Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ChemMedChem ; 16(1): 164-178, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-32700391

RESUMO

G protein-coupled receptors (GPCRs) can be used to shuttle peptide-drug conjugates into cells. But, for efficient therapy, a high concentration of cargo needs to be delivered. To explore this, we studied the pharmacologically interesting neuropeptide Y1 receptor (Y1 R) in one recombinant and three oncogenic cell systems that endogenously express the receptor. We demonstrate that recycled receptors behave identically to newly synthesized receptors with respect to ligand binding and internalization pathways. Depending on the cell system, biosynthesis, recycling efficiency, and peptide uptake differ partially, but shuttling was efficient in all systems. However, by comparing continuous application of the ligand for four hours to four cycles of internalization and recycling in between, a significantly higher amount of peptide uptake was achieved in the pulsed application (150-250 % to 300-400 %). Accordingly, in this well-suited drug shuttle system pulsed application is superior under all investigated conditions and should be considered for innovative, targeted drug delivery in general.


Assuntos
Neuropeptídeo Y/química , Preparações Farmacêuticas/química , Receptores de Neuropeptídeo Y/metabolismo , Arrestina/química , Arrestina/metabolismo , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Corantes Fluorescentes/química , Células HEK293 , Humanos , Ligantes , Microscopia Confocal , Neuropeptídeo Y/metabolismo , Ligação Proteica , Receptores de Neuropeptídeo Y/química
2.
Nat Chem Biol ; 16(12): 1343-1350, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32778842

RESUMO

The adhesion G-protein-coupled receptor (GPCR) latrophilin 3 (ADGRL3) has been associated with increased risk of attention deficit hyperactivity disorder (ADHD) and substance use in human genetic studies. Knockdown in multiple species leads to hyperlocomotion and altered dopamine signaling. Thus, ADGRL3 is a potential target for treatment of neuropsychiatric disorders that involve dopamine dysfunction, but its basic signaling properties are poorly understood. Identification of adhesion GPCR signaling partners has been limited by a lack of tools to acutely activate these receptors in living cells. Here, we design a novel acute activation strategy to characterize ADGRL3 signaling by engineering a receptor construct in which we could trigger acute activation enzymatically. Using this assay, we found that ADGRL3 signals through G12/G13 and Gq, with G12/13 the most robustly activated. Gα12/13 is a new player in ADGRL3 biology, opening up unexplored roles for ADGRL3 in the brain. Our methodological advancements should be broadly useful in adhesion GPCR research.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Peptídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Fator 6 Ativador da Transcrição/agonistas , Fator 6 Ativador da Transcrição/química , Fator 6 Ativador da Transcrição/genética , Animais , Arrestina/química , Arrestina/genética , Arrestina/metabolismo , Sistemas CRISPR-Cas , Engenharia Celular , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/química , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Células HEK293 , Humanos , Cinética , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/química , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/química , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Ligação Proteica , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/química , Receptores de Peptídeos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
3.
Nat Chem Biol ; 16(10): 1096-1104, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32632293

RESUMO

Peptide ligands of class B G-protein-coupled receptors act via a two-step binding process, but the essential mechanisms that link their extracellular binding to intracellular receptor-arrestin interactions are not fully understood. Using NMR, crosslinking coupled to mass spectrometry, signaling experiments and computational approaches on the parathyroid hormone (PTH) type 1 receptor (PTHR), we show that initial binding of the PTH C-terminal part constrains the conformation of the flexible PTH N-terminal signaling epitope before a second binding event occurs. A 'hot-spot' PTH residue, His9, that inserts into the PTHR transmembrane domain at this second step allosterically engages receptor-arrestin coupling. A conformational change in PTHR intracellular loop 3 permits favorable interactions with ß-arrestin's finger loop. These results unveil structural determinants for PTHR-arrestin complex formation and reveal that the two-step binding mechanism proceeds via cooperative fluctuations between ligand and receptor, which extend to other class B G-protein-coupled receptors.


Assuntos
Arrestina/metabolismo , Hormônio Paratireóideo/metabolismo , Arrestina/química , Fosfatos de Cálcio , Microscopia Crioeletrônica , AMP Cíclico , Escherichia coli , Células HEK293 , Humanos , Simulação de Dinâmica Molecular , Hormônio Paratireóideo/química , Receptores Acoplados a Proteínas G
4.
J Biol Chem ; 295(19): 6498-6508, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32238431

RESUMO

Arrestin-1 is the arrestin family member responsible for inactivation of the G protein-coupled receptor rhodopsin in photoreceptors. Arrestin-1 is also well-known to interact with additional protein partners and to affect other signaling cascades beyond phototransduction. In this study, we investigated one of these alternative arrestin-1 binding partners, the glycolysis enzyme enolase-1, to map the molecular contact sites between these two proteins and investigate how the binding of arrestin-1 affects the catalytic activity of enolase-1. Using fluorescence quench protection of strategically placed fluorophores on the arrestin-1 surface, we observed that arrestin-1 primarily engages enolase-1 along a surface that is opposite of the side of arrestin-1 that binds photoactivated rhodopsin. Using this information, we developed a molecular model of the arrestin-1-enolase-1 complex, which was validated by targeted substitutions of charge-pair interactions. Finally, we identified the likely source of arrestin's modulation of enolase-1 catalysis, showing that selective substitution of two amino acids in arrestin-1 can completely remove its effect on enolase-1 activity while still remaining bound to enolase-1. These findings open up opportunities for examining the functional effects of arrestin-1 on enolase-1 activity in photoreceptors and their surrounding cells.


Assuntos
Arrestina/química , Biomarcadores Tumorais/química , Proteínas de Ligação a DNA/química , Modelos Moleculares , Complexos Multienzimáticos/química , Fosfopiruvato Hidratase/química , Rodopsina/química , Proteínas Supressoras de Tumor/química , Arrestina/genética , Sítios de Ligação , Biomarcadores Tumorais/genética , Catálise , Proteínas de Ligação a DNA/genética , Humanos , Complexos Multienzimáticos/genética , Fosfopiruvato Hidratase/genética , Rodopsina/genética , Proteínas Supressoras de Tumor/genética
5.
Int J Mol Sci ; 20(3)2019 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-30691068

RESUMO

The adenosine monophosphate-activated protein kinase (AMPK) plays a central role in the regulation of cellular metabolism. Recent studies reveal a novel role for AMPK in the regulation of glucose and other carbohydrates flux by controlling the endocytosis of transporters. The first step in glucose metabolism is glucose uptake, a process mediated by members of the GLUT/SLC2A (glucose transporters) or HXT (hexose transporters) family of twelve-transmembrane domain glucose transporters in mammals and yeast, respectively. These proteins are conserved from yeast to humans, and multiple transporters-each with distinct kinetic properties-compete for plasma membrane occupancy in order to enhance or limit the rate of glucose uptake. During growth in the presence of alternative carbon sources, glucose transporters are removed and replaced with the appropriate transporter to help support growth in response to this environment. New insights into the regulated protein trafficking of these transporters reveal the requirement for specific α-arrestins, a little-studied class of protein trafficking adaptor. A defining feature of the α-arrestins is that each contains PY-motifs, which can bind to the ubiquitin ligases from the NEDD4/Rsp5 (Neural precursor cell Expressed, Developmentally Down-regulated 4 and Reverses Spt- Phenotype 5, respectively) family. Specific association of α-arrestins with glucose and carbohydrate transporters is thought to bring the ubiquitin ligase in close proximity to its membrane substrate, and thereby allows the membrane cargo to become ubiquitinated. This ubiquitination in turn serves as a mark to stimulate endocytosis. Recent results show that AMPK phosphorylation of the α-arrestins impacts their abundance and/or ability to stimulate carbohydrate transporter endocytosis. Indeed, AMPK or glucose limitation also controls α-arrestin gene expression, adding an additional layer of complexity to this regulation. Here, we review the recent studies that have expanded the role of AMPK in cellular metabolism to include regulation of α-arrestin-mediated trafficking of transporters and show that this mechanism of regulation is conserved over the ~150 million years of evolution that separate yeast from man.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Arrestina/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Animais , Arrestina/química , Sítios de Ligação , Endocitose , Proteínas Fúngicas/metabolismo , Glucose/metabolismo , Humanos , Transporte Proteico , Ubiquitinação
6.
Biochemistry ; 53(20): 3294-307, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24724832

RESUMO

Various studies have implicated the concave surface of arrestin in the binding of the cytosolic surface of rhodopsin. However, specific sites of contact between the two proteins have not previously been defined in detail. Here, we report that arrestin shares part of the same binding site on rhodopsin as does the transducin Gα subunit C-terminal tail, suggesting binding of both proteins to rhodopsin may share some similar underlying mechanisms. We also identify two areas of contact between the proteins near this region. Both sites lie in the arrestin N-domain, one in the so-called "finger" loop (residues 67-79) and the other in the 160 loop (residues 155-165). We mapped these sites using a novel tryptophan-induced quenching method, in which we introduced Trp residues into arrestin and measured their ability to quench the fluorescence of bimane probes attached to cysteine residues on TM6 of rhodopsin (T242C and T243C). The involvement of finger loop binding to rhodopsin was expected, but the evidence of the arrestin 160 loop contacting rhodopsin was not. Remarkably, our data indicate one site on rhodopsin can interact with multiple structurally separate sites on arrestin that are almost 30 Å apart. Although this observation at first seems paradoxical, in fact, it provides strong support for recent hypotheses that structural plasticity and conformational changes are involved in the arrestin-rhodopsin binding interface and that the two proteins may be able to interact through multiple docking modes, with arrestin binding to both monomeric and dimeric rhodopsin.


Assuntos
Arrestina/química , Arrestina/metabolismo , Rodopsina/química , Rodopsina/metabolismo , Animais , Sítios de Ligação/fisiologia , Células COS , Bovinos , Chlorocebus aethiops , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
7.
Proc Natl Acad Sci U S A ; 109(11): 4146-51, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22315426

RESUMO

Mammalian cells are capable of delivering multiple types of membrane capsules extracellularly. The limiting membrane of late endosomes can fuse with the plasma membrane, leading to the extracellular release of multivesicular bodies (MVBs), initially contained within the endosomes, as exosomes. Budding viruses exploit the TSG101 protein and endosomal sorting complex required for transport (ESCRT) machinery used for MVB formation to mediate the egress of viral particles from host cells. Here we report the discovery of a virus-independent cellular process that generates microvesicles that are distinct from exosomes and which, like budding viruses, are produced by direct plasma membrane budding. Such budding is driven by a specific interaction of TSG101 with a tetrapeptide PSAP motif of an accessory protein, arrestin domain-containing protein 1 (ARRDC1), which we show is localized to the plasma membrane through its arrestin domain. This interaction results in relocation of TSG101 from endosomes to the plasma membrane and mediates the release of microvesicles that contain TSG101, ARRDC1, and other cellular proteins. Unlike exosomes, which are derived from MVBs, ARRDC1-mediated microvesicles (ARMMs) lack known late endosomal markers. ARMMs formation requires VPS4 ATPase and is enhanced by the E3 ligase WWP2, which interacts with and ubiquitinates ARRDC1. ARRDC1 protein discharged into ARMMs was observed in co-cultured cells, suggesting a role for ARMMs in intercellular communication. Our findings reveal an intrinsic cellular mechanism that results in direct budding of microvesicles from the plasma membrane, providing a formal paradigm for the evolutionary recruitment of ESCRT proteins in the release of budding viruses.


Assuntos
Arrestina/química , Arrestina/metabolismo , Membrana Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Fatores de Transcrição/metabolismo , Vesículas Transportadoras/metabolismo , Adenosina Trifosfatases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Biomarcadores/metabolismo , Membrana Celular/ultraestrutura , Exossomos/metabolismo , Espaço Extracelular/metabolismo , Células HEK293 , Humanos , Fusão de Membrana , Modelos Biológicos , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Vesículas Transportadoras/ultraestrutura , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Liberação de Vírus
8.
PLoS One ; 6(12): e28723, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22174878

RESUMO

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.


