Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
J Comp Neurol ; 527(12): 2069-2085, 2019 08 15.
Article de Anglais | MEDLINE | ID: mdl-30809795

RÉSUMÉ

Central activation of fibroblast growth factor (FGF) receptors regulates peripheral glucose homeostasis and reduces food intake in preclinical models of obesity and diabetes. The current work was undertaken to advance our understanding of the receptor expression, as sites of ligand action by FGF19, FGF21, and FGF1 in the mammalian brain remains unresolved. Recent advances in automated RNAscope in situ hybridization and droplet digital PCR (ddPCR) technology allowed us to interrogate central FGFR/beta klotho (Klb) system at the cellular level in the mouse, with relevant comparisons to nonhuman primate and human brain. FGFR1-3 gene expression was broadly distributed throughout the CNS in Mus musculus, with FGFR1 exhibiting the greatest heterogeneity. FGFR4 expression localized only in the medial habenula and subcommissural organ of mice. Likewise, Klb mRNA was restricted to the suprachiasmatic nucleus (SCh) and select midbrain and hindbrain nuclei. ddPCR in the rodent hypothalamus confirmed that, although expression levels are indeed low for Klb, there is nonetheless a bonafide subpopulation of Klb+ cells in the hypothalamus. In NHP and human midbrain and hindbrain, Klb + cells are quite rare, as is expression of FGFR4. Collectively, these data provide the most robust central map of the FGFR/Klb system to date and highlight central regions that may be of critical importance to assess central ligand effects with pharmacological dosing, such as the putative interactions between the endocrine FGFs and FGFR1/Klb, or FGF19 with FGFR4.


Sujet(s)
Cartographie cérébrale/méthodes , Encéphale/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Glucuronidase/métabolisme , Hybridation in situ/méthodes , Animaux , Facteurs de croissance fibroblastique/analyse , Glucuronidase/analyse , Humains , Protéines Klotho , Macaca fascicularis , Mâle , Souris , Souris de lignée C57BL
2.
Nat Chem Biol ; 12(11): 896-898, 2016 Nov.
Article de Anglais | MEDLINE | ID: mdl-27595330

RÉSUMÉ

The With-No-Lysine (K) (WNK) kinases play a critical role in blood pressure regulation and body fluid and electrolyte homeostasis. Herein, we introduce the first orally bioavailable pan-WNK-kinase inhibitor, WNK463, that exploits unique structural features of the WNK kinases for both affinity and kinase selectivity. In rodent models of hypertension, WNK463 affects blood pressure and body fluid and electro-lyte homeostasis, consistent with WNK-kinase-associated physiology and pathophysiology.


Sujet(s)
Système cardiovasculaire/effets des médicaments et des substances chimiques , Imidazoles/pharmacologie , Rein/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyrrolidines/pharmacologie , Bibliothèques de petites molécules/pharmacologie , Animaux , Système cardiovasculaire/métabolisme , Humains , Imidazoles/composition chimique , Rein/métabolisme , Tests de la fonction rénale , Souris , Souris de lignée C57BL , Souris transgéniques , Inhibiteurs de protéines kinases/composition chimique , Protein-Serine-Threonine Kinases/métabolisme , Pyrrolidines/composition chimique , Rats , Rat Sprague-Dawley , Bibliothèques de petites molécules/composition chimique
3.
Microvasc Res ; 91: 22-9, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24246569

RÉSUMÉ

PURPOSE: The objectives of this study were (i) to characterize the hemodynamic responses caused by controlled hemorrhage (HEM) in pentobarbital-anesthetized rats, and (ii) to determine the responses elicited by systemic bolus injections of isotonic saline (0.15M) or hypertonic saline (3M) given 5min after completion of HEM. RESULTS: Controlled HEM (4.3±0.2ml/rat at 1.5ml/min) resulted in a pronounced and sustained fall in mean arterial blood pressure (MAP) to about 40mmHg. The fall in MAP was associated with a reduction in hindquarter vascular resistance (HQR) but no changes in renal (RR) or mesenteric (MR) vascular resistances. Systemic injections of isotonic saline (96-212µmol/kg i.v., in 250-550µl) did not produce immediate responses but promoted the recovery of MAP to levels below pre-HEM values. Systemic injections of hypertonic saline (750-3000µmol/kg, i.v., in 250-550µl) produced immediate and pronounced falls in MAP, RR, MR and especially HQR of 30-120s in duration. However, hypertonic saline prompted a full recovery of MAP, HQR and RR to pre-HEM levels and an increase in MR to levels above pre-HEM values. CONCLUSIONS: This study demonstrates that (i) HEM induced a pronounced fall in MAP which likely involved a fall in cardiac output and HQR, (ii) isotonic saline did not fully normalize MAP, and (iii) hypertonic saline produced dramatic initial responses, and promoted normalization of MAP probably by restoring blood volume and cardiac output through sequestration of fluid from intracellular compartments.


Sujet(s)
Pression artérielle/effets des médicaments et des substances chimiques , Hydrodynamique , Solution isotonique/composition chimique , Solution saline hypertonique/composition chimique , Choc hémorragique/anatomopathologie , Résistance vasculaire , Animaux , Pression sanguine , Hémodynamique/effets des médicaments et des substances chimiques , Hémorragie/anatomopathologie , Mâle , Artères mésentériques/anatomopathologie , Pentobarbital/composition chimique , Rats , Rat Sprague-Dawley , Artère rénale/anatomopathologie , Facteurs temps , Vasodilatation/effets des médicaments et des substances chimiques
4.
Blood ; 119(5): 1217-27, 2012 Feb 02.
Article de Anglais | MEDLINE | ID: mdl-22147898

RÉSUMÉ

The core pathology of sickle cell disease (SCD) starts with the erythrocyte (RBC). Aberration in MAPK/ERK1/2 signaling, which can regulate cell adhesion, occurs in diverse pathologies. Because RBCs contain abundant ERK1/2, we predicted that ERK1/2 is functional in sickle (SS) RBCs and promotes adherence, a hallmark of SCD. ERK1/2 remained active in SS but not normal RBCs. ß(2)-adrenergic receptor stimulation by epinephrine can enhance ERK1/2 activity only in SS RBCs via PKA- and tyrosine kinase p72(syk)-dependent pathways. ERK signaling is implicated in RBC ICAM-4 phosphorylation, promoting SS RBC adhesion to the endothelium. SS RBC adhesion and phosphorylation of both ERK and ICAM-4 all decreased with continued cell exposure to epinephrine, implying that activation of ICAM-4-mediated SS RBC adhesion is temporally associated with ERK1/2 activation. Furthermore, recombinant ERK2 phosphorylated α- and ß-adducins and dematin at the ERK consensus motif. Cytoskeletal protein 4.1 also showed dynamic phosphorylation but not at the ERK consensus motif. These results demonstrate that ERK activation induces phosphorylation of cytoskeletal proteins and the adhesion molecule ICAM-4, promoting SS RBC adhesion to the endothelium. Thus, blocking RBC ERK1/2 activation, such as that promoted by catecholamine stress hormones, could ameliorate SCD pathophysiology.


Sujet(s)
Drépanocytose/sang , Molécules d'adhérence cellulaire/physiologie , Endothélium vasculaire/physiologie , Membrane érythrocytaire/métabolisme , Érythrocytes anormaux/physiologie , Système de signalisation des MAP kinases/physiologie , Cellules 3T3 , Drépanocytose/métabolisme , Drépanocytose/anatomopathologie , Animaux , Adhérence cellulaire , Molécules d'adhérence cellulaire/métabolisme , Membrane cellulaire/métabolisme , Cellules cultivées , Endothélium vasculaire/métabolisme , Activation enzymatique/physiologie , Érythrocytes anormaux/métabolisme , Érythrocytes anormaux/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/physiologie , Humains , Souris , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 1/physiologie , Mitogen-Activated Protein Kinase 3/métabolisme , Mitogen-Activated Protein Kinase 3/physiologie , Modèles biologiques , Culture de cellules primaires
5.
Circ Heart Fail ; 4(6): 770-8, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21835984

RÉSUMÉ

BACKGROUND: The angiotensin II type 1 receptor (AT1R) plays a key role in regulating cardiorenal function. Classic "unbiased" AT1R antagonists block receptor coupling to both G(αq) and ß-arrestin-mediated signals, which desensitize G-protein signaling as well as transduce G-protein-independent signals. TRV120027 is a novel ß-arrestin-biased AT1R ligand, which engages ß-arrestins while blocking G-protein signaling. At the AT1R, TRV120027 can inhibit angiotensin II-mediated vasoconstriction, whereas, through ß-arrestin coupling, increase cardiomyocyte contractility. We defined for the first time the acute cardiorenal actions of TRV120027 in healthy and heart failure (HF) canines. METHODS AND RESULTS: Healthy and HF canines (induced by tachypacing) were anesthetized. After instrumentation and equilibration, a 30-minute baseline clearance was performed, followed by further clearance with escalating doses of intravenous TRV120027 (0.01, 0.1, 1, 10, and 100 µg/kg per minute) and a 30-minute washout. In healthy canines, TRV120027 decreased pulmonary capillary wedge pressure and systemic and renal vascular resistances, while increasing cardiac output, renal blood flow, glomerular filtration rate, and urinary sodium excretion. In HF canines, TRV120027 decreased mean arterial pressure, right atrial pressure, and pulmonary capillary wedge pressure, systemic and renal vascular resistances and increased cardiac output and renal blood flow. Glomerular filtration rate and urinary sodium excretion were maintained. CONCLUSIONS: We report for the first time the cardiorenal actions of the novel ß-arrestin-biased AT1R ligand TRV120027. In both normal and HF canines, TRV120027 demonstrated cardiac unloading actions while preserving renal function. With this beneficial pharmacological profile, TRV120027 represents a novel strategy for the treatment of HF.


Sujet(s)
Arrestines/métabolisme , Défaillance cardiaque/traitement médicamenteux , Coeur/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Oligopeptides/pharmacologie , Oligopeptides/usage thérapeutique , Récepteur de type 1 à l'angiotensine-II/métabolisme , Animaux , Pression sanguine/effets des médicaments et des substances chimiques , Pression sanguine/physiologie , Débit cardiaque/effets des médicaments et des substances chimiques , Débit cardiaque/physiologie , Modèles animaux de maladie humaine , Chiens , Relation dose-effet des médicaments , Débit de filtration glomérulaire/effets des médicaments et des substances chimiques , Débit de filtration glomérulaire/physiologie , Coeur/physiopathologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/physiopathologie , Rein/physiopathologie , Ligands , Mâle , Sodium/urine , Résultat thérapeutique , Résistance vasculaire/effets des médicaments et des substances chimiques , Résistance vasculaire/physiologie , bêta-Arrestines
6.
Nature ; 477(7364): 349-53, 2011 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-21857681

RÉSUMÉ

The human mind and body respond to stress, a state of perceived threat to homeostasis, by activating the sympathetic nervous system and secreting the catecholamines adrenaline and noradrenaline in the 'fight-or-flight' response. The stress response is generally transient because its accompanying effects (for example, immunosuppression, growth inhibition and enhanced catabolism) can be harmful in the long term. When chronic, the stress response can be associated with disease symptoms such as peptic ulcers or cardiovascular disorders, and epidemiological studies strongly indicate that chronic stress leads to DNA damage. This stress-induced DNA damage may promote ageing, tumorigenesis, neuropsychiatric conditions and miscarriages. However, the mechanisms by which these DNA-damage events occur in response to stress are unknown. The stress hormone adrenaline stimulates ß(2)-adrenoreceptors that are expressed throughout the body, including in germline cells and zygotic embryos. Activated ß(2)-adrenoreceptors promote Gs-protein-dependent activation of protein kinase A (PKA), followed by the recruitment of ß-arrestins, which desensitize G-protein signalling and function as signal transducers in their own right. Here we elucidate a molecular mechanism by which ß-adrenergic catecholamines, acting through both Gs-PKA and ß-arrestin-mediated signalling pathways, trigger DNA damage and suppress p53 levels respectively, thus synergistically leading to the accumulation of DNA damage. In mice and in human cell lines, ß-arrestin-1 (ARRB1), activated via ß(2)-adrenoreceptors, facilitates AKT-mediated activation of MDM2 and also promotes MDM2 binding to, and degradation of, p53, by acting as a molecular scaffold. Catecholamine-induced DNA damage is abrogated in Arrb1-knockout (Arrb1(-/-)) mice, which show preserved p53 levels in both the thymus, an organ that responds prominently to acute or chronic stress, and in the testes, in which paternal stress may affect the offspring's genome. Our results highlight the emerging role of ARRB1 as an E3-ligase adaptor in the nucleus, and reveal how DNA damage may accumulate in response to chronic stress.


Sujet(s)
Arrestines/métabolisme , Altération de l'ADN , Récepteurs bêta-2 adrénergiques/métabolisme , Stress physiologique/physiologie , Animaux , Arrestines/déficit , Arrestines/génétique , Catécholamines/pharmacologie , Lignée cellulaire , Noyau de la cellule/enzymologie , Noyau de la cellule/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Fibroblastes , Humains , Mâle , Souris , Souris de lignée C57BL , Maturation post-traductionnelle des protéines/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Testicule/métabolisme , Thymus (glande)/métabolisme , Protéine p53 suppresseur de tumeur/composition chimique , Protéine p53 suppresseur de tumeur/métabolisme , bêta-Arrestine 1 , bêta-Arrestines
7.
Trends Mol Med ; 17(3): 126-39, 2011 Mar.
Article de Anglais | MEDLINE | ID: mdl-21183406

RÉSUMÉ

Members of the seven-transmembrane receptor (7TMR), or G protein-coupled receptor (GPCR), superfamily represent some of the most successful targets of modern drug therapy, with proven efficacy in the treatment of a broad range of human conditions and disease processes. It is now appreciated that ß-arrestins, once viewed simply as negative regulators of traditional 7TMR-stimulated G protein signaling, act as multifunctional adapter proteins that regulate 7TMR desensitization and trafficking and promote distinct intracellular signals in their own right. Moreover, several 7TMR biased agonists, which selectively activate these divergent signaling pathways, have been identified. Here we highlight the diversity of G protein- and ß-arrestin-mediated functions and the therapeutic potential of selective targeting of these in disease states.


Sujet(s)
Arrestines/agonistes , Arrestines/usage thérapeutique , Traitement médicamenteux , Protéines G/agonistes , Protéines G/usage thérapeutique , Animaux , Humains , Transduction du signal , bêta-Arrestines
8.
J Pharmacol Exp Ther ; 335(3): 572-9, 2010 Dec.
Article de Anglais | MEDLINE | ID: mdl-20801892

RÉSUMÉ

Biased G protein-coupled receptor ligands engage subsets of the receptor signals normally stimulated by unbiased agonists. However, it is unclear whether ligand bias can elicit differentiated pharmacology in vivo. Here, we describe the discovery of a potent, selective ß-arrestin biased ligand of the angiotensin II type 1 receptor. TRV120027 (Sar-Arg-Val-Tyr-Ile-His-Pro-D-Ala-OH) competitively antagonizes angiotensin II-stimulated G protein signaling, but stimulates ß-arrestin recruitment and activates several kinase pathways, including p42/44 mitogen-activated protein kinase, Src, and endothelial nitric-oxide synthase phosphorylation via ß-arrestin coupling. Consistent with ß-arrestin efficacy, and unlike unbiased antagonists, TRV120027 increased cardiomyocyte contractility in vitro. In rats, TRV120027 reduced mean arterial pressure, as did the unbiased antagonists losartan and telmisartan. However, unlike the unbiased antagonists, which decreased cardiac performance, TRV120027 increased cardiac performance and preserved cardiac stroke volume. These striking differences in vivo between unbiased and ß-arrestin biased ligands validate the use of biased ligands to selectively target specific receptor functions in drug discovery.


Sujet(s)
Angiotensine-II/analogues et dérivés , Angiotensine-II/pharmacologie , Arrestines/métabolisme , Pression sanguine/effets des médicaments et des substances chimiques , Phénomènes physiologiques cardiovasculaires/effets des médicaments et des substances chimiques , Récepteur de type 1 à l'angiotensine-II/agonistes , Transduction du signal/effets des médicaments et des substances chimiques , Angiotensine-II/métabolisme , Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Animaux , Arrestines/génétique , Fixation compétitive , Lignée cellulaire tumorale , Interactions médicamenteuses , Extracellular Signal-Regulated MAP Kinases/métabolisme , Focal adhesion kinase 1/métabolisme , Protéines G/métabolisme , Cellules HEK293 , Humains , Mâle , Souris , Contraction myocardique/effets des médicaments et des substances chimiques , Contraction myocardique/physiologie , Myocytes cardiaques/cytologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Nitric oxide synthase type III/métabolisme , Oligopeptides/métabolisme , Oligopeptides/pharmacologie , Protéines proto-oncogènes c-jun/métabolisme , Petit ARN interférent/génétique , Rats , Récepteur de type 1 à l'angiotensine-II/génétique , Transduction du signal/physiologie , Transfection , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Fonction ventriculaire gauche/physiologie , bêta-Arrestines , src-Family kinases/métabolisme
9.
Sci Signal ; 2(78): ra33, 2009 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-19584355

RÉSUMÉ

Agonist-induced ubiquitylation and degradation of heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) play an essential role in surface receptor homeostasis, thereby tuning many physiological processes. Although beta-arrestin and affiliated E3 ligases mediate agonist-stimulated lysosomal degradation of the beta(2)-adrenergic receptor (beta(2)AR), a prototypic GPCR, the molecular cues that mark receptors for ubiquitylation and the regulation of receptor degradation by the proteasome remain poorly understood. We show that the von Hippel-Lindau tumor suppressor protein (pVHL)-E3 ligase complex, known for its regulation of hypoxia-inducible factor (HIF) proteins, interacts with and ubiquitylates the beta(2)AR, thereby decreasing receptor abundance. We further show that the interaction of pVHL with beta(2)AR is dependent on proline hydroxylation (proline-382 and -395) and that the dioxygenase EGLN3 interacts directly with the beta(2)AR to serve as an endogenous beta(2)AR prolyl hydroxylase. Under hypoxic conditions, receptor hydroxylation and subsequent ubiquitylation decrease dramatically, thus attenuating receptor degradation and down-regulation. Notably, in both cells and tissue, the abundance of endogenous beta(2)AR is shown to reflect constitutive turnover by EGLN3 and pVHL. Our findings provide insight into GPCR regulation, broaden the functional scope of prolyl hydroxylation, and expand our understanding of the cellular response to hypoxia.


Sujet(s)
Dioxygenases/physiologie , Oxygène/physiologie , Récepteurs bêta-2 adrénergiques/métabolisme , Protéine Von Hippel-Lindau supresseur de tumeur/physiologie , Lignée cellulaire , Régulation négative , Humains , Hydroxylation , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Hypoxia-inducible factor-proline dioxygenases , Oxygène/pharmacologie , Procollagen-Proline Dioxygenase/métabolisme , Proline/métabolisme , Liaison aux protéines , Ubiquitination
10.
Proc Natl Acad Sci U S A ; 106(24): 9649-54, 2009 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-19497875

RÉSUMÉ

CCL19 and CCL21 are endogenous agonists for the seven-transmembrane receptor CCR7. They are equally active in promoting G protein stimulation and chemotaxis. Yet, we find that they result in striking differences in activation of the G protein-coupled receptor kinase (GRK)/ss-arrestin system. CCL19 leads to robust CCR7 phosphorylation and beta-arrestin2 recruitment catalyzed by both GRK3 and GRK6 whereas CCL21 activates GRK6 alone. This differential GRK activation leads to distinct functional consequences. Although each ligand leads to beta-arrestin2 recruitment, only CCL19 leads to redistribution of beta-arrestin2-GFP into endocytic vesicles and classical receptor desensitization. In contrast, these agonists are both capable of signaling through GRK6 and beta-arrestin2 to ERK kinases. Thus, this mechanism for "ligand bias" whereby endogenous agonists activate different GRK isoforms leads to functionally distinct pools of beta-arrestin.


Sujet(s)
Kinases associées à des récepteurs couplés à une protéine G/métabolisme , Arrestines/métabolisme , Lignée cellulaire , Humains , Ligands , Mitogen-Activated Protein Kinases/métabolisme , Phosphorylation , Petit ARN interférent , Récepteurs CCR7/métabolisme , Transduction du signal , bêta-Arrestines
11.
J Clin Invest ; 119(5): 1312-21, 2009 May.
Article de Anglais | MEDLINE | ID: mdl-19349687

RÉSUMÉ

Nicotinic acid is one of the most effective agents for both lowering triglycerides and raising HDL. However, the side effect of cutaneous flushing severely limits patient compliance. As nicotinic acid stimulates the GPCR GPR109A and Gi/Go proteins, here we dissected the roles of G proteins and the adaptor proteins, beta-arrestins, in nicotinic acid-induced signaling and physiological responses. In a human cell line-based signaling assay, nicotinic acid stimulation led to pertussis toxin-sensitive lowering of cAMP, recruitment of beta-arrestins to the cell membrane, an activating conformational change in beta-arrestin, and beta-arrestin-dependent signaling to ERK MAPK. In addition, we found that nicotinic acid promoted the binding of beta-arrestin1 to activated cytosolic phospholipase A2 as well as beta-arrestin1-dependent activation of cytosolic phospholipase A2 and release of arachidonate, the precursor of prostaglandin D2 and the vasodilator responsible for the flushing response. Moreover, beta-arrestin1-null mice displayed reduced cutaneous flushing in response to nicotinic acid, although the improvement in serum free fatty acid levels was similar to that observed in wild-type mice. These data suggest that the adverse side effect of cutaneous flushing is mediated by beta-arrestin1, but lowering of serum free fatty acid levels is not. Furthermore, G protein-biased ligands that activate GPR109A in a beta-arrestin-independent fashion may represent an improved therapeutic option for the treatment of dyslipidemia.


Sujet(s)
Arrestines/métabolisme , Rougeur de la face/métabolisme , Lipolyse/effets des médicaments et des substances chimiques , Acide nicotinique/pharmacologie , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Animaux , Arrestines/composition chimique , Arrestines/génétique , AMP cyclique/métabolisme , Oreille/vascularisation , Éicosanoïdes/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Acide gras libre/sang , Rougeur de la face/induit chimiquement , Humains , Cellules de Langerhans/effets des médicaments et des substances chimiques , Cellules de Langerhans/métabolisme , Lipolyse/physiologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Agonistes nicotiniques/pharmacologie , Cytosolic phospholipases A2/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Conformation des protéines/effets des médicaments et des substances chimiques , Pyrazoles/pharmacologie , Récepteurs couplés aux protéines G/agonistes , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Récepteurs nicotiniques/génétique , Récepteurs nicotiniques/métabolisme , Débit sanguin régional/effets des médicaments et des substances chimiques , Tétrazoles/pharmacologie , bêta-Arrestines
12.
Proc Natl Acad Sci U S A ; 105(38): 14555-60, 2008 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-18787115

RÉSUMÉ

Recent evidence suggests that binding of agonist to its cognate receptor initiates not only classical G protein-mediated signaling, but also beta-arrestin-dependent signaling. One such beta-arrestin-mediated pathway uses the beta(1)-adrenergic receptor (beta(1)AR) to transactivate the EGFR. To determine whether beta-adrenergic ligands that do not activate G protein signaling (i.e., beta-blockers) can stabilize the beta(1)AR in a signaling conformation, we screened 20 beta-blockers for their ability to stimulate beta-arrestin-mediated EGFR transactivation. Here we show that only alprenolol (Alp) and carvedilol (Car) induce beta(1)AR-mediated transactivation of the EGFR and downstream ERK activation. By using mutants of the beta(1)AR lacking G protein-coupled receptor kinase phosphorylation sites and siRNA directed against beta-arrestin, we show that Alp- and Car-stimulated EGFR transactivation requires beta(1)AR phosphorylation at consensus G protein-coupled receptor kinase sites and beta-arrestin recruitment to the ligand-occupied receptor. Moreover, pharmacological inhibition of Src and EGFR blocked Alp- and Car-stimulated EGFR transactivation. Our findings demonstrate that Alp and Car are ligands that not only act as classical receptor antagonists, but can also stimulate signaling pathways in a G protein-independent, beta-arrestin-dependent fashion.


Sujet(s)
Antagonistes bêta-adrénergiques/pharmacologie , Alprénolol/pharmacologie , Arrestines/métabolisme , Carbazoles/pharmacologie , Récepteurs ErbB/métabolisme , Propanolamines/pharmacologie , Activation de la transcription/effets des médicaments et des substances chimiques , Animaux , Carvédilol , Lignée de cellules transformées , Relation dose-effet des médicaments , Activation enzymatique/effets des médicaments et des substances chimiques , Enzymes/métabolisme , Chlorhydrate d'erlotinib , Gènes erbB-1/génétique , Coeur/effets des médicaments et des substances chimiques , Humains , Souris , Souris de lignée C57BL , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Quinazolines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , bêta-Arrestines
13.
Mol Cell ; 31(3): 395-405, 2008 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-18691971

RÉSUMÉ

Signal transduction through G protein-coupled receptors (GPCRs) is regulated by receptor desensitization and internalization that follow agonist stimulation. Nitric oxide (NO) can influence these processes, but the cellular source of NO bioactivity and the effects of NO on GPCR-mediated signal transduction are incompletely understood. Here, we show in cells and mice that beta-arrestin 2, a central element in GPCR trafficking, interacts with and is S-nitrosylated at a single cysteine by endothelial NO synthase (eNOS), and that S-nitrosylation of beta-arrestin 2 is promoted by endogenous S-nitrosogluthathione. S-nitrosylation after agonist stimulation of the beta-adrenergic receptor, a prototypical GPCR, dissociates eNOS from beta-arrestin 2 and promotes binding of beta-arrestin 2 to clathrin heavy chain/beta-adaptin, thereby accelerating receptor internalization. The agonist- and NO-dependent shift in the affiliations of beta-arrestin 2 is followed by denitrosylation. Thus, beta-arrestin subserves the functional coupling of eNOS and GPCRs, and dynamic S-nitrosylation/denitrosylation of beta-arrestin 2 regulates stimulus-induced GPCR trafficking.


Sujet(s)
Arrestines/métabolisme , Composés nitrosés/métabolisme , Récepteurs bêta-2 adrénergiques/métabolisme , Complexe protéique adaptateur 2/métabolisme , Animaux , Bovins , Lignée cellulaire , Clathrine/métabolisme , Cystéine/analogues et dérivés , Cystéine/métabolisme , Endocytose , Humains , Ligands , Souris , Modèles biologiques , Nitric oxide synthase type III/métabolisme , Liaison aux protéines , Transport des protéines , S-Nitrosothiols/métabolisme , bêta-Arrestine 2 , bêta-Arrestines
14.
Proc Natl Acad Sci U S A ; 105(29): 9988-93, 2008 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-18621717

RÉSUMÉ

Beta-arrestins critically regulate G protein-coupled receptors (GPCRs), also known as seven-transmembrane receptors (7TMRs), both by inhibiting classical G protein signaling and by initiating distinct beta-arrestin-mediated signaling. The recent discovery of beta-arrestin-biased ligands and receptor mutants has allowed characterization of these independent "G protein-mediated" and "beta-arrestin-mediated" signaling mechanisms of 7TMRs. However, the molecular mechanisms underlying the dual functions of beta-arrestins remain unclear. Here, using an intramolecular BRET (bioluminescence resonance energy transfer)-based biosensor of beta-arrestin 2 and a combination of biased ligands and/or biased mutants of three different 7TMRs, we provide evidence that beta-arrestin can adopt multiple "active" conformations. Surprisingly, phosphorylation-deficient mutants of the receptors are also capable of directing similar conformational changes in beta-arrestin as is the wild-type receptor. This indicates that distinct receptor conformations induced and/or stabilized by different ligands can promote distinct and functionally specific conformations in beta-arrestin even in the absence of receptor phosphorylation. Our data thus highlight another interesting aspect of 7TMR signaling--i.e., functionally specific receptor conformations can be translated to downstream effectors such as beta-arrestins, thereby governing their functional specificity.


Sujet(s)
Arrestines/composition chimique , Récepteurs couplés aux protéines G/composition chimique , Arrestines/génétique , Arrestines/métabolisme , Phénomènes biophysiques , Biophysique , Techniques de biocapteur , Cellules , Transfert d'énergie par résonance de fluorescence , Humains , Ligands , Mutagenèse dirigée , Phosphorylation , Conformation des protéines , Récepteur de type 1 à l'angiotensine-II/agonistes , Récepteur de type 1 à l'angiotensine-II/composition chimique , Récepteur de type 1 à l'angiotensine-II/génétique , Récepteurs couplés aux protéines G/agonistes , Récepteurs couplés aux protéines G/génétique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , bêta-Arrestine 2 , bêta-Arrestines
15.
J Biol Chem ; 283(30): 21093-101, 2008 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-18534983

RÉSUMÉ

Members of the seven-transmembrane receptor (7TMR) superfamily are sequestered from the plasma membrane following stimulation both to limit cellular responses as well as to initiate novel G protein-independent signaling pathways. The best studied mechanism for 7TMR internalization is via clathrin-coated pits, where clathrin and adaptor protein complex 2 nucleate and polymerize upon encountering the membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PIP(2)) to form the outer layer of the clathrin-coated vesicle. Activated receptors are recruited to clathrin-coated pits by beta-arrestins, scaffolding proteins that interact with agonist-occupied 7TMRs as well as adaptor protein complex 2 and clathrin. We report here that following stimulation of the beta2-adrenergic receptor (beta2-AR), a prototypical 7TMR, beta-arrestins bind phosphatidylinositol 4-phosphate 5-kinase (PIP5K) Ialpha, a PIP(2)-producing enzyme. Furthermore, beta-arrestin2 is required to form a complex with PIP5K Ialpha and agonist-occupied beta2-AR, and beta-arrestins synergize with the kinase to produce PIP(2) in response to isoproterenol stimulation. Interestingly, beta-arrestins themselves bind PIP(2), and a beta-arrestin mutant deficient in PIP(2) binding no longer internalizes 7TMRs, fails to interact with PIP5K Ialpha, and is not associated with PIP kinase activity assayed in vitro. However, a chimeric protein in which the core kinase domain of PIP5K Ialpha has been fused to the same beta-arrestin mutant rescues internalization of beta2-ARs. Collectively, these data support a model in which beta-arrestins direct the localization of PIP5K Ialpha and PIP(2) production to agonist-activated 7TMRs, thereby regulating receptor internalization.


Sujet(s)
Arrestines/composition chimique , Endocytose , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Récepteurs bêta-2 adrénergiques/métabolisme , Animaux , Arrestines/métabolisme , Lignée cellulaire , Membrane cellulaire/métabolisme , Clathrine/composition chimique , Humains , Modèles biologiques , Mutation , Plasmides/métabolisme , Liaison aux protéines , Conformation des protéines , Protéines de fusion recombinantes/composition chimique , bêta-Arrestines
16.
Science ; 320(5884): 1777-81, 2008 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-18497258

RÉSUMÉ

beta-Arrestins have important roles in the regulation of seven-transmembrane receptors (7TMRs). Smoothened (Smo) is a 7TMR that mediates effects of Hedgehog on developmental processes and whose dysregulation may cause tumorigenesis. beta-Arrestins are required for endocytosis of Smo and signaling to Gli transcription factors. In mammalian cells, Smo-dependent signaling requires translocation to primary cilia. We demonstrated that beta-arrestins mediate the activity-dependent interaction of Smo and the kinesin motor protein Kif3A. This multimeric complex localized to primary cilia and was disrupted in cells transfected with beta-arrestin small interfering RNA. beta-Arrestin 1 or beta-arrestin 2 depletion prevented the localization of Smo to primary cilia and the Smo-dependent activation of Gli. These results suggest roles for beta-arrestins in mediating the intracellular transport of a 7TMR to its obligate subcellular location for signaling.


Sujet(s)
Arrestines/métabolisme , Cils vibratiles/métabolisme , Kinésine/métabolisme , Moteurs moléculaires/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Animaux , Arrestines/génétique , Protéines Hedgehog/métabolisme , Souris , Microscopie confocale , Cellules NIH 3T3 , Transport des protéines , Interférence par ARN , Protéines de fusion recombinantes/métabolisme , Transduction du signal , Récepteur Smoothened , Facteurs de transcription/métabolisme , Transfection , bêta-Arrestine 1 , bêta-Arrestine 2 , bêta-Arrestines
17.
J Biol Chem ; 283(16): 10611-20, 2008 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-18276584

RÉSUMÉ

Seven transmembrane receptors (7TMRs) exert strong regulatory influences on virtually all physiological processes. Although it is historically assumed that heterotrimeric G proteins mediate these actions, there is a newer appreciation that beta-arrestins, originally thought only to desensitize G protein signaling, also serve as independent receptor signal transducers. Recently, we found that activation of ERK1/2 by the angiotensin receptor occurs via both of these distinct pathways. In this work, we explore the physiological consequences of beta-arrestin ERK1/2 signaling and delineate a pathway that regulates mRNA translation and protein synthesis via Mnk1, a protein that both physically interacts with and is activated by beta-arrestins. We show that beta-arrestin-dependent activation of ERK1/2, Mnk1, and eIF4E are responsible for increasing translation rates in both human embryonic kidney 293 and rat vascular smooth muscle cells. This novel demonstration that beta-arrestins regulate protein synthesis reveals that the spectrum of beta-arrestin-mediated signaling events is broader than previously imagined.


Sujet(s)
Arrestines/métabolisme , Animaux , Cellules COS , Chlorocebus aethiops , Activation enzymatique , Cellules HeLa , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Mâle , Souris , Souris de lignée C57BL , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Muscles lisses vasculaires/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Rats , Rat Sprague-Dawley , bêta-Arrestines
18.
J Biol Chem ; 283(9): 5669-76, 2008 Feb 29.
Article de Anglais | MEDLINE | ID: mdl-18086673

RÉSUMÉ

Classically, the beta 2-adrenergic receptor (beta 2AR) and other members of the seven-transmembrane receptor (7TMR) superfamily activate G protein-dependent signaling pathways in response to ligand stimulus. It has recently been discovered, however, that a number of 7TMRs, including beta 2AR, can signal via beta-arrestin-dependent pathways independent of G protein activation. It is currently unclear if among beta 2AR agonists there exist ligands that disproportionately signal via G proteins or beta-arrestins and are hence "biased." Using a variety of approaches that include highly sensitive fluorescence resonance energy transfer-based methodologies, including a novel assay for receptor internalization, we show that the majority of known beta 2AR agonists exhibit relative efficacies for beta-arrestin-associated activities (beta-arrestin membrane translocation and beta 2AR internalization) identical to the irrelative efficacies for G protein-dependent signaling (cyclic AMP generation). However, for three betaAR ligands there is a marked bias toward beta-arrestin signaling; these ligands stimulate beta-arrestin-dependent receptor activities to a much greater extent than would be expected given their efficacy for G protein-dependent activity. Structural comparison of these biased ligands reveals that all three are catecholamines containing an ethyl substitution on the alpha-carbon, a motif absent on all of the other, unbiased ligands tested. Thus, these studies demonstrate the potential for developing a novel class of 7TMR ligands with a distinct bias for beta-arrestin-mediated signaling.


Sujet(s)
Agonistes adrénergiques/pharmacologie , Agonistes des récepteurs béta-2 adrénergiques , Arrestines/métabolisme , Membrane cellulaire/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Agonistes adrénergiques/composition chimique , Lignée cellulaire , AMP cyclique/métabolisme , Transfert d'énergie par résonance de fluorescence , Protéines G/génétique , Protéines G/métabolisme , Humains , Ligands , Transport des protéines/effets des médicaments et des substances chimiques , Récepteurs bêta-2 adrénergiques/métabolisme , Transduction du signal/physiologie , bêta-Arrestines
19.
Proc Natl Acad Sci U S A ; 104(42): 16657-62, 2007 Oct 16.
Article de Anglais | MEDLINE | ID: mdl-17925438

RÉSUMÉ

For many years, beta-adrenergic receptor antagonists (beta-blockers or betaAR antagonists) have provided significant morbidity and mortality benefits in patients who have sustained acute myocardial infarction. More recently, beta-adrenergic receptor antagonists have been found to provide survival benefits in patients suffering from heart failure, although the efficacy of different beta-blockers varies widely in this condition. One drug, carvedilol, a nonsubtype-selective betaAR antagonist, has proven particularly effective in the treatment of heart failure, although the mechanism(s) responsible for this are controversial. Here, we report that among 16 clinically relevant betaAR antagonists, carvedilol displays a unique profile of in vitro signaling characteristics. We observed that in beta2 adrenergic receptor (beta2AR)-expressing HEK-293 cells, carvedilol has inverse efficacy for stimulating G(s)-dependent adenylyl cyclase but, nonetheless, stimulates (i) phosphorylation of the receptor's cytoplasmic tail on previously documented G protein-coupled receptor kinase sites; (ii) recruitment of beta-arrestin to the beta2AR; (iii) receptor internalization; and (iv) activation of extracellular regulated kinase 1/2 (ERK 1/2), which is maintained in the G protein-uncoupled mutant beta2AR(T68F,Y132G,Y219A) (beta2AR(TYY)) and abolished by beta-arrestin2 siRNA. Taken together, these data indicate that carvedilol is able to stabilize a receptor conformation which, although uncoupled from G(s), is nonetheless able to stimulate beta-arrestin-mediated signaling. We hypothesize that such signaling may contribute to the special efficacy of carvedilol in the treatment of heart failure and may serve as a prototype for a new generation of therapeutic beta2AR ligands.


Sujet(s)
Antagonistes bêta-adrénergiques/pharmacologie , Arrestines/métabolisme , Carbazoles/pharmacologie , Propanolamines/pharmacologie , Récepteurs bêta-2 adrénergiques/métabolisme , Antagonistes des récepteurs bêta-2 adrénergiques , Arrestines/analyse , Carvédilol , Lignée cellulaire , Humains , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Phosphorylation , Récepteurs bêta-2 adrénergiques/analyse , Transduction du signal , bêta-Arrestines
20.
Blood ; 110(7): 2708-17, 2007 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-17609430

RÉSUMÉ

Sickle red cell (SS RBC) adhesion is believed to contribute to the process of vaso-occlusion in sickle cell disease (SCD). We previously found that the LW RBC adhesion receptor can be activated by epinephrine to mediate SS RBC adhesion to endothelial alphavbeta3 integrin. To determine the contribution of LW activation to vaso-occlusive events in vivo, we investigated whether in vitro treatment of SS RBCs by epinephrine resulted in vaso-occlusion in intact microvasculature after RBC infusion into nude mice. Epinephrine enhanced human SS but not normal RBC adhesion to murine endothelial cells in vitro and to endothelium in vivo, promoting vaso-occlusion and RBC organ sequestration. Murine sickle RBCs also responded to epinephrine with increased adhesion to postcapillary endothelium in nude mice. Epinephrine-induced SS RBC adhesion, vaso-occlusion, and RBC organ trapping could be prevented by the beta-adrenergic receptor (beta-AR) antagonist, propranolol. Infusion of soluble recombinant LW also significantly reduced adhesion and vaso-occlusion. In addition, epinephrine-treated SS RBCs induced activation of murine leukocyte adhesion to endothelium as well. We conclude that LW activation by epinephrine via beta-AR stimulation can promote both SS RBC and leukocyte adhesion as well as vaso-occlusion, suggesting that both epinephrine and LW play potentially pathophysiological roles in SCD.


Sujet(s)
Drépanocytose/anatomopathologie , Molécules d'adhérence cellulaire/métabolisme , Épinéphrine/pharmacologie , Drépanocytose/métabolisme , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire , Cellules cultivées , Endothélium/vascularisation , Endothélium/effets des médicaments et des substances chimiques , Endothélium/anatomopathologie , Érythrocytes/cytologie , Érythrocytes/effets des médicaments et des substances chimiques , Humains , Leucocytes/cytologie , Leucocytes/effets des médicaments et des substances chimiques , Leucocytes/immunologie , Souris , Souris nude , Récepteurs bêta-2 adrénergiques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE