Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Cell Sci ; 129(7): 1366-77, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26869225

RESUMEN

The G-protein-coupled receptor 37 ( GPR37) has been implicated in the juvenile form of Parkinson's disease, in dopamine signalling and in the survival of dopaminergic cells in animal models. The structure and function of the receptor, however, have remained enigmatic. Here, we demonstrate that although GPR37 matures and is exported from the endoplasmic reticulum in a normal manner upon heterologous expression in HEK293 and SH-SY5Y cells, its long extracellular N-terminus is subject to metalloproteinase-mediated limited proteolysis between E167 and Q168. The proteolytic processing is a rapid and efficient process that occurs constitutively. Moreover, the GPR37 ectodomain is released from cells by shedding, a phenomenon rarely described for GPCRs. Immunofluorescence microscopy further established that although full-length receptors are present in the secretory pathway until the trans-Golgi network, GPR37 is expressed at the cell surface predominantly in the N-terminally truncated form. This notion was verified by flow cytometry and cell surface biotinylation assays. These new findings on the GPR37 N-terminal limited proteolysis may help us to understand the role of this GPCR in the pathophysiology of Parkinson's disease and in neuronal function in general.


Asunto(s)
Metaloproteasas/metabolismo , Enfermedad de Parkinson/patología , Proteolisis , Receptores Acoplados a Proteínas G/metabolismo , Línea Celular , Dipéptidos/farmacología , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Metaloproteasas/antagonistas & inhibidores , Estructura Terciaria de Proteína
2.
Traffic ; 10(1): 116-29, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19000170

RESUMEN

The human delta opioid receptor (hdeltaOR) is a G-protein-coupled receptor that is mainly involved in the modulation of pain and mood. Only one nonsynonymous single nucleotide polymorphism (T80G) has been described, causing Phe27Cys substitution in the receptor N-terminus and showing association with substance dependence. In this study, we expressed the two hdeltaOR variants in a heterologous expression system with an identical genetic background. They differed greatly during early steps of biosynthesis, displaying a significant difference in the maturation efficiency (50% and 85% for the Cys27 and Phe27 variants, respectively). The Cys27 variant also showed accumulation in pre-Golgi compartments of the secretory pathway and impaired targeting to endoplasmic reticulum (ER)-associated degradation following long-term expression. In addition, the cell surface receptors of the Cys27 variant internalized constitutively. Replacement of phenylalanine with other amino acids revealed that cysteine at position 27 decreased the mature receptor/precursor ratio most extensively, suggesting a thiol-mediated retention of precursors in the ER. However, cysteine did not cause a major folding defect because pharmacological characteristics and the maturation kinetics of the variants were identical, and an opioid antagonist was able to enhance the maturation of both variants. We conclude that, instead of causing loss of function, Phe27Cys polymorphism of the hdeltaOR causes a gain-of-function phenotype, which may have implications for the regulation of receptor expression at the cell surface and possibly also for the susceptibility to pathophysiological states.


Asunto(s)
Polimorfismo Genético/genética , Procesamiento Proteico-Postraduccional/genética , Receptores Opioides delta/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Secuencia Conservada , Cisteína/genética , Cisteína/metabolismo , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Humanos , Cinética , Datos de Secuencia Molecular , Fenilalanina/genética , Fenilalanina/metabolismo , Transporte de Proteínas , Receptores Opioides delta/química , Receptores Opioides delta/genética , Alineación de Secuencia
3.
J Biol Chem ; 285(37): 28850-61, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20587416

RESUMEN

The beta(1)-adrenergic receptor (beta(1)AR) is the predominant betaAR in the heart, mediating the catecholamine-stimulated increase in cardiac rate and force of contraction. Regulation of this important G protein-coupled receptor is nevertheless poorly understood. We describe here the biosynthetic profile of the human beta(1)AR and reveal novel features relevant to its regulation using an inducible heterologous expression system in HEK293(i) cells. Metabolic pulse-chase labeling and cell surface biotinylation assays showed that the synthesized receptors are efficiently and rapidly transported to the cell surface. The N terminus of the mature receptor is extensively modified by sialylated mucin-type O-glycosylation in addition to one N-glycan attached to Asn(15). Furthermore, the N terminus was found to be subject to limited proteolysis, resulting in two membrane-bound C-terminal fragments. N-terminal sequencing of the fragments identified two cleavage sites between Arg(31) and Leu(32) and Pro(52) and Leu(53), which were confirmed by cleavage site and truncation mutants. Metalloproteinase inhibitors were able to inhibit the cleavage, suggesting that it is mediated by a matrix metalloproteinase or a disintegrin and metalloproteinase (ADAM) family member. Most importantly, the N-terminal cleavage was found to occur not only in vitro but also in vivo. Receptor activation mediated by the betaAR agonist isoproterenol enhanced the cleavage in a concentration- and time-dependent manner, and it was also enhanced by direct stimulation of protein kinase C and adenylyl cyclase. Mutation of the Arg(31)-Leu(32) cleavage site stabilized the mature receptor. We hypothesize that the N-terminal cleavage represents a novel regulatory mechanism of cell surface beta(1)ARs.


Asunto(s)
Péptido Hidrolasas/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Agonistas Adrenérgicos beta/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Glicosilación/efectos de los fármacos , Humanos , Isoproterenol/farmacología , Mutación , Péptido Hidrolasas/genética , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Receptores Adrenérgicos beta 1/genética
4.
Mol Cell Biochem ; 351(1-2): 173-81, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21234650

RESUMEN

A quarter of the human population with European background carries at least one allele of the OPRD1 gene that encodes the delta opioid receptor with cysteine at the amino acid position 27 (hδOR(Cys27)) instead of the evolutionary conserved phenylalanine (hδOR(Phe27)). The two variants have indistinguishable pharmacological properties but, importantly, hδOR(Cys27) differs from hδOR(Phe27) in having low maturation efficiency, lower stability at the cell surface and pronounced intracellular location. Both variants were previously shown to interact with the Sarco(endo)plasmic reticulum Ca²+ ATPase (SERCA) 2b in the early phase of their biosynthesis. We analyzed by pulse-chase assays, whether cellular signaling can affect hδOR(Cys27) maturation. Neither activation of the receptor by a δOR-specific agonist Leu-enkephalin, induction of intracellular calcium (Ca²+) release by ATP nor the direct stimulation of SERCA 2b by protein kinase C activation affected receptor maturation in HEK-293 cells. No signaling-mediated regulation of receptor maturation could therefore be demonstrated. Instead, we found by using single cell Ca²+ measurements that over-expression of hδOR(Cys27), but not hδOR(Phe27), compromised ATP-induced intracellular Ca²+-signaling. Furthermore, hδOR(Cys27) precursors showed slower dissociation from SERCA2b and hδOR(Cys27) expression caused down-regulation of the homocysteine-inducible endoplasmic reticulum-resident ubiquitin domain-like member 1 protein (HERP). We suggest that aging individuals with at least one hδOR(Cys27) encoding allele might have lowered threshold for Ca²+ dysregulation in neurons expressing hδOR.


Asunto(s)
Señalización del Calcio/genética , Cisteína/genética , Fenilalanina/genética , Polimorfismo Genético , Receptores Opioides delta/genética , Adenosina Trifosfato/metabolismo , Western Blotting , Línea Celular , Activación Enzimática , Humanos , Proteína Quinasa C/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
5.
Mol Biol Cell ; 17(5): 2243-55, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16495341

RESUMEN

The luteinizing hormone receptor (LHR) is a G protein-coupled receptor that is expressed in multiple RNA messenger forms. The common rat ectodomain splice variant is expressed concomitantly with the full-length LHR in tissues and is a truncated transcript corresponding to the partial ectodomain with a unique C-terminal end. Here we demonstrate that the variant alters the behavior of the full-length receptor by misrouting it away from the normal secretory pathway in human embryonic kidney 293 cells. The variant was expressed as two soluble forms of M(r) 52,000 and M(r) 54,000, but although the protein contains a cleavable signal sequence, no secretion to the medium was observed. Only a very small fraction of the protein was able to gain hormone-binding ability, suggesting that it is retained in the endoplasmic reticulum (ER) by its quality control due to misfolding. This was supported by the finding that the variant was found to interact with calnexin and calreticulin and accumulated together with these ER chaperones in a specialized juxtanuclear subcompartment of the ER. Only proteasomal blockade with lactacystin led to accumulation of the variant in the cytosol. Importantly, coexpression of the variant with the full-length LHR resulted in reduction in the number of receptors that were capable of hormone binding and were expressed at the cell surface and in targeting of immature receptors to the juxtanuclear ER subcompartment. Thus, the variant mediated misrouting of the newly synthesized full-length LHRs may provide a way to regulate the number of cell surface receptors.


Asunto(s)
Empalme Alternativo , Retículo Endoplásmico/metabolismo , Receptores de HL/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Calnexina/metabolismo , Calreticulina/metabolismo , Células Cultivadas , Gonadotropina Coriónica/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Retículo Endoplásmico/química , Humanos , Chaperonas Moleculares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Pliegue de Proteína , Estructura Terciaria de Proteína/genética , Transporte de Proteínas , Ratas , Receptores de HL/análisis , Receptores de HL/genética
6.
J Mol Biol ; 371(3): 622-38, 2007 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-17588601

RESUMEN

Calcium (Ca(2+)) plays a pivotal role in both cellular signaling and protein synthesis. However, it is not well understood how calcium metabolism and synthesis of secreted and membrane-bound proteins are related. Here we demonstrate that the sarco(endo)plasmic reticulum Ca(2+) ATPase 2b (SERCA2b), which maintains high Ca(2+) concentration in the lumen of the endoplasmic reticulum, interacts specifically with the human delta opioid receptor during early steps of receptor biogenesis in human embryonic kidney 293 cells. The interaction involves newly synthesized incompletely folded receptor precursors, because the association between the delta opioid receptor and SERCA2b (i) was short-lived and took place soon after receptor translation, (ii) was not affected by misfolding of the receptor, and (iii) decreased if receptor folding was enhanced by opioid receptor pharmacological chaperone. The physical association with SERCA2b was found to be a universal feature among G protein-coupled receptors within family A and was shown to occur also between the endogenously expressed luteinizing hormone receptor and SERCA2b in rat ovaries. Importantly, active SERCA2b rather than undisturbed Ca(2+) homeostasis was found to be essential for delta opioid receptor biogenesis, as inhibition of its Ca(2+) pumping activity by thapsigargin reduced the interaction and impaired the efficiency of receptor maturation, two phenomena that were not affected by a Ca(2+) ionophore A23187. Nevertheless, inhibition of SERCA2b did not compromise the functionality of receptors that were able to mature. Thus, we propose that the association with SERCA2b is required for efficient folding and/or membrane integration of G protein-coupled receptors.


Asunto(s)
Retículo Endoplásmico/enzimología , Receptores Opioides delta/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Calnexina/metabolismo , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Inmunoprecipitación , Ovario/efectos de los fármacos , Ovario/enzimología , Embarazo , Unión Proteica/efectos de los fármacos , Pliegue de Proteína , Precursores de Proteínas/metabolismo , Ratas , Receptores de HL/metabolismo , Receptores Opioides delta/química , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , Especificidad por Sustrato/efectos de los fármacos , Tapsigargina/farmacología
7.
Mol Cell Biol ; 31(11): 2326-40, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21464208

RESUMEN

Agonist-induced activation of the δ-opioid receptor (δOR) was recently shown to augment ß- and γ-secretase activities, which increased the production of ß-amyloid peptide (Aß), known to accumulate in the brain tissues of Alzheimer's disease (AD) patients. Previously, the δOR variant with a phenylalanine at position 27 (δOR-Phe27) exhibited more efficient receptor maturation and higher stability at the cell surface than did the less common cysteine (δOR-Cys27) variant. For this study, we expressed these variants in human SH-SY5Y and HEK293 cells expressing exogenous or endogenous amyloid precursor protein (APP) and assessed the effects on APP processing. Expression of δOR-Cys27, but not δOR-Phe27, resulted in a robust accumulation of the APP C83 C-terminal fragment and the APP intracellular domain, while the total soluble APP and, particularly, the ß-amyloid 40 levels were decreased. These changes upon δOR-Cys27 expression coincided with decreased localization of APP C-terminal fragments in late endosomes and lysosomes. Importantly, a long-term treatment with a subset of δOR-specific ligands or a c-Src tyrosine kinase inhibitor suppressed the δOR-Cys27-induced APP phenotype. These data suggest that an increased constitutive internalization and/or concurrent signaling of the δOR-Cys27 variant affects APP processing through altered endocytic trafficking of APP.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Endocitosis , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Enfermedad de Alzheimer/metabolismo , Sustitución de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Western Blotting , Citometría de Flujo , Células HEK293 , Humanos , Inmunoprecipitación , Mutación , Polimorfismo de Nucleótido Simple , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Receptores Opioides delta/química , Transducción de Señal
8.
FEBS J ; 277(13): 2815-29, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20528919

RESUMEN

Sarco(endo)plasmic reticulum calcium ATPase (SERCA)2b maintains the cellular Ca(2+) homeostasis by transferring Ca(2+) from the cytosol to the lumen of the endoplasmic reticulum (ER). Recently, SERCA2b has also been shown to be involved in the biosynthesis of secreted and membrane proteins via direct protein-protein interactions, involving components of the ER folding and quality-control machinery, as well as newly synthesized G protein-coupled receptors. Here we demonstrate that the human delta opioid receptor (hdeltaOR) exists in a ternary complex with SERCA2b and the ER molecular chaperone calnexin. The interaction between SERCA2b and hdeltaOR in vivo did not require calnexin as it was independent of the C-terminal calnexin-interacting domain of SERCA2b. However, the receptor was able to mediate co-immunoprecipitation of calnexin with the C-terminally truncated SERCA2b. The association of SERCA2b with hdeltaOR was regulated in vitro by Ca(2+) and ATP in a manner that was opposite to the calnexin-hdeltaOR interaction. Importantly, co-expression of the catalytically inactive SERCA2b(D351A) or calnexin binding-compromised SERCA2bDeltaC mutants with the receptor decreased the expression of mature receptors in a manner that did not directly relate to changes in the ER Ca(2+) concentration. We conclude that dynamic interactions among SERCA2b, calnexin and the hdeltaOR precursor orchestrate receptor biogenesis and are regulated by Ca(2+) and ATP. We further hypothesize that the primary role of SERCA2b in this process is to act as a Ca(2+) sensor in the vicinity of active translocons, integrating protein folding with local fluctuations of ER Ca(2+) levels.


Asunto(s)
Calnexina/metabolismo , Receptores Opioides delta/biosíntesis , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Humanos
9.
J Biol Chem ; 282(32): 23171-83, 2007 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-17550902

RESUMEN

Accumulating evidence has indicated that membrane-permeable G protein-coupled receptor ligands can enhance cell surface targeting of their cognate wild-type and mutant receptors. This pharmacological chaperoning was thought to result from ligand-mediated stabilization of immature receptors in the endoplasmic reticulum (ER). In the present study, we directly tested this hypothesis using wild-type and mutant forms of the human delta-opioid receptor as models. ER-localized receptors were isolated by expressing the receptors in HEK293 cells under tightly controlled tetracycline induction and blocking their ER export with brefeldin A. The ER-retained delta-opioid receptor precursors were able to bind [(3)H]diprenorphine with high affinity, and treatment of cells with an opioid antagonist naltrexone led to a 2-fold increase in the number of binding sites. After removing the transport block, the antagonist-mediated increase in the number of receptors was detectable at the cell surface by flow cytometry and cell surface biotinylation assay. Importantly, opioid ligands, both antagonists and agonists, were found to stabilize the ER-retained receptor precursors in an in vitro heat inactivation assay and the treatment enhanced dissociation of receptor precursors from the molecular chaperone calnexin. Thus, we conclude that pharmacological chaperones facilitate plasma membrane targeting of delta-opioid receptors by binding and stabilizing receptor precursors, thereby promoting their release from the stringent ER quality control.


Asunto(s)
Retículo Endoplásmico/metabolismo , Receptores Opioides/metabolismo , Sitios de Unión , Biotinilación , Brefeldino A/química , Calnexina/química , Línea Celular , Membrana Celular/metabolismo , ADN/química , Humanos , Cinética , Ligandos , Chaperonas Moleculares/metabolismo , Unión Proteica , Receptores Opioides/química , Receptores Opioides delta/química
10.
J Biol Chem ; 281(23): 15780-9, 2006 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-16595649

RESUMEN

Protein palmitoylation is a reversible lipid modification that plays important roles for many proteins involved in signal transduction, but relatively little is known about the regulation of this modification and the cellular location where it occurs. We demonstrate that the human delta opioid receptor is palmitoylated at two distinct cellular locations in human embryonic kidney 293 cells and undergoes dynamic regulation at one of these sites. Although palmitoylation could be readily observed for the mature receptor (Mr 55,000), [3H]palmitate incorporation into the receptor precursor (Mr 45,000) could be detected only following transport blockade with brefeldin A, nocodazole, and monensin, indicating that the modification occurs initially during or shortly after export from the endoplasmic reticulum. Blocking of palmitoylation with 2-bromopalmitate inhibited receptor cell surface expression, indicating that it is needed for efficient intracellular transport. However, cell surface biotinylation experiments showed that receptors can also be palmitoylated once they have reached the plasma membrane. At this location, palmitoylation is regulated in a receptor activation-dependent manner, as was indicated by the opioid agonist-promoted increase in the turnover of receptor-bound palmitate. This agonist-mediated effect did not require receptor-G protein coupling and occurred at the cell surface without the need for internalization or recycling. The activation-dependent modulation of receptor palmitoylation may thus contribute to the regulation of receptor function at the plasma membrane.


Asunto(s)
Ácido Palmítico/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Fracciones Subcelulares/metabolismo , Línea Celular , Humanos
11.
J Biol Chem ; 280(28): 26622-9, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15901736

RESUMEN

Increasing evidence suggests that the folding and maturation of monomeric proteins and assembly of multimeric protein complexes in the endoplasmic reticulum (ER) may be inefficient not only for mutants that carry changes in the primary structure but also for wild type proteins. In the present study, we demonstrate that the rat luteinizing hormone receptor, a G protein-coupled receptor, is one of these proteins that matures inefficiently and appears to be very prone to premature degradation. A substantial portion of the receptors in stably transfected human embryonic kidney 293 cells existed in immature form of M(r) 73,000, containing high mannose-type N-linked glycans. In metabolic pulse-chase studies, only approximately 20% of these receptor precursors were found to gain hormone binding ability and matured to a form of M(r) 90,000, containing bi- and multiantennary sialylated N-linked glycans. The rest had a propensity to form disulfide-bonded complexes with a M(r) 120,000 protein in the ER membrane and were eventually targeted for degradation in proteasomes. The number of membrane-bound receptor precursors increased when proteasomal degradation was inhibited, and no cytosolic receptor forms were detected, suggesting that retrotranslocation of the misfolded/incompletely folded receptors is tightly coupled to proteasomal function. Furthermore, a proteasomal blockade was found to increase the number of receptors that were capable of hormone binding. Thus, these results raise the interesting possibility that luteinizing hormone receptor expression at the cell surface may be controlled at the ER level by regulating the number of newly synthesized proteins that will mature and escape the ER quality control and premature degradation.


Asunto(s)
Membrana Celular/metabolismo , Regulación de la Expresión Génica , Receptores de HL/química , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Western Blotting , Línea Celular , Citosol/metabolismo , ADN/metabolismo , Disulfuros/química , Electroforesis en Gel Bidimensional , Electroforesis en Gel de Poliacrilamida , Retículo Endoplásmico/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Ligandos , Masculino , Oligosacáridos/química , Polisacáridos/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores de HL/fisiología , Fracciones Subcelulares/metabolismo , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA