Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 13: 848816, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35721749

RESUMEN

The angiotensin type 2 (AT2) receptor and the bradykinin type 2 (B2) receptor are G protein-coupled receptors (GPCRs) that have major roles in the cardiovascular system. The two receptors are known to functionally interact at various levels, and there is some evidence that the observed crosstalk may occur as a result of heteromerization. We investigated evidence for heteromerization of the AT2 receptor and the B2 receptor in HEK293FT cells using various bioluminescence resonance energy transfer (BRET)-proximity based assays, including the Receptor Heteromer Investigation Technology (Receptor-HIT) and the NanoBRET ligand-binding assay. The Receptor-HIT assay showed that Gαq, GRK2 and ß-arrestin2 recruitment proximal to AT2 receptors only occurred upon B2 receptor coexpression and activation, all of which is indicative of AT2-B2 receptor heteromerization. Additionally, we also observed specific coupling of the B2 receptor with the Gαz protein, and this was found only in cells coexpressing both receptors and stimulated with bradykinin. The recruitment of Gαz, Gαq, GRK2 and ß-arrestin2 was inhibited by B2 receptor but not AT2 receptor antagonism, indicating the importance of B2 receptor activation within AT2-B2 heteromers. The close proximity between the AT2 receptor and B2 receptor at the cell surface was also demonstrated with the NanoBRET ligand-binding assay. Together, our data demonstrate functional interaction between the AT2 receptor and B2 receptor in HEK293FT cells, resulting in novel pharmacology for both receptors with regard to Gαq/GRK2/ß-arrestin2 recruitment (AT2 receptor) and Gαz protein coupling (B2 receptor). Our study has revealed a new mechanism for the enigmatic and poorly characterized AT2 receptor to be functionally active within cells, further illustrating the role of heteromerization in the diversity of GPCR pharmacology and signaling.


Asunto(s)
Receptor de Angiotensina Tipo 2 , Receptor de Bradiquinina B2 , Bradiquinina/farmacología , Ligandos , Receptor de Angiotensina Tipo 2/fisiología , Receptor de Bradiquinina B2/fisiología , Receptores Acoplados a Proteínas G , Arrestina beta 2
2.
Biochem Pharmacol ; 188: 114521, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33741329

RESUMEN

Transactivation of the epidermal growth factor receptor (EGFR) by the angiotensin II (AngII) type 1 (AT1) receptor is involved in AT1 receptor-dependent growth effects and cardiovascular pathologies, however the mechanisms underpinning this transactivation are yet to be fully elucidated. Recently, a potential intermediate of this process was identified following the discovery that a kinase called TRIO was involved in AngII/AT1 receptor-mediated transactivation of EGFR. To investigate the mechanisms by which TRIO acts as an intermediate in AngII/AT1 receptor-mediated EGFR transactivation we used bioluminescence resonance energy transfer (BRET) assays to investigate proximity between the AT1 receptor, EGFR, TRIO and other proteins of interest. We found that AngII/AT1 receptor activation caused a Gαq-dependent increase in proximity of TRIO with Gγ2 and the AT1-EGFR heteromer, as well as trafficking of TRIO towards the Kras plasma membrane marker and into early, late and recycling endosomes. In contrast, we found that AngII/AT1 receptor activation caused a Gαq-independent increase in proximity of TRIO with Grb2, GRK2 and PKCζ, as well as trafficking of TRIO up to the plasma membrane from the Golgi. Furthermore, we confirmed the proximity between the AT1 receptor and the EGFR using the Receptor-Heteromer Investigation Technology, which showed AngII-induced recruitment of Grb2, GRK2, PKCζ, Gγ2 and TRIO to the EGFR upon AT1 coexpression. In summary, our results provide further evidence for the existence of the AT1-EGFR heteromer and reveal potential mechanisms by which TRIO contributes to the transactivation process.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Transducción de Señal/fisiología , Angiotensina II/farmacología , Relación Dosis-Respuesta a Droga , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/agonistas , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Receptor de Angiotensina Tipo 2/agonistas , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología
3.
J Clin Invest ; 129(1): 406-421, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30530993

RESUMEN

Activation of the type 1 angiotensin II receptor (AT1) triggers proinflammatory signaling through pathways independent of classical Gq signaling that regulate vascular homeostasis. Here, we report that the AT1 receptor preformed a heteromeric complex with the receptor for advanced glycation endproducts (RAGE). Activation of the AT1 receptor by angiotensin II (Ang II) triggered transactivation of the cytosolic tail of RAGE and NF-κB-driven proinflammatory gene expression independently of the liberation of RAGE ligands or the ligand-binding ectodomain of RAGE. The importance of this transactivation pathway was demonstrated by our finding that adverse proinflammatory signaling events induced by AT1 receptor activation were attenuated when RAGE was deleted or transactivation of its cytosolic tail was inhibited. At the same time, classical homeostatic Gq signaling pathways were unaffected by RAGE deletion or inhibition. These data position RAGE transactivation by the AT1 receptor as a target for vasculoprotective interventions. As proof of concept, we showed that treatment with the mutant RAGE peptide S391A-RAGE362-404 was able to inhibit transactivation of RAGE and attenuate Ang II-dependent inflammation and atherogenesis. Furthermore, treatment with WT RAGE362-404 restored Ang II-dependent atherogenesis in Ager/Apoe-KO mice, without restoring ligand-mediated signaling via RAGE, suggesting that the major effector of RAGE activation was its transactivation.


Asunto(s)
Aterosclerosis/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal , Activación Transcripcional , Animales , Aterosclerosis/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Eliminación de Gen , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados para ApoE , Dominios Proteicos , Receptor de Angiotensina Tipo 1/genética
4.
Mol Endocrinol ; 30(8): 889-904, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27355191

RESUMEN

Nephrogenic syndrome of inappropriate antidiuresis (NSIAD) is a genetic disease first described in 2 unrelated male infants with severe symptomatic hyponatremia. Despite undetectable arginine vasopressin levels, patients have inappropriately concentrated urine resulting in hyponatremia, hypoosmolality, and natriuresis. Here, we describe and functionally characterize a novel vasopressin type 2 receptor (V2R) gain-of-function mutation. An L312S substitution in the seventh transmembrane domain was identified in a boy presenting with water-induced hyponatremic seizures at the age of 5.8 years. We show that, compared with wild-type V2R, the L312S mutation results in the constitutive production of cAMP, indicative of the gain-of-function NSIAD profile. Interestingly, like the previously described F229V and I130N NSIAD-causing mutants, this appears to both occur in the absence of notable constitutive ß-arrestin2 recruitment and can be reduced by the inverse agonist Tolvaptan. In addition, to understand the effect of various V2R substitutions on the full receptor "life-cycle," we have used and further developed a bioluminescence resonance energy transfer intracellular localization assay using multiple localization markers validated with confocal microscopy. This allowed us to characterize differences in the constitutive and ligand-induced localization and trafficking profiles of the novel L312S mutation as well as for previously described V2R gain-of-function mutants (NSIAD; R137C and R137L), loss-of-function mutants (nephrogenic diabetes insipidus; R137H, R181C, and M311V), and a putative silent V266A V2R polymorphism. In doing so, we describe differences in trafficking between unique V2R substitutions, even at the same amino acid position, therefore highlighting the value of full and thorough characterization of receptor function beyond simple signaling pathway analysis.


Asunto(s)
Mutación/genética , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo , Preescolar , AMP Cíclico/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Microscopía Confocal , Polimorfismo Genético , Unión Proteica , Transducción de Señal/genética , Transducción de Señal/fisiología , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
5.
Br J Pharmacol ; 171(2): 364-74, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24206104

RESUMEN

BACKGROUND AND PURPOSE: The orexin system regulates a multitude of key physiological processes, particularly involving maintenance of metabolic homeostasis. Consequently, there is considerable potential for pharmaceutical development for the treatment of disorders from narcolepsy to metabolic syndrome. It acts through the hormonal activity of two endogenous peptides, orexin A binding to orexin receptors 1 and 2 (OX1 and OX2) with similar affinity, and orexin B binding to OX2 with higher affinity than OX1 receptors. We have previously revealed data differentiating orexin receptor subtypes with respect to their relative stability in forming orexin receptor-arrestin-ubiquitin complexes measured by BRET. Recycling and cellular signalling distinctions were also observed. Here, we have investigated, using BRET, the molecular determinants involved in providing OX2 receptors with greater ß-arrestin-ubiquitin complex stability. EXPERIMENTAL APPROACH: The contribution of the C-terminal tail of the OX receptors was investigated by bulk substitution and site-specific mutagenesis using BRET and inositol phosphate assays. KEY RESULTS: Replacement of the OX1 receptor C-terminus with that of the OX2 receptor did not result in the expected gain of function, indicating a role for intracellular domain configuration in addition to primary structure. Furthermore, two out of the three putative serine/threonine clusters in the C-terminus were found to be involved in OX2 receptor-ß-arrestin-ubiquitin complex formation. CONCLUSIONS AND IMPLICATIONS: This study provides fundamental insights into the molecular elements that influence receptor-arrestin-ubiquitin complex formation. Understanding how and why the orexin receptors can be functionally differentiated brings us closer to exploiting these receptors as drug targets.


Asunto(s)
Arrestina/metabolismo , Receptores de Orexina/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Arrestina/genética , ADN Complementario/biosíntesis , ADN Complementario/genética , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Fosfatos de Inositol/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Mutagénesis , Neuropéptidos , Receptores de Orexina/genética , Orexinas , Serina/química , Serina/metabolismo , Treonina/química , Treonina/metabolismo , Ubiquitina/genética
6.
PLoS One ; 8(6): e65885, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762448

RESUMEN

Arginine vasopressin (AVP) is released from the posterior pituitary and controls water homeostasis. AVP binding to vasopressin V2 receptors (V2Rs) located on kidney collecting duct epithelial cells triggers activation of Gs proteins, leading to increased cAMP levels, trafficking of aquaporin-2 water channels, and consequent increased water permeability and antidiuresis. Typically, loss-of-function V2R mutations cause nephrogenic diabetes insipidus (NDI), whereas gain-of-function mutations cause nephrogenic syndrome of inappropriate antidiuresis (NSIAD). Here we provide further characterization of two mutant V2Rs, R181C and M311V, reported to cause complete and partial NDI respectively, together with a V266A variant, in a patient diagnosed with NSIAD. Our data in HEK293FT cells revealed that for cAMP accumulation, AVP was about 500- or 30-fold less potent at the R181C and M311V mutants than at the wild-type receptor respectively (and about 4000- and 60-fold in COS7 cells respectively). However, in contrast to wild type V2R, the R181C mutant failed to increase inositol phosphate production, while with the M311V mutant, AVP exhibited only partial agonism in addition to a 37-fold potency decrease. Similar responses were detected in a BRET assay for ß-arrestin recruitment, with the R181C receptor unresponsive to AVP, and partial agonism with a 23-fold decrease in potency observed with M311V in both HEK293FT and COS7 cells. Notably, the V266A V2R appeared functionally identical to the wild-type receptor in all assays tested, including cAMP and inositol phosphate accumulation, ß-arrestin interaction, and in a BRET assay of receptor ubiquitination. Each receptor was expressed at comparable levels. Hence, the M311V V2R retains greater activity than the R181C mutant, consistent with the milder phenotype of NDI associated with this mutant. Notably, the R181C mutant appears to be a Gs protein-biased receptor incapable of signaling to inositol phosphate or recruiting ß-arrestin. The etiology of NSIAD in the patient with V266A V2R remains unknown.


Asunto(s)
Diabetes Insípida Nefrogénica/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Síndrome de Secreción Inadecuada de ADH/genética , Mutación , Polimorfismo Genético , Receptores de Vasopresinas/genética , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Arginina Vasopresina/metabolismo , Arrestinas/genética , Arrestinas/metabolismo , Células COS , Chlorocebus aethiops , AMP Cíclico/metabolismo , Diabetes Insípida Nefrogénica/metabolismo , Diabetes Insípida Nefrogénica/patología , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Regulación de la Expresión Génica , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Células HEK293 , Humanos , Síndrome de Secreción Inadecuada de ADH/metabolismo , Síndrome de Secreción Inadecuada de ADH/patología , Fosfatos de Inositol/metabolismo , Receptores de Vasopresinas/metabolismo , Transducción de Señal , beta-Arrestinas
7.
PLoS One ; 8(5): e64672, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23700486

RESUMEN

Heteromerization can play an important role in regulating the activation and/or signal transduction of most forms of receptors, including receptor tyrosine kinases (RTKs). The study of receptor heteromerization has evolved extensively with the emergence of resonance energy transfer based approaches such as bioluminescence resonance energy transfer (BRET). Here, we report an adaptation of our Receptor-Heteromer Investigation Technology (Receptor-HIT) that has recently been published as the G protein-coupled receptor (GPCR) Heteromer Identification Technology (GPCR-HIT). We now demonstrate the utility of this approach for investigating RTK heteromerization by examining the functional interaction between the epidermal growth factor (EGF) receptor (EGFR; also known as erbB1/HER1) and heregulin (HRG) receptor 3 (HER3; also known as erbB3) in live HEK293FT cells using recruitment of growth factor receptor-bound protein 2 (Grb2) to the activated receptors. We found that EGFR and HER3 heteromerize specifically as demonstrated by HRG inducing a BRET signal between EGFR/Rluc8 and Grb2/Venus only when HER3 was co-expressed. Similarly, EGF stimulation promoted a specific BRET signal between HER3/Rluc8 and Grb2/Venus only when EGFR was co-expressed. Both EGF and HRG effects on Grb2 interaction are dose-dependent, and specifically blocked by EGFR inhibitor AG-1478. Furthermore, truncation of HER3 to remove the putative Grb2 binding sites appears to abolish EGF-induced Grb2 recruitment to the EGFR-HER3 heteromer. Our results support the concept that EGFR interacts with Grb2 in both constitutive and EGF-dependent manners and this interaction is independent of HER3 co-expression. In contrast, HER3-Grb2 interaction requires the heteromerization between EGFR and HER3. These findings clearly indicate the importance of EGFR-HER3 heteromerization in HER3-mediated Grb2-dependent signaling pathways and supports the central role of HER3 in the diversity and regulation of HER family functioning.


Asunto(s)
Receptores ErbB/metabolismo , Proteína Adaptadora GRB2/metabolismo , Multimerización de Proteína , Receptor ErbB-3/metabolismo , Sitios de Unión , Transferencia Resonante de Energía de Fluorescencia , Proteína Adaptadora GRB2/antagonistas & inhibidores , Células HEK293 , Humanos , Cinética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Quinazolinas/farmacología , Transducción de Señal , Tirfostinos/farmacología
8.
J Biol Chem ; 287(16): 12952-65, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22371491

RESUMEN

We have provided the first evidence for specific heteromerization between the α(1A)-adrenoceptor (α(1A)AR) and CXC chemokine receptor 2 (CXCR2) in live cells. α(1A)AR and CXCR2 are both expressed in areas such as the stromal smooth muscle layer of the prostate. By utilizing the G protein-coupled receptor (GPCR) heteromer identification technology on the live cell-based bioluminescence resonance energy transfer (BRET) assay platform, our studies in human embryonic kidney 293 cells have identified norepinephrine-dependent ß-arrestin recruitment that was in turn dependent upon co-expression of α(1A)AR with CXCR2. These findings have been supported by co-localization observed using confocal microscopy. This norepinephrine-dependent ß-arrestin recruitment was inhibited not only by the α(1)AR antagonist Terazosin but also by the CXCR2-specific allosteric inverse agonist SB265610. Furthermore, Labetalol, which is marketed for hypertension as a nonselective ß-adrenoceptor antagonist with α(1)AR antagonist properties, was identified as a heteromer-specific-biased agonist exhibiting partial agonism for inositol phosphate production but essentially full agonism for ß-arrestin recruitment at the α(1A)AR-CXCR2 heteromer. Finally, bioluminescence resonance energy transfer studies with both receptors tagged suggest that α(1A)AR-CXCR2 heteromerization occurs constitutively and is not modulated by ligand. These findings support the concept of GPCR heteromer complexes exhibiting distinct pharmacology, thereby providing additional mechanisms through which GPCRs can potentially achieve their diverse biological functions. This has important implications for the use and future development of pharmaceuticals targeting these receptors.


Asunto(s)
Próstata/metabolismo , Estructura Cuaternaria de Proteína , Receptores Adrenérgicos alfa 1/química , Receptores de Interleucina-8B/química , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Regulación Alostérica/fisiología , Animales , Arrestinas/metabolismo , Células CHO , Quimiocinas/metabolismo , Cricetinae , Células HEK293 , Humanos , Fosfatos de Inositol/metabolismo , Labetalol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/farmacología , Prazosina/análogos & derivados , Prazosina/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interleucina-8B/metabolismo , beta-Arrestinas
9.
Cell Signal ; 23(11): 1767-76, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21740964

RESUMEN

The cardiovascular hormone angiotensin II (AngII) exerts its actions via two G protein-coupled receptor (GPCR) subtypes, AT(1) and AT(2), which often display antagonistic functions. Methodological constraints have so far precluded detailed analyses of the ligand-dependency, cellular localization, and functional relevance of AngII receptor interactions in live cells. In this study, we utilize a protein-fragment complementation assay (PCA) and GPCR-Heteromer Identification Technology (GPCR-HIT) to provide the first detailed investigation of the ligand-dependency and cellular localization of AngII receptor interactions in human embryonic kidney 293 cells. Fluorescent-tagged receptor constructs for PCA and GPCR-HIT displayed normal affinity and selectivity for AngII (AT(1): IC(50)=1.0-1.6nM; AT(2): IC(50)=2.0-3.0nM). Well-characterized angiotensin receptor interactions were used as positive and negative controls to demonstrate the sensitivity and specificity of these fluorescence-based assays. We report that AT(1)-AT(2) receptor heteromers form constitutively, are localized to the plasma membrane and perinuclear compartments, and do not internalize following AngII stimulation despite arrestin being recruited specifically to the heteromer. Our findings using novel fluorescence-based technologies reveal a previously unrecognized mechanism of angiotensin receptor cross-talk involving cross-inhibition of AT(1) receptor internalization through heteromerization with the AT(2) receptor subtype.


Asunto(s)
Angiotensina II/metabolismo , Arrestinas/metabolismo , Bioensayo , Fragmentos de Péptidos/análisis , Receptor Cross-Talk/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Sistema Renina-Angiotensina/fisiología , Angiotensina II/farmacología , Arrestinas/farmacología , Membrana Celular/metabolismo , Movimiento Celular , Fluorescencia , Células HEK293 , Humanos , Ligandos , Microscopía Confocal , Fragmentos de Péptidos/química , Fosforilación , Plásmidos , Polimerizacion , Unión Proteica , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 2/genética , Sensibilidad y Especificidad , Transfección
10.
Assay Drug Dev Technol ; 9(1): 21-30, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21133678

RESUMEN

Understanding the role of G protein-coupled receptor (GPCR; also known as a 7 transmembrane receptor) heteromerization in the physiology and pathophysiology of cellular function has now become a major research focus. However, there is currently a lack of cell-based assays capable of profiling the specific functional consequences of heteromerization in a ligand-dependent manner. Understanding the pharmacology specifically associated with heteromer function in contrast to monomer or homomer function enables the so-called biochemical fingerprints of the receptor heteromer to be ascertained. This is the first step in establishing the physiological relevance of heteromerization, the goal of everyone in the field, as these fingerprints can then be utilized in future endeavors to elucidate heteromer function in native tissues. The simple, robust, ligand-dependent methodology described in this study utilizes a novel configuration of components of a proximity-based reporter system. This is exemplified by the use of bioluminescence resonance energy transfer due to the advantages of real-time live cell monitoring of proximity specifically between the heteromer complex and a protein that is recruited in a ligand-dependent manner, in this case, ß-arrestin 2. Further, the demonstration of Z'-factor values in excess of 0.6 shows the potential of the method for screening compounds for heteromer-selective or biased activity. Three previously characterized GPCR heteromers, the chemokine receptor heteromers CCR2-CCR5 and CCR2-CXCR4, as well as the angiotensin II receptor type 1-bradykinin receptor type 2 heteromer, have been used to illustrate the profiling capability and specificity of the GPCR heteromer identification technology.


Asunto(s)
Arrestinas/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Mapeo de Interacción de Proteínas/métodos , Receptores Acoplados a Proteínas G/metabolismo , Biotecnología/métodos , Células HEK293 , Humanos , Multimerización de Proteína , Arrestina beta 2 , beta-Arrestinas
11.
Artículo en Inglés | MEDLINE | ID: mdl-22654789

RESUMEN

The bioluminescence resonance energy transfer (BRET) technique has become extremely valuable for the real-time monitoring of protein-protein interactions in live cells. This method is highly amenable to the detection of G protein-coupled receptor (GPCR) interactions with proteins critical for regulating their function, such as ß-arrestins. Of particular interest to endocrinologists is the ability to monitor interactions involving endocrine receptors, such as orexin receptor 2 or vasopressin type II receptor. The BRET method utilizes heterologous co-expression of fusion proteins linking one protein of interest (GPCR) to a bioluminescent donor enzyme, a variant of Renilla luciferase, and a second protein of interest (ß-arrestin) to an acceptor fluorophore. If in close proximity, energy resulting from oxidation of the coelenterazine substrate by the donor will transfer to the acceptor, which in turn fluoresces. Using novel luciferase constructs, we were able to monitor interactions not detectable using less sensitive BRET combinations in the same configuration. In particular, we were able to show receptor/ß-arrestin interactions in an agonist-independent manner using Rluc8-tagged mutant receptors, in contrast to when using Rluc. Therefore, the enhanced BRET methodology has not only enabled live cell compound screening as we have recently published, it now provides a new level of sensitivity for monitoring specific transient, weak or hardly detectable protein-protein complexes, including agonist-independent GPCR/ß-arrestin interactions. This has important implications for the use of BRET technologies in endocrine drug discovery programs as well as academic research.

12.
J Biomol Screen ; 13(9): 888-98, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18812574

RESUMEN

The bioluminescence resonance energy transfer (BRET) technique has become extremely popular for studying protein-protein interactions in living cells and real time. Of particular interest is the ability to monitor interactions between G protein-coupled receptors, such as the thyrotropin-releasing hormone receptor (TRHR), and proteins critical for regulating their function, such as beta-arrestin. Using TRHR/beta-arrestin interactions, we have demonstrated improvements to all 3 generations of BRET (BRET(1), BRET(2), and eBRET) by using the novel forms of luciferase, Rluc2 and Rluc8, developed by the Gambhir laboratory. Furthermore, for the 1st time it was possible to use the BRET2 system to detect ligand-induced G protein-coupled receptor/beta-arrestin interactions over prolonged periods (on the scale of hours rather than seconds) with a very stable signal. As demonstrated by our Z'-factor data, these luciferases increase the sensitivity of BRET to such an extent that they substantially increase the potential applicability of this technology for effective drug discovery high-throughput screening.


Asunto(s)
Transferencia de Energía , Receptores Acoplados a Proteínas G/química , Línea Celular , Diseño de Fármacos , Descubrimiento de Drogas , Humanos , Fosfatos de Inositol/química , Cinética , Luciferasas/química , Proteínas Luminiscentes/química , Receptores de Hormona Liberadora de Tirotropina/química , Proteínas Recombinantes de Fusión/química , Factores de Tiempo
13.
Mol Pharmacol ; 73(5): 1362-70, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18230715

RESUMEN

The Duffy antigen/receptor for chemokines (DARC) is an unusual chemokine receptor that binds a large number of inflammatory chemokines of both the CC and CXC families with nanomolar affinity, yet it lacks the ability to signal upon ligand binding. Using bioluminescent resonant energy transfer, we have demonstrated for the first time that DARC exists as a constitutive homo-oligomer in living cells and furthermore that DARC hetero-oligomerizes with the CC chemokine receptor CCR5. DARC-CCR5 interaction impairs chemotaxis and calcium flux through CCR5, whereas internalization of CCR5 in response to ligand binding remains unchanged. These results suggest a novel mechanism by which DARC could modulate inflammatory responses to chemokines in vivo.


Asunto(s)
Antagonistas de los Receptores CCR5 , Sistema del Grupo Sanguíneo Duffy/química , Sistema del Grupo Sanguíneo Duffy/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Animales , Arrestinas/metabolismo , Sitios de Unión , Calcio/metabolismo , Línea Celular , Supervivencia Celular , Quimiotaxis , Dimerización , Endocitosis , Células Endoteliales/citología , Células Endoteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Ligandos , Ratones , Unión Proteica , Estructura Cuaternaria de Proteína , Transfección , beta-Arrestinas
14.
Nat Protoc ; 1(1): 337-45, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17406254

RESUMEN

A substantial range of protein-protein interactions can be readily monitored in real time using bioluminescence resonance energy transfer (BRET). The procedure involves heterologous coexpression of fusion proteins, which link proteins of interest to a bioluminescent donor enzyme or acceptor fluorophore. Energy transfer between these proteins is then detected. This protocol encompasses BRET1, BRET2 and the recently described eBRET, including selection of the donor, acceptor and substrate combination, fusion construct generation and validation, cell culture, fluorescence and luminescence detection, BRET detection and data analysis. The protocol is particularly suited to studying protein-protein interactions in live cells (adherent or in suspension), but cell extracts and purified proteins can also be used. Furthermore, although the procedure is illustrated with references to mammalian cell culture conditions, this protocol can be readily used for bacterial or plant studies. Once fusion proteins are generated and validated, the procedure typically takes 48-72 h depending on cell culture requirements.


Asunto(s)
Mediciones Luminiscentes/métodos , Mapeo de Interacción de Proteínas/métodos , Técnicas de Cultivo , Proteínas Luminiscentes/análisis , Proteínas/análisis , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/análisis
15.
Nat Struct Mol Biol ; 12(9): 814-21, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116438

RESUMEN

Growth hormone is believed to activate the growth hormone receptor (GHR) by dimerizing two identical receptor subunits, leading to activation of JAK2 kinase associated with the cytoplasmic domain. However, we have reported previously that dimerization alone is insufficient to activate full-length GHR. By comparing the crystal structure of the liganded and unliganded human GHR extracellular domain, we show here that there is no substantial change in its conformation on ligand binding. However, the receptor can be activated by rotation without ligand by inserting a defined number of alanine residues within the transmembrane domain. Fluorescence resonance energy transfer (FRET), bioluminescence resonance energy transfer (BRET) and coimmunoprecipitation studies suggest that receptor subunits undergo specific transmembrane interactions independent of hormone binding. We propose an activation mechanism involving a relative rotation of subunits within a dimeric receptor as a result of asymmetric placement of the receptor-binding sites on the ligand.


Asunto(s)
Modelos Biológicos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Receptores de Somatotropina/química , Receptores de Somatotropina/metabolismo , Rotación , Secuencia de Aminoácidos , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Cristalografía por Rayos X , Dimerización , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Subunidades de Proteína/genética , Receptores de Somatotropina/genética , Espectrometría de Fluorescencia
16.
J Biol Chem ; 279(36): 37726-33, 2004 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-15187087

RESUMEN

The alpha(v)beta(3) integrin is known to cooperate with receptor tyrosine kinases to enhance cellular responses. To determine whether alpha(v)beta(3) regulates transforming growth factor beta (TGFbeta) 1-induced responses, we investigated the interaction between alpha(v)beta(3) and TGFbeta type II receptor (TGFbetaIIR) in primary human lung fibroblasts. We report that TGFbeta1 up-regulates cell surface and mRNA expression of alpha(v)beta(3) in a time- and dose-dependent manner. Co-immunoprecipitation and confocal microscopy showed that TGFbetaRII associates and clusters with alpha(v)beta(3), following TGFbeta1 exposure. This association was not observed with alpha(v)beta(5) or alpha(5)beta(1). We also used a novel molecular proximity assay, bioluminescence resonance energy transfer (BRET), to quantify this dynamic interaction in living cells. TGFbeta1 stimulation resulted in a BRET signal within 5 min, whereas tenascin, which binds alpha(v)beta(3), did not induce a substantial BRET signal. Co-exposure to tenascin and TGFbeta1 produced no further increases in BRET than TGFbeta1 alone. Cyclin D1 was rapidly induced in cells co-exposed to TGFbeta1 and tenascin, and as a consequence proliferation induced by TGFbeta1 was dramatically enhanced in cells co-exposed to tenascin or vitronectin. Cholesterol depletion inhibited the interaction between TGFbetaRII and alpha(v)beta(3) and abrogated the proliferative effect. The cyclic RGD peptide, GpenGRGDSPCA, which blocks alpha(v)beta(3), also abolished the synergistic proliferative effect seen. These results indicate a new interaction partner for the alpha(v)beta(3) integrin, the TGFbetaIIR, in which TGFbeta1-induced responses are potentiated in the presence alpha(v)beta(3) ligands. Our data provide a novel mechanism by which TGFbeta1 may contribute to abnormal wound healing and tissue fibrosis.


Asunto(s)
División Celular/fisiología , Integrina alfaVbeta3/metabolismo , Pulmón/citología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Línea Celular , Fibroblastos/citología , Humanos , Ligandos , Unión Proteica , ARN Mensajero/genética , Factor de Crecimiento Transformador beta/genética
17.
J Biol Chem ; 277(52): 50422-30, 2002 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-12393857

RESUMEN

G-protein-coupled receptors (GPCRs) are regulated by a complex network of mechanisms such as oligomerization and internalization. Using the GPCR subtypes for thyrotropin-releasing hormone (TRHR1 and TRHR2), the aim of this study was to determine if subtype-specific differences exist in the trafficking process. If so, we wished to determine the impact of homo- and hetero-oligomerization on TRHR subtype trafficking as a potential mechanism for the differential cellular responses induced by TRH. Expression of either beta-arrestin 1 or 2 promoted TRHR1 internalization. In contrast, only beta-arrestin 2 could enhance TRHR2 internalization. The preference for beta-arrestin 2 by TRHR2 was supported by the impairment of TRHR2 trafficking in mouse embryonic fibroblasts (MEFs) from either a beta-arrestin 2 knockout or a beta-arrestin 1/2 knockout, while TRHR1 trafficking was only abolished in MEFs lacking both beta-arrestins. The differential beta-arrestin-dependence of TRHR2 was directly measured in live cells using bioluminescence resonance energy transfer (BRET). Both BRET and confocal microscopy were also used to demonstrate that TRHR subtypes form hetero-oligomers. In addition, these hetero-oligomers have altered internalization kinetics compared with the homo-oligomer. The formation of TRHR1/2 heteromeric complexes increased the interaction between TRHR2 and beta-arrestin 1. This may be due to conformational differences between TRHR1/2 hetero-oligomers versus TRHR2 homo-oligomers as a mutant TRHR1 (TRHR1 C335Stop) that does not interact with beta-arrestins, could also enhance TRHR2/beta-arrestin 1 interaction. This study demonstrates that TRHR subtypes are differentially regulated by the beta-arrestins and also provides the first evidence that the interactions of TRHRs with beta-arrestin may be altered by hetero-oligomer formation.


Asunto(s)
Arrestinas/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Hormona Liberadora de Tirotropina/química , Animales , Arrestinas/química , Células COS , Línea Celular , Chlorocebus aethiops , Clonación Molecular , Fibroblastos/metabolismo , Vectores Genéticos , Proteínas Fluorescentes Verdes , Riñón , Cinética , Proteínas Luminiscentes/metabolismo , Sustancias Macromoleculares , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas/química , Ratas , Receptores de Hormona Liberadora de Tirotropina/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Transfección , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
18.
Biol Reprod ; 66(6): 1762-7, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12021059

RESUMEN

Leptin, the 16-kDa peptide hormone product of the ob gene, regulates body weight via the hypothalamus but also influences several aspects of reproductive function. Results of previous studies have suggested that pregnancy is a state of leptin resistance, because food consumption remains stable or increases despite a progressive rise in plasma leptin across most of gestation. In the present study, we assessed whether this apparent leptin resistance during rat pregnancy was due to either increased plasma leptin-binding activity and/or reduced expression of hypothalamic leptin receptor. Plasma leptin increased from 2.2 +/- 0.4 ng/ml before pregnancy to a maximum at midgestation (4.2 +/- 0.8 ng/ml on Day 12) and then fell by Day 22 and remained low throughout lactation. Despite the higher plasma leptin levels in pregnancy, food consumption increased from a minimum of 13.6 +/- 0.5 g/day before pregnancy to a peak of 21.9 +/- 0.6 g/day on Day 19, then fell before parturition (11.9 +/- 0.4 g/day on Day 22). At least part of the increase in plasma leptin during pregnancy was attributable to a marked increase (P < 0.001) in plasma leptin-binding activity between diestrus and late pregnancy, which then fell after birth but remained at midpregnancy levels to at least Day 12 of lactation. Hypothalamic expression of mRNA encoding the long form of the leptin receptor (Ob-Rb) was elevated in early pregnancy (Day 7) but returned to prepregnancy levels by midgestation and remained stable thereafter. The results of this study confirm that pregnancy in the rat is a state of relative leptin resistance, which is due primarily to increased plasma leptin-binding activity rather than to changes in hypothalamic Ob-Rb expression.


Asunto(s)
Expresión Génica , Hipotálamo/metabolismo , Lactancia , Leptina/sangre , Preñez/fisiología , Receptores de Superficie Celular/genética , Animales , Proteínas Sanguíneas/metabolismo , Resistencia a Medicamentos , Ingestión de Alimentos , Femenino , Edad Gestacional , Embarazo , Unión Proteica , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptores de Leptina , Valores de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...