impaired oligomerization causes apoptotic rod death in mice. Dark rearing does not prevent mutant-induced cell death, ruling out the role of arrestin complexes with light-activated rhodopsin. Similar expression of WT arrestin-1 that robustly oligomerizes, which leads to only modest increase in the monomer concentration, does not affect rod survival. Moreover, WT arrestin-1 co-expressed with the mutant delays retinal degeneration. Thus, arrestin-1 mutant directly affects cell survival via binding partner(s) other than light-activated rhodopsin. Due to impaired self-association of the mutant its high expression dramatically increases the concentration of the monomer. The data suggest that monomeric arrestin-1 is cytotoxic and WT arrestin-1 protects rods by forming mixed oligomers with the mutant and/or competing with it for the binding to non-receptor partners. Thus, arrestin-1 self-association likely serves to keep low concentration of the toxic monomer. The reduction of the concentration of harmful monomer is an earlier unappreciated biological function of protein oligomerization.


Asunto(s)
Arrestina/genética , Arrestina/metabolismo , Mutación , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Animales , Arrestina/química , Muerte Celular , MAP Quinasa Quinasa 4/metabolismo , Ratones , Multimerización de Proteína , Células Fotorreceptoras Retinianas Bastones/citología , Rodopsina/metabolismo
5.
Proc Natl Acad Sci U S A ; 109(45): 18407-12, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-23091036

RESUMEN

Arrestin-1 (visual arrestin) binds to light-activated phosphorylated rhodopsin (P-Rh*) to terminate G-protein signaling. To map conformational changes upon binding to the receptor, pairs of spin labels were introduced in arrestin-1 and double electron-electron resonance was used to monitor interspin distance changes upon P-Rh* binding. The results indicate that the relative position of the N and C domains remains largely unchanged, contrary to expectations of a "clam-shell" model. A loop implicated in P-Rh* binding that connects ß-strands V and VI (the "finger loop," residues 67-79) moves toward the expected location of P-Rh* in the complex, but does not assume a fully extended conformation. A striking and unexpected movement of a loop containing residue 139 away from the adjacent finger loop is observed, which appears to facilitate P-Rh* binding. This change is accompanied by smaller movements of distal loops containing residues 157 and 344 at the tips of the N and C domains, which correspond to "plastic" regions of arrestin-1 that have distinct conformations in monomers of the crystal tetramer. Remarkably, the loops containing residues 139, 157, and 344 appear to have high flexibility in both free arrestin-1 and the P-Rh*complex.


Asunto(s)
Arrestina/química , Arrestina/metabolismo , Rodopsina/metabolismo , Cristalografía por Rayos X , Electrones , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilación , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Secundaria de Proteína , Eliminación de Secuencia , Soluciones , Coloración y Etiquetado , Temperatura
6.
PLoS One ; 6(12): e28723, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174878

RESUMEN

Arrestins are multifunctional signaling adaptors originally discovered as proteins that "arrest" G protein activation by G protein-coupled receptors (GPCRs). Recently GPCR complexes with arrestins have been proposed to activate G protein-independent signaling pathways. In particular, arrestin-dependent activation of extracellular signal-regulated kinase 1/2 (ERK1/2) has been demonstrated. Here we have performed in vitro binding assays with pure proteins to demonstrate for the first time that ERK2 directly binds free arrestin-2 and -3, as well as receptor-associated arrestins-1, -2, and -3. In addition, we showed that in COS-7 cells arrestin-2 and -3 association with ß(2)-adrenergic receptor (ß2AR) significantly enhanced ERK2 binding, but showed little effect on arrestin interactions with the upstream kinases c-Raf1 and MEK1. Arrestins exist in three conformational states: free, receptor-bound, and microtubule-associated. Using conformationally biased arrestin mutants we found that ERK2 preferentially binds two of these: the "constitutively inactive" arrestin-Δ7 mimicking microtubule-bound state and arrestin-3A, a mimic of the receptor-bound conformation. Both rescue arrestin-mediated ERK1/2/activation in arrestin-2/3 double knockout fibroblasts. We also found that arrestin-2-c-Raf1 interaction is enhanced by receptor binding, whereas arrestin-3-c-Raf1 interaction is not.


Asunto(s)
Arrestina/química , Arrestina/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Arrestinas/química , Arrestinas/metabolismo , Células COS , Bovinos , Chlorocebus aethiops , Embrión de Mamíferos/citología , Activación Enzimática , Fibroblastos/enzimología , Células HEK293 , Humanos , Ligandos , Ratones , Ratones Noqueados , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Receptores Adrenérgicos beta 2/metabolismo , Relación Estructura-Actividad , beta-Arrestinas
7.
Prog Retin Eye Res ; 30(6): 405-30, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21824527

RESUMEN

Visual arrestin-1 plays a key role in the rapid and reproducible shutoff of rhodopsin signaling. Its highly selective binding to light-activated phosphorylated rhodopsin is an integral part of the functional perfection of rod photoreceptors. Structure-function studies revealed key elements of the sophisticated molecular mechanism ensuring arrestin-1 selectivity and paved the way to the targeted manipulation of the arrestin-1 molecule to design mutants that can compensate for congenital defects in rhodopsin phosphorylation. Arrestin-1 self-association and light-dependent translocation in photoreceptor cells work together to keep a constant supply of active rhodopsin-binding arrestin-1 monomer in the outer segment. Recent discoveries of arrestin-1 interaction with other signaling proteins suggest that it is a much more versatile signaling regulator than previously thought, affecting the function of the synaptic terminals and rod survival. Elucidation of the fine molecular mechanisms of arrestin-1 interactions with rhodopsin and other binding partners is necessary for the comprehensive understanding of rod function and for devising novel molecular tools and therapeutic approaches to the treatment of visual disorders.


Asunto(s)
Arrestinas/metabolismo , Células Fotorreceptoras/metabolismo , Transducción de Señal/fisiología , Animales , Arrestinas/química , Humanos , Transporte de Proteínas/fisiología
8.
J Biol Chem ; 286(27): 24288-99, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21471193

RESUMEN

Arrestins bind active phosphorylated forms of G protein-coupled receptors, terminating G protein activation, orchestrating receptor trafficking, and redirecting signaling to alternative pathways. Visual arrestin-1 preferentially binds rhodopsin, whereas the two non-visual arrestins interact with hundreds of G protein-coupled receptor subtypes. Here we show that an extensive surface on the concave side of both arrestin-2 domains is involved in receptor binding. We also identified a small number of residues on the receptor binding surface of the N- and C-domains that largely determine the receptor specificity of arrestins. We show that alanine substitution of these residues blocks the binding of arrestin-1 to rhodopsin in vitro and of arrestin-2 and -3 to ß2-adrenergic, M2 muscarinic cholinergic, and D2 dopamine receptors in intact cells, suggesting that these elements critically contribute to the energy of the interaction. Thus, in contrast to arrestin-1, where direct phosphate binding is crucial, the interaction of non-visual arrestins with their cognate receptors depends to a lesser extent on phosphate binding and more on the binding to non-phosphorylated receptor elements.


Asunto(s)
Arrestina/química , Sitios de Unión/fisiología , Receptores Acoplados a Proteínas G/química , Sustitución de Aminoácidos , Animales , Arrestina/genética , Arrestina/metabolismo , Bovinos , Humanos , Mutación Missense , Mapeo Peptídico , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
9.
Biochemistry ; 50(12): 2235-42, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21288033

RESUMEN

Arrestin-1 binds light-activated phosphorhodopsin and ensures rapid signal termination. Its deficiency in humans and mice results in prolonged signaling and rod degeneration. However, most of the biochemical studies were performed on bovine arrestin-1, which was shown to self-associate forming dimers and tetramers, although only the monomer binds rhodopsin. It is unclear whether self-association is a property of arrestin-1 in all mammals or a specific feature of bovine protein. To address this issue, we compared self-association parameters of purified human and mouse arrestin-1 with those of its bovine counterpart using multiangle light scattering. We found that mouse and human arrestin-1 also robustly self-associate, existing in a monomer-dimer-tetramer equilibrium. Interestingly, the combination of dimerization and tetramerization constants in these three species is strikingly different. While tetramerization of bovine arrestin-1 is highly cooperative (K(D,dim)(4) > K(D,tet)), K(D,dim) ∼ K(D,tet) in the mouse form and K(D,dim) ≪ K(D,tet) in the human form. Importantly, in all three species at very high physiological concentrations of arrestin-1 in rod photoreceptors, most of it is predicted to exist in oligomeric form, with a relatively low concentration of the free monomer. Thus, it appears that maintenance of low levels of the active monomer is the biological role of arrestin-1 self-association.


Asunto(s)
Arrestinas/química , Arrestinas/metabolismo , Multimerización de Proteína , Animales , Arrestinas/genética , Bovinos , Humanos , Ratones , Modelos Moleculares , Mutación Puntual , Estructura Cuaternaria de Proteína , Conejos , Células Fotorreceptoras Retinianas Bastones/metabolismo
10.
Br J Pharmacol ; 162(3): 673-87, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20942818

RESUMEN

BACKGROUND AND SIGNIFICANCE: Although the functional effects of benzodiazepines (BZDs) on GABA(A) receptors have been well characterized, the structural mechanism by which these modulators alter activation of the receptor by GABA is still undefined. EXPERIMENTAL APPROACH: We used disulphide trapping between engineered cysteines to probe BZD-induced conformational changes within the γ2 subunit and at the α1/γ2 coupling interface (Loops 2, 7 and 9) of α1ß2γ2 GABA(A) receptors. KEY RESULTS: Crosslinking γ2 Loop 9 to γ2ß-strand 9 (via γ2 S195C/F203C and γ2 S187C/L206C) significantly decreased maximum potentiation by flurazepam, suggesting that modulation of GABA-induced current (I(GABA)) by flurazepam involves movements of γ2 Loop 9 relative to γ2ß-strand 9. In contrast, tethering γ2ß-strand 9 to the γ2 pre-M1 region (via γ2S202C/S230C) significantly enhanced potentiation by both flurazepam and zolpidem, indicating γ2S202C/S230C trapped the receptor in a more favourable conformation for positive modulation by BZDs. Intersubunit disulphide bonds formed at the α/γ coupling interface between α1 Loop 2 and γ2Loop 9 (α1D56C/γ2L198C) prevented flurazepam and zolpidem from efficiently modulating I(GABA) . Disulphide trapping α1 Loop 2 (α1D56C) to γ2ß-strand 1 (γ2P64C) decreased maximal I(GABA) as well as flurazepam potentiation. None of the disulphide bonds affected the ability of the negative modulator, 3-carbomethoxy-4-ethyl-6,7-dimethoxy-ß-carboline (DMCM), to inhibit I(GABA) . CONCLUSIONS AND IMPLICATIONS: Positive modulation of GABA(A) receptors by BZDs requires reorganization of the loops in the α1/γ2 coupling interface. BZD-induced movements at the α/γ coupling interface likely synergize with rearrangements induced by GABA binding at the ß/α subunit interfaces to enhance channel activation by GABA.


Asunto(s)
Benzodiazepinas/farmacología , Carbolinas/farmacología , Flurazepam/farmacología , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/farmacología , Animales , Benzodiazepinas/metabolismo , Carbolinas/metabolismo , Disulfuros/metabolismo , Ditiotreitol/metabolismo , Relación Dosis-Respuesta a Droga , Flurazepam/metabolismo , Peróxido de Hidrógeno/metabolismo , Mutagénesis Sitio-Dirigida , Oocitos , Técnicas de Placa-Clamp , Xenopus laevis , Ácido gamma-Aminobutírico/metabolismo
11.
J Mol Biol ; 395(1): 42-54, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19883657

RESUMEN

Arrestins rapidly bind phosphorylated activated forms of their cognate G protein-coupled receptors, thereby preventing G protein coupling and often switching signaling to other pathways. Amphipathic alpha-helix I (residues 100-111) has been implicated in receptor binding, but the mechanism of its action has not been determined yet. Here we show that several mutations in the helix itself and in adjacent hydrophobic residues in the body of the N-domain reduce arrestin1 binding to light-activated phosphorylated rhodopsin (P-Rh*). On the background of phosphorylation-independent mutants that bind with high affinity to both P-Rh* and light-activated unphosphorylated rhodopsin, these mutations reduce the stability of the arrestin complex with P-Rh*, but not with light-activated unphosphorylated rhodopsin. Using site-directed spin labeling, we found that the local structure around alpha-helix I changes upon binding to rhodopsin. However, the intramolecular distances between alpha-helix I and adjacent beta-strand I (or the rest of the N-domain), measured using double electron-electron resonance, do not change, ruling out relocation of the helix due to receptor binding. Collectively, these data demonstrate that alpha-helix I plays an indirect role in receptor binding, likely keeping beta-strand I, which carries several phosphate-binding residues, in a position favorable for its interaction with receptor-attached phosphates.


Asunto(s)
Arrestina/química , Arrestina/metabolismo , Rodopsina/metabolismo , Sustitución de Aminoácidos/genética , Animales , Bovinos , Interacciones Hidrofóbicas e Hidrofílicas , Mutación/genética , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína
12.
J Med Chem ; 51(22): 7243-52, 2008 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-18973287

RESUMEN

The sleep-aids zolpidem and eszopiclone exert their effects by binding to and modulating gamma-aminobutyric acid type-A receptors (GABA(A)Rs), but little is known about the structural requirements for their actions. We made 24 cysteine mutations in the benzodiazepine (BZD) binding site of alpha(1)beta(2)gamma(2) GABA(A)Rs and measured zolpidem, eszopiclone, and BZD-site antagonist binding. Mutations in gamma(2)loop D and alpha(1)loops A and B altered the affinity of all ligands tested, indicating that these loops are important for BZD pocket structural integrity. In contrast, gamma(2)loop E and alpha(1)loop C mutations differentially affected ligand affinity, suggesting that these loops are important for ligand selectivity. In agreement with our mutagenesis data, eszopiclone docking yielded a single model stabilized by several hydrogen bonds. Zolpidem docking yielded three equally populated orientations with few polar interactions, suggesting that unlike eszopiclone, zolpidem relies more on shape recognition of the binding pocket than on specific residue interactions and may explain why zolpidem is highly alpha(1)- and gamma(2)-subunit selective.


Asunto(s)
Compuestos de Azabiciclo/química , Compuestos de Azabiciclo/metabolismo , Piperazinas/química , Piperazinas/metabolismo , Piridinas/química , Piridinas/metabolismo , Receptores de GABA-A/metabolismo , Benzodiazepinas/química , Benzodiazepinas/metabolismo , Sitios de Unión , Línea Celular , Simulación por Computador , Cisteína/genética , Cisteína/metabolismo , Análisis Mutacional de ADN , Eszopiclona , Agonistas de Receptores de GABA-A , Humanos , Enlace de Hidrógeno , Ligandos , Modelos Químicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Receptores de GABA-A/química , Receptores de GABA-A/genética , Relación Estructura-Actividad , Zolpidem
13.
Structure ; 16(6): 924-34, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18547524

RESUMEN

Visual rod arrestin has the ability to self-associate at physiological concentrations. We previously demonstrated that only monomeric arrestin can bind the receptor and that the arrestin tetramer in solution differs from that in the crystal. We employed the Rosetta docking software to generate molecular models of the physiologically relevant solution tetramer based on the monomeric arrestin crystal structure. The resulting models were filtered using the Rosetta energy function, experimental intersubunit distances measured with DEER spectroscopy, and intersubunit contact sites identified by mutagenesis and site-directed spin labeling. This resulted in a unique model for subsequent evaluation. The validity of the model is strongly supported by model-directed crosslinking and targeted mutagenesis that yields arrestin variants deficient in self-association. The structure of the solution tetramer explains its inability to bind rhodopsin and paves the way for experimental studies of the physiological role of rod arrestin self-association.


Asunto(s)
Arrestina/química , Modelos Moleculares , Arrestina/genética , Disulfuros/química , Espectroscopía de Resonancia por Spin del Electrón , Conformación Proteica , Subunidades de Proteína/química , Eliminación de Secuencia , Programas Informáticos , Soluciones , Marcadores de Spin
14.
J Neurosci ; 28(13): 3490-9, 2008 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-18367615

RESUMEN

Many clinically important drugs target ligand-gated ion channels; however, the mechanisms by which these drugs modulate channel function remain elusive. Benzodiazepines (BZDs), anesthetics, and barbiturates exert their CNS actions by binding to GABA(A) receptors and modulating their function. The structural mechanisms by which BZD binding is transduced to potentiation or inhibition of GABA-induced current (I(GABA)) are essentially unknown. Here, we explored the role of the gamma(2)Q182-R197 region (Loop F/9) in the modulation of I(GABA) by positive (flurazepam, zolpidem) and negative [3-carbomethoxy-4-ethyl-6,7-dimethoxy-beta-carboline (DMCM)] BZD ligands. Each residue was individually mutated to cysteine, coexpressed with wild-type alpha(1) and beta(2) subunits in Xenopus oocytes, and analyzed using two-electrode voltage clamp. Individual mutations differentially affected BZD modulation of I(GABA). Mutations affecting positive modulation span the length of this region, whereas gamma(2)W183C at the beginning of Loop F was the only mutation that adversely affected DMCM inhibition. Radioligand binding experiments demonstrate that mutations in this region do not alter BZD binding, indicating that the observed changes in modulation result from changes in BZD efficacy. Flurazepam and zolpidem significantly slowed covalent modification of gamma(2)R197C, whereas DMCM, GABA, and the allosteric modulator pentobarbital had no effects, demonstrating that gamma(2)Loop F is a specific transducer of positive BZD modulator binding. Therefore, gamma(2)Loop F plays a key role in defining BZD efficacy and is part of the allosteric pathway allowing positive BZD modulator-induced structural changes at the BZD binding site to propagate through the protein to the channel domain.


Asunto(s)
Benzodiazepinas/farmacología , Activación del Canal Iónico/efectos de los fármacos , Receptores de GABA-A/química , Receptores de GABA-A/efectos de los fármacos , Regulación Alostérica , Animales , Sitios de Unión/efectos de los fármacos , Carbolinas/farmacología , Convulsivantes/farmacología , Cisteína/genética , Relación Dosis-Respuesta a Droga , Indicadores y Reactivos/farmacología , Larva , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Mesilatos/farmacología , Microinyecciones/métodos , Mutagénesis Sitio-Dirigida/métodos , Oocitos , Técnicas de Placa-Clamp , Unión Proteica/efectos de los fármacos , Ratas , Receptores de GABA-A/genética , Relación Estructura-Actividad , Xenopus , Ácido gamma-Aminobutírico/farmacología
15.
Biochemistry ; 47(3): 1070-5, 2008 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-18161994

RESUMEN

The robust cooperative formation of rod arrestin tetramers has been well-established, whereas the ability of other members of the arrestin family to self-associate remains controversial. Here, we used purified arrestins and multi-angle light scattering to quantitatively compare the propensity of the four mammalian arrestin subtypes to self-associate. Both non-visual and cone arrestins only form oligomers at very high non-physiological concentrations. However, inositol hexakisphosphate (IP6), a fairly abundant form of inositol in the cytoplasm, greatly facilitates self-association of arrestin2. Arrestin2 self-association equilibrium constants in the presence of 100 microM IP6 suggest that an appreciable proportion could exist in an oligomeric state but only in intracellular compartments where its concentration is 5-10-fold higher than average. In contrast to arrestin2, IP6 inhibits self-association of rod arrestin, indicating that the structure of these two tetramers in solution is likely different.


Asunto(s)
Arrestina/química , Arrestinas/química , Ácido Fítico/química , Animales , Arrestina/metabolismo , Arrestinas/metabolismo , Bovinos , Cromatografía en Gel , Dimerización , Humanos , Rayos Láser , Luz , Peso Molecular , Ácido Fítico/metabolismo , Ácido Fítico/farmacología , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Dispersión de Radiación , beta-Arrestinas
16.
Invest Ophthalmol Vis Sci ; 48(5): 1968-75, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17460248

RESUMEN

PURPOSE: To examine the biochemical characteristics of rod and cone arrestin with respect to their ability to quench the activity of light-activated rhodopsin in transgenic mice. METHODS: The mouse rod opsin promoter was used to drive expression of mouse cone arrestin in rod photoreceptor cells of rod arrestin knockout (arr1-/-) mice. Suction electrode recordings from single rods were performed to investigate cone arrestin's ability to quench the catalytic activity of light-activated rhodopsin. In addition, the ability of cone arrestin to prevent light-induced retinal damage caused by prolonged activation of the phototransduction cascade was assessed. RESULTS: Two independent lines of transgenic mice were obtained that expressed cone arrestin in rod photoreceptors, and each was bred into the arr1-/- background. Flash responses measured by suction electrode recordings showed that cone arrestin reduced signaling from photolyzed rhodopsin but was unable to quench its activity completely. Consistent with this observation, expression of mouse cone arrestin conferred dose-dependent protection against photoreceptor cell death caused by low light exposure to arr1-/- retinas, but did not appear to be as effective as rod arrestin. CONCLUSIONS: Cone arrestin can partially substitute for rod arrestin in arr1-/- rods, offering a degree of protection from light-induced damage and increasing the extent of rhodopsin deactivation in response to flashes of light. Although earlier work has shown that rod arrestin can bind and deactivate cone pigments efficiently, the results suggest that cone arrestin binds light-activated, phosphorylated rhodopsin less efficiently than does rod arrestin in vivo. These results suggest that the structural requirements for high-affinity binding are fundamentally distinct for rod and cone arrestins.


Asunto(s)
Arrestinas/fisiología , Células Fotorreceptoras Retinianas Conos/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Animales , Western Blotting , Electroforesis en Gel de Poliacrilamida , Electrofisiología , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica , Luz , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Estimulación Luminosa , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Células Fotorreceptoras Retinianas Bastones/efectos de la radiación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodopsina/metabolismo , Visión Ocular/fisiología , beta-Arrestina 1 , beta-Arrestinas
17.
EMBO J ; 26(6): 1726-36, 2007 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-17332750

RESUMEN

A distinguishing feature of rod arrestin is its ability to form oligomers at physiological concentrations. Using visible light scattering, we show that rod arrestin forms tetramers in a cooperative manner in solution. To investigate the structure of the tetramer, a nitroxide side chain (R1) was introduced at 18 different positions. The effects of R1 on oligomer formation, EPR spectra, and inter-spin distance measurements all show that the structures of the solution and crystal tetramers are different. Inter-subunit distance measurements revealed that only arrestin monomer binds to light-activated phosphorhodopsin, whereas both monomer and tetramer bind microtubules, which may serve as a default arrestin partner in dark-adapted photoreceptors. Thus, the tetramer likely serves as a 'storage' form of arrestin, increasing the arrestin-binding capacity of microtubules while readily dissociating to supply active monomer when it is needed to quench rhodopsin signaling.


Asunto(s)
Arrestina/química , Arrestina/fisiología , Modelos Moleculares , Oligodesoxirribonucleótidos/química , Animales , Arrestina/genética , Bovinos , Cristalización , Humanos , Luz , Espectroscopía de Resonancia Magnética/métodos , Microtúbulos/metabolismo , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Rodopsina/metabolismo , Dispersión de Radiación
18.
J Mol Biol ; 368(2): 375-87, 2007 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-17359998

RESUMEN

Arrestins regulate the activity and subcellular localization of G protein-coupled receptors and other signaling molecules. Here, we demonstrate that arrestins bind microtubules (MTs) in vitro and in vivo. The MT-binding site on arrestins overlaps significantly with the receptor-binding site, but the conformations of MT-bound and receptor-bound arrestin are different. Arrestins recruit ERK1/2 and the E3 ubiquitin ligase Mdm2 to MTs in cells, similar to the arrestin-dependent mobilization of these proteins to the receptor. Arrestin-mediated sequestration of ERK to MTs reduces the level of ERK activation. In contrast, recruitment of Mdm2 to MTs by arrestin channels Mdm2 activity toward cytoskeleton-associated proteins, increasing their ubiquitination dramatically. The mobilization of signaling molecules to MTs is a novel biological function of arrestin proteins.


Asunto(s)
Arrestina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal , Animales , Arrestina/química , Sitios de Unión , Células COS , Línea Celular , Supervivencia Celular , Chlorocebus aethiops , Dimerización , Humanos , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Tubulina (Proteína)/metabolismo
19.
Proc Natl Acad Sci U S A ; 104(9): 3125-8, 2007 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-17360618

RESUMEN

Arrestins (Arrs) are ubiquitous regulators of the most numerous family of signaling proteins, G protein-coupled receptors. Two models of the Arr-receptor interaction have been proposed: the binding of one Arr to an individual receptor or to two receptors in a dimer. To determine the binding stoichiometry in vivo, we used rod photoreceptors where rhodopsin (Rh) and Arr are expressed at comparably high levels and where Arr localization in the light is determined by its binding to activated Rh. Genetic manipulation of the expression of both proteins shows that the maximum amount of Arr that moves to the Rh-containing compartment exceeds 80%, but not 100%, of the molar amount of Rh present. In vitro experiments with purified proteins confirm that Arr "saturates" Rh at a 1:1 ratio. Thus, a single Rh molecule is necessary and sufficient to bind Arr. Remarkable structural conservation among receptors and Arrs strongly suggests that all Arr subtypes bind individual molecules of their cognate receptors.


Asunto(s)
Arrestina/metabolismo , Modelos Moleculares , Unión Proteica , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/metabolismo , Animales , Arrestina/genética , Dimerización , Inmunohistoquímica , Ratones , Ratones Noqueados , Transporte de Proteínas , Rodopsina/genética
20.
J Mol Biol ; 364(5): 955-63, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17054984

RESUMEN

Arrestins serve as multi-functional regulators of G-protein coupled receptors, interacting with hundreds of different receptor subtypes and a variety of other signaling proteins. Here we identify calmodulin as a novel arrestin interaction partner using three independent methods in vitro and in cells. Arrestin preferentially binds calcium-loaded calmodulin with a Kd value of approximately 7 microM, which is within range of endogenous calmodulin concentrations. The calmodulin binding site is localized on the concave side of the C-domain and a loop in the center of the arrestin molecule, significantly overlapping with receptor and microtubule-binding sites. Using purified proteins, we found that arrestins sequester calmodulin, preventing its binding to microtubules. Nanomolar affinity of arrestins for their cognate receptors makes calmodulin an ineffective competitor for arrestin binding at relatively high receptor concentrations. The arrestin-calmodulin interaction likely regulates the localization of both proteins and their availability for other interaction partners.


Asunto(s)
Arrestinas/metabolismo , Calmodulina/metabolismo , Arrestinas/genética , Sitios de Unión , Calcio/metabolismo , Calmodulina/genética , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Inmunoprecipitación , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...