Assuntos
Arrestina/química , Arrestina/metabolismo , MAP Quinase Quinase 1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Animais , Arrestinas/química , Arrestinas/metabolismo , Células COS , Bovinos , Chlorocebus aethiops , Embrião de Mamíferos/citologia , Ativação Enzimática , Fibroblastos/enzimologia , Células HEK293 , Humanos , Ligantes , Camundongos , Camundongos Knockout , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilação , Ligação Proteica , Conformação Proteica , Receptores Adrenérgicos beta 2/metabolismo , Relação Estrutura-Atividade , beta-Arrestinas
9.
Biochemistry ; 50(18): 3749-63, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21466165

RESUMO

Numerous mutations in E3 ubiquitin ligase parkin were shown to associate with familial Parkinson's disease. Here we show that parkin binds arrestins, versatile regulators of cell signaling. Arrestin-parkin interaction was demonstrated by coimmunoprecipitation of endogenous proteins from brain tissue and shown to be direct using purified proteins. Parkin binding enhances arrestin interactions with another E3 ubiquitin ligase, Mdm2, apparently by shifting arrestin conformational equilibrium to the basal state preferred by Mdm2. Although Mdm2 was reported to ubiquitinate arrestins, parkin-dependent increase in Mdm2 binding dramatically reduces the ubiquitination of both nonvisual arrestins, basal and stimulated by receptor activation, without affecting receptor internalization. Several disease-associated parkin mutations differentially affect the stimulation of Mdm2 binding. All parkin mutants tested effectively suppress arrestin ubiquitination, suggesting that bound parkin shields arrestin lysines targeted by Mdm2. Parkin binding to arrestins along with its effects on arrestin interaction with Mdm2 and ubiquitination is a novel function of this protein with implications for Parkinson's disease pathology.


Assuntos
Arrestina/química , Proteínas Proto-Oncogênicas c-mdm2/química , Ubiquitina-Proteína Ligases/química , Ubiquitina/química , Animais , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Lisina/química , Camundongos , Doença de Parkinson/metabolismo , Ligação Proteica , Coelhos , Espectrometria de Fluorescência/métodos
10.
Arch Biochem Biophys ; 507(2): 219-31, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21176771

RESUMO

A 50-kDa-polypeptide band peripherally bound to retinal rod outer segment (ROS) membranes was purified by anion-exchange chromatography. When the 50-kDa protein was compared with purified arrestin-1, it was observed that: (1) both proteins comigrated on sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and were recognized by either anti-50-kDa protein polyclonal antibodies or anti-arrestin-1 monoclonal antibodies; (2) protein fragments and peptide fingerprint maps obtained following limited and complete proteolysis with specific proteases were very similar for both molecules; and (3) several chromatographically-purified tryptic peptides from the 50-kDa protein possessed the same amino acid composition as tryptic peptides deduced from the reported arrestin-1 primary structure. Consequently, arrestin-1 and the purified 50-kDa protein must correspond to variants of the same molecule. However, in contrast to arrestin-1 that associated to the ROS membranes only in the presence of light and ATP, the 50-kDa protein interacted with the ROS membranes in a light-independent manner, either in the presence or absence of ATP. These results clearly established that phosphorylated and illuminated rhodopsin is not the membrane anchor for this variant of arrestin-1.


Assuntos
Arrestina/metabolismo , Membrana Celular/metabolismo , Luz , Segmento Externo da Célula Bastonete/metabolismo , Animais , Arrestina/química , Arrestina/isolamento & purificação , Bovinos , Membrana Celular/efeitos da radiação , Peso Molecular , Fosforilação/efeitos da radiação , Ligação Proteica/efeitos da radiação , Segmento Externo da Célula Bastonete/efeitos da radiação , Solubilidade , Especificidade por Substrato
11.
Mol Pharmacol ; 75(1): 19-26, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18820126

RESUMO

Dopamine D(2) and D(3) receptors are similar subtypes with distinct interactions with arrestins; the D(3) receptor mediates less agonist-induced translocation of arrestins than the D(2) receptor. The goals of this study were to compare nonphosphorylated arrestin-binding determinants in the second intracellular domain (IC2) of the D(2) and D(3) receptors to identify residues that contribute to the differential binding of arrestin to the subtypes. Arrestin 3 bound to glutathione transferase (GST) fusion proteins of the D(2) receptor IC2 more avidly than to the D(3) receptor IC2. Mutagenesis of the fusion proteins identified a residue at the C terminus of IC2, Lys149, that was important for the preferential binding of arrestin 3 to D(2)-IC2; arrestin binding to D(2)-IC2-K149C was greatly decreased compared with wild-type D(2)-IC2, whereas binding to the reciprocal mutant D(3)-IC2-C147K was enhanced compared with wild-type D(3)-IC2. Mutating this lysine in the full-length D(2) receptor to cysteine decreased the ability of the D(2) receptor to mediate agonist-induced arrestin 3 translocation to the membrane and decreased agonist-induced receptor internalization in human embryonic kidney 293 cells. The reciprocal mutation in the D(3) receptor increased receptor-mediated translocation of arrestin 3 without affecting agonist-induced receptor internalization. G protein-coupled receptor crystal structures suggest that Lys149, at the junction of IC2 and the fourth membrane-spanning helix, has intramolecular interactions that contribute to maintaining an inactive receptor state. It is suggested that the preferential agonist-induced binding of arrestin3 to the D(2) receptor over the D(3) receptor is due in part to Lys149, which could be exposed as a result of receptor activation.


Assuntos
Arrestina/química , Arrestina/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Arrestina/genética , Arrestina/isolamento & purificação , Sítios de Ligação , Fenômenos Biofísicos , Linhagem Celular , Cisteína/metabolismo , Glutationa Transferase/metabolismo , Humanos , Ligação de Hidrogênio , Rim/citologia , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Receptores de Dopamina D2/química , Receptores de Dopamina D2/genética , Receptores de Dopamina D3/química , Receptores de Dopamina D3/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos
12.
Expert Opin Biol Ther ; 8(7): 941-50, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18549324

RESUMO

BACKGROUND: Evidence suggests that the p53 tumor suppressor protein functions, in part, by limiting tumor angiogenesis. This effect is partly mediated by the ability of p53 to increase production of endogenous angiogenesis inhibitors, such as the collagen-derived antiangiogenic factors (CDAFs), endostatin and tumstatin. OBJECTIVE: To review the clinical and therapeutic implications of CDAFs and their regulation by p53. METHODS: We highlight the inhibitory role of CDAFs in angiogenesis and summarize evidence that p53 regulates the transcriptional program leading to their expression, synthesis, assembly and activation. RESULTS/CONCLUSION: The p53 gene is mutated in half of all human tumors and such cancers would be predicted to produce lower levels of CDAFs. We therefore believe that p53 function can be partially compensated by therapeutic use of CDAFs, which offers a promising new avenue for cancer treatment.


Assuntos
Inibidores da Angiogênese/farmacologia , Colágeno/química , Regulação da Expressão Gênica , Terapia Genética/métodos , Neovascularização Patológica , Proteína Supressora de Tumor p53/metabolismo , Animais , Arrestina/química , Autoantígenos/química , Colágeno Tipo IV/química , Endostatinas/química , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/química , Proteínas de Neoplasias/química , Fragmentos de Peptídeos/química
13.
J Neurochem ; 103(3): 1053-62, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17680991

RESUMO

Arrestins are multi-functional regulators of G protein-coupled receptors. Receptor-bound arrestins interact with >30 remarkably diverse proteins and redirect the signaling to G protein-independent pathways. The functions of free arrestins are poorly understood, and the interaction sites of the non-receptor arrestin partners are largely unknown. In this study, we show that cone arrestin, the least studied member of the family, binds c-Jun N-terminal kinase (JNK3) and Mdm2 and regulates their subcellular distribution. Using arrestin mutants with increased or reduced structural flexibility, we demonstrate that arrestin in all conformations binds JNK3 comparably, whereas Mdm2 preferentially binds cone arrestin 'frozen' in the basal state. To localize the interaction sites, we expressed separate N- and C-domains of cone and rod arrestins and found that individual domains bind JNK3 and remove it from the nucleus as efficiently as full-length proteins. Thus, the arrestin binding site for JNK3 includes elements in both domains with the affinity of partial sites on individual domains sufficient for JNK3 relocalization. N-domain of rod arrestin binds Mdm2, which localizes its main interaction site to this region. Comparable binding of JNK3 and Mdm2 to four arrestin subtypes allowed us to identify conserved residues likely involved in these interactions.


Assuntos
Arrestina/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Ambystoma , Animais , Arrestina/química , Arrestina/genética , Sítios de Ligação/fisiologia , Compartimento Celular/fisiologia , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Fluorescência Verde , Humanos , Indóis , Ligação Proteica/fisiologia , Conformação Proteica , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia
14.
Arch Immunol Ther Exp (Warsz) ; 55(4): 261-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17659379

RESUMO

INTRODUCTION: Cancer-associated retinopathy (CAR) is a paraneoplastic neurological syndrome resulting in progressive loss of vision and clinical signs of retinal degeneration. It is associated with various types of cancer and is also considered to be an autoimmune disorder that involves cross-reaction between autoantibodies and retinal proteins. The aim of this study was to establish whether immunoreactivity to retinal antigens (RAs) observed in patients with breast cancer is accompanied by any visual impairments. MATERIALS AND METHODS: Sera of 295 patients with diagnosed breast cancer were screened for the presence of anti-RAs antibodies using immunoblotting. Cellular immunoreactivity to RAs present in retinal extracts and to purified recoverin and arrestin was determined by means of a lymphocyte proliferation assay. Six patients with high-titer antibodies to RAs then underwent ophthalmic and neurological examinations. RESULTS: Four serum samples contained high-titer antibodies to a 46-kDa protein, most probably retinal alpha-enolase, three had antibodies to a 48-kDa protein identified as retinal arrestin, while 56-, 43-, 41-, and 34-kDa antigens were recognized only by one serum sample each. Moreover, weak cellular response to all the RAs tested was observed in one patient and another patient responded only to retinal extract. Two of the examined patients displayed symptoms of CAR. CONCLUSIONS: Immunoreactivity to RAs in patients with breast cancer may also be present in cases without clinical signs of CAR.


Assuntos
Arrestina/imunologia , Autoanticorpos/sangue , Doenças Autoimunes/imunologia , Neoplasias da Mama/complicações , Proteínas do Olho/imunologia , Síndromes Paraneoplásicas/imunologia , Fosfopiruvato Hidratase/imunologia , Doenças Retinianas/imunologia , Transtornos da Visão/imunologia , Idoso , Arrestina/química , Doenças Autoimunes/fisiopatologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/fisiopatologia , Eletrorretinografia , Potenciais Evocados Visuais , Proteínas do Olho/química , Feminino , Humanos , Pessoa de Meia-Idade , Peso Molecular , Oftalmoscopia , Síndromes Paraneoplásicas/fisiopatologia , Fosfopiruvato Hidratase/química , Doenças Retinianas/fisiopatologia , Transtornos da Visão/fisiopatologia , Acuidade Visual , Campos Visuais
15.
J Biol Chem ; 282(35): 25560-8, 2007 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-17606620

RESUMO

In this study we investigate conformational changes in Loop V-VI of visual arrestin during binding to light-activated, phosphorylated rhodopsin (Rho*-P) using a combination of site-specific cysteine mutagenesis and intramolecular fluorescence quenching. Introduction of cysteines at positions in the N-domain at residues predicted to be in close proximity to Ile-72 in Loop V-VI of arrestin (i.e. Glu-148 and Lys-298) appear to form an intramolecular disulfide bond with I72C, significantly diminishing the binding of arrestin to Rho*-P. Using a fluorescence approach, we show that the steady-state emission from a monobromobimane fluorophore in Loop V-VI is quenched by tryptophan residues placed at 148 or 298. This quenching is relieved upon binding of arrestin to Rho*-P. These results suggest that arrestin Loop V-VI moves during binding to Rho*-P and that conformational flexibility of this loop is essential for arrestin to adopt a high affinity binding state.


Assuntos
Arrestina/química , Rodopsina/química , Substituição de Aminoácidos , Animais , Arrestina/genética , Arrestina/metabolismo , Sítios de Ligação , Compostos Bicíclicos com Pontes/química , Compostos Bicíclicos com Pontes/metabolismo , Bovinos , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica , Estrutura Secundária de Proteína , Rodopsina/genética , Rodopsina/metabolismo , Espectrometria de Fluorescência
16.
J Mol Biol ; 368(2): 375-87, 2007 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-17359998

RESUMO

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.


Assuntos
Arrestina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Animais , Arrestina/química , Sítios de Ligação , Células COS , Linhagem Celular , Sobrevivência Celular , Chlorocebus aethiops , Dimerização , Humanos , Ligação Proteica , Conformação Proteica , Transporte Proteico , Tubulina (Proteína)/metabolismo
17.
Photochem Photobiol ; 83(2): 385-92, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17132044

RESUMO

Visual arrestin terminates the signal amplification cascade in photoreceptor cells by blocking the interaction of light activated phosphorylated rhodopsin with the G-protein transducin. Although crystal structures of arrestin and rhodopsin are available, it is still unknown how the complex of the two proteins is formed. To investigate the interaction sites of arrestin with rhodopsin various surface regions of recombinant arrestin were sterically blocked by different numbers of fluorophores (Alexa 633). The binding was recorded by time-resolved light scattering. To accomplish site-specific shielding of protein regions, in a first step all three wild-type cysteines were replaced by alanines. Nevertheless, regarding the magnitude and specificity of rhodopsin binding, the protein is still fully active. In a second step, new cysteines were introduced at selected sites to allow covalent binding of fluorophores. Upon attachment of Alexa 633 to the recombinant cysteines we observed that these bulky labels residing in the concave area of either the N- or the C-terminal domain do not perturb the activity of arrestin. By simultaneously modifying both domains with one Alexa 633 the binding capacity was reduced. The presence of two Alexa 633 molecules in each domain prevented binding of rhodopsin to arrestin. This observation indicates that both concave sites participate in binding.


Assuntos
Arrestina/química , Rodopsina/metabolismo , Animais , Arrestina/genética , Arrestina/metabolismo , Sequência de Bases , Sítios de Ligação , Bovinos , Cisteína/química , Primers do DNA/genética , Corantes Fluorescentes , Técnicas In Vitro , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fotoquímica , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
18.
Methods Enzymol ; 414: 50-63, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17110186

RESUMO

Finding natural and/or synthetic ligands that activate orphan G protein-coupled receptors (oGPCRs) is a major focus in current drug discovery efforts. Transfluor is a cell-based GPCR screening platform that utilizes an arrestin-green fluorescent protein conjugate (arrestin-GFP) to detect ligand interactions with GPCRs. The assay is ideally suited for oGPCRs because binding of arrestin-GFP to activated receptors is independent of the interacting G protein. Before embarking on a high-throughput screen, it is important to know that the target oGPCR can actually bind arrestin-GFP. This information was thought to be inaccessible, however, as arrestin-GFP recruitment is an agonist-driven process. This chapter describes an assay that enables GPCRs to be validated in Transfluor in the absence of ligand. This assay, termed the ligand-independent translocation (LITe) assay, utilizes a modified G protein-coupled receptor kinase to bypass the requirement of ligand for initiating arrestin-GFP translocation. Using the LITe assay, one can determine if an oGPCR binds arrestin-GFP and if the response is quantifiable by high-content screening instruments. In addition, the assay expedites the development and identification of oGPCR stable cell lines with the best Transfluor properties. In this way, the assay provides criteria for selecting the best oGPCRs to move forward for a Transfluor screening campaign. Moreover, the assay can be used for quality control purposes during the orphan receptor screen itself by providing positive translocation responses for calculation of Z prime values. In summary, the LITe assay is a powerful new technology that enables a faster and more reliable path forward in the deorphanization of GPCRs with Transfluor.


Assuntos
Arrestina/química , Bioquímica/métodos , Receptores Acoplados a Proteínas G/química , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Proteínas de Fluorescência Verde/química , Humanos , Ligantes , Ligação Proteica , Isoformas de Proteínas , Transporte Proteico , Controle de Qualidade , Receptores de Superfície Celular/química
19.
Methods Enzymol ; 414: 63-78, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17110187

RESUMO

G protein-coupled receptors (GPCRs) have proven to be one of the most successful target classes for drug discovery. Accordingly, many assays are available to screen GPCRs, including radioactive-binding assays, second messenger signaling assays, and downstream reporter assays. One of the more novel approaches is the Transfluor technology, a cell-based assay that uses a detectable tag on a cytosolic protein, called arrestin, that is involved in the desensitization or inactivation of GPCRs. Monitoring the translocation of GFP-tagged arrestin from the cytosol to activated GPCRs at the plasma membrane measures the pharmacological effect of test compounds that bind the receptor target. Moreover, the Transfluor assay provides further, high-content information on the test compound itself and its effects on cell processes due to the fluorescent imaging of whole cells used in this screen. Screening known GPCRs with Transfluor against large compound libraries is best accomplished in cell lines stably expressing an optimum level of the target receptor. This chapter describes how to generate a clonal cell line stably expressing the known GPCR with suitable Transfluor properties. It then describes the steps involved in performing a Transfluor screen and discusses high content data resulting from the screen.


Assuntos
Arrestina/química , Bioquímica/métodos , Receptores Acoplados a Proteínas G/química , Algoritmos , Arrestinas/química , Linhagem Celular Tumoral , Desenho de Fármacos , Proteínas de Fluorescência Verde/química , Humanos , Proteínas Luminescentes , Modelos Biológicos , Transporte Proteico , Transfecção
20.
Invest Ophthalmol Vis Sci ; 47(9): 3745-53, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936082

RESUMO

PURPOSE: Cone function and survival are compromised in the guanylate cyclase-1 (GC1) knockout mouse. Disruption of the light-driven translocation of cone arrestin is one of the phenotypes of cone cells in this retina: the cone arrestin in these cells is localized to the outer segments and synaptic terminals, regardless of the state of light adaptation. The purpose of this study was to determine whether the expression of GC1 restores cone arrestin translocation in the cone cells of postnatal GC1 knockout mouse retina. METHODS: Subretinal injections of AAV-GC1 were performed on 3-week-old GC1 KO mice. Electroretinographic and immunohistochemical analyses of treated retinas were carried out 5 weeks after injection. GC1 and cone arrestin antibodies were used to identify photoreceptors transduced by the AAV vector and to localize cone arrestin within cone cells, respectively. RESULTS: Treatment of GC1 knockout retinas with AAV-GC1 restored the light-driven translocation of cone arrestin in transduced cone cells. Staining patterns for cone arrestin in transduced and wild-type cone cells were indistinguishable after dark and light adaptation. In dark-adapted retinas, cone arrestin was distributed throughout the subcellular compartments of the cone cells. In light-adapted retinas, cone arrestin was concentrated in the cone outer segments. Successful restoration of cone arrestin translocation did not translate to a restoration of cone ERG responses, which remained undetectable in the treated retinas. CONCLUSIONS: AAV-mediated expression of GC1 in a subpopulation of cone cells in postnatal GC1 knockout retina restores light-driven translocation of cone arrestin in these cells. These findings, which show that fully developed cone cells that have developed in the absence of GC1 can respond to viral-mediated expression of this enzyme, support further analysis of this animal model of Leber congenital amaurosis type 1 (LCA1), a disease that results from null mutations in the gene encoding this enzyme.


Assuntos
Arrestina/metabolismo , Dependovirus/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Guanilato Ciclase/genética , Luz , Transporte Proteico/efeitos da radiação , Receptores de Superfície Celular/genética , Células Fotorreceptoras Retinianas Cones/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Arrestina/química , Eletrorretinografia , Técnica Indireta de Fluorescência para Anticorpo , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Microscopia Confocal , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia , Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/efeitos da radiação , Transgenes , Visão Ocular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA