Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Pharmacol Exp Ther ; 387(2): 214-225, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37643795

RESUMO

Hereditary angioedema (HAE) is a rare autosomal dominant disorder caused by a deficiency in functional C1 esterase inhibitor, a serpin family protein that blocks the activity of plasma kallikrein. Insufficient inhibition of plasma kallikrein results in the overproduction of bradykinin, a vasoactive inflammatory mediator that produces both pain and unpredictable swelling during HAE attacks, with potentially life-threatening consequences. We describe the generation of STAR-0215, a humanized IgG1 antibody with a long circulating half-life (t1/2) that potently inhibits plasma kallikrein activity, with a >1000-fold lower affinity for prekallikrein and no measurable inhibitory activity against other serine proteases. The high specificity and inhibitory effect of STAR-0215 is demonstrated through a unique allosteric mechanism involving N-terminal catalytic domain binding, destabilization of the activation domain, and reversion of the active site to the inactive zymogen state. The YTE (M252Y/S254T/T256E) modified fragment crystallizable (Fc) domain of STAR-0215 enhances pH-dependent neonatal Fc receptor binding, resulting in a prolonged t1/2 in vivo (∼34 days in cynomolgus monkeys) compared with antibodies without this modification. A single subcutaneous dose of STAR-0215 (≥100 mg) was predicted to be active in patients for 3 months or longer, based on simulations using a minimal physiologically based pharmacokinetic model. These data indicate that STAR-0215, a highly potent and specific antibody against plasma kallikrein with extended t1/2, is a potential agent for long-term preventative HAE therapy administered every 3 months or less frequently. SIGNIFICANCE STATEMENT: STAR-0215 is a YTE-modified immunoglobulin G1 monoclonal antibody with a novel binding mechanism that specifically and potently inhibits the enzymatic activity of plasma kallikrein and prevents the generation of bradykinin. It has been designed to be a long-lasting prophylactic treatment to prevent attacks of HAE and to decrease the burden of disease and the burden of treatment for people with HAE.

2.
J Pharmacol Exp Ther ; 362(2): 254-262, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28533287

RESUMO

Prescription opioids are a mainstay in the treatment of acute moderate to severe pain. However, chronic use leads to a host of adverse consequences including tolerance and opioid-induced hyperalgesia (OIH), leading to more complex treatment regimens and diminished patient compliance. Patients with OIH paradoxically experience exaggerated nociceptive responses instead of pain reduction after chronic opioid usage. The development of OIH and tolerance tend to occur simultaneously and, thus, present a challenge when studying the molecular mechanisms driving each phenomenon. We tested the hypothesis that a G protein-biased µ-opioid peptide receptor (MOPR) agonist would not induce symptoms of OIH, such as mechanical allodynia, following chronic administration. We observed that the development of opioid-induced mechanical allodynia (OIMA), a model of OIH, was absent in ß-arrestin1-/- and ß-arrestin2-/- mice in response to chronic administration of conventional opioids such as morphine, oxycodone and fentanyl, whereas tolerance developed independent of OIMA. In agreement with the ß-arrestin knockout mouse studies, chronic administration of TRV0109101, a G protein-biased MOPR ligand and structural analog of oliceridine, did not promote the development of OIMA but did result in drug tolerance. Interestingly, following induction of OIMA by morphine or fentanyl, TRV0109101 was able to rapidly reverse allodynia. These observations establish a role for ß-arrestins in the development of OIH, independent of tolerance, and suggest that the use of G protein-biased MOPR ligands, such as oliceridine and TRV0109101, may be an effective therapeutic avenue for managing chronic pain with reduced propensity for opioid-induced hyperalgesia.


Assuntos
Analgésicos Opioides/administração & dosagem , Proteínas de Ligação ao GTP/agonistas , Hiperalgesia/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Receptores Opioides mu/agonistas , Animais , Relação Dose-Resposta a Droga , Esquema de Medicação , Proteínas de Ligação ao GTP/fisiologia , Células HEK293 , Humanos , Hiperalgesia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Medição da Dor/métodos , Receptores Opioides mu/fisiologia
3.
J Biol Chem ; 289(20): 14211-24, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24668815

RESUMO

The concept of "biased agonism" arises from the recognition that the ability of an agonist to induce a receptor-mediated response (i.e. "efficacy") can differ across the multiple signal transduction pathways (e.g. G protein and ß-arrestin (ßarr)) emanating from a single GPCR. Despite the therapeutic promise of biased agonism, the molecular mechanism(s) whereby biased agonists selectively engage signaling pathways remain elusive. This is due in large part to the challenges associated with quantifying ligand efficacy in cells. To address this, we developed a cell-free approach to directly quantify the transducer-specific molecular efficacies of balanced and biased ligands for the angiotensin II type 1 receptor (AT1R), a prototypic GPCR. Specifically, we defined efficacy in allosteric terms, equating shifts in ligand affinity (i.e. KLo/KHi) at AT1R-Gq and AT1R-ßarr2 fusion proteins with their respective molecular efficacies for activating Gq and ßarr2. Consistent with ternary complex model predictions, transducer-specific molecular efficacies were strongly correlated with cellular efficacies for activating Gq and ßarr2. Subsequent comparisons across transducers revealed that biased AT1R agonists possess biased molecular efficacies that were in strong agreement with the signaling bias observed in cellular assays. These findings not only represent the first measurements of the thermodynamic driving forces underlying differences in ligand efficacy between transducers but also support a molecular mechanism whereby divergent transducer-specific molecular efficacies generate biased agonism at a GPCR.


Assuntos
Receptor Tipo 1 de Angiotensina/agonistas , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais , Regulação Alostérica , Células HEK293 , Humanos , Ligantes , Proteínas Recombinantes de Fusão/metabolismo , Termodinâmica
4.
Am J Physiol Heart Circ Physiol ; 305(6): H856-66, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23873795

RESUMO

In the present study, we compared the cardioprotective effects of TRV120023, a novel angiotensin II (ANG II) type 1 receptor (AT1R) ligand, which blocks G protein coupling but stimulates ß-arrestin signaling, against treatment with losartan, a conventional AT1R blocker in the treatment of cardiac hypertrophy and regulation of myofilament activity and phosphorylation. Rats were subjected to 3 wk of treatment with saline, ANG II, ANG II + losartan, ANG II + TRV120023, or TRV120023 alone. ANG II induced increased left ventricular mass compared with rats that received ANG II + losartan or ANG II + TRV120023. Compared with saline controls, ANG II induced a significant increase in pCa50 and maximum Ca(2+)-activated myofilament tension but reduced the Hill coefficient (nH). TRV120023 increased maximum tension and pCa50, although to lesser extent than ANG II. In contrast to ANG II, TRV120023 increased nH. Losartan blocked the effects of ANG II on pCa50 and nH and reduced maximum tension below that of saline controls. ANG II + TRV120023 showed responses similar to those of TRV120023 alone; compared with ANG II + losartan, ANG II + TRV120023 preserved maximum tension and increased both pCa50 and cooperativity. Tropomyosin phosphorylation was lower in myofilaments from saline-treated hearts compared with the other groups. Phosphorylation of cardiac troponin I was significantly reduced in ANG II + TRV120023 and TRV120023 groups versus saline controls, and myosin-binding protein C phosphorylation at Ser(282) was unaffected by ANG II or losartan but significantly reduced with TRV120023 treatment compared with all other groups. Our data indicate that TRV120023-related promotion of ß-arrestin signaling and enhanced contractility involves a mechanism promoting the myofilament response to Ca(2+) via altered protein phosphorylation. Selective activation of ß-arrestin-dependent pathways may provide advantages over conventional AT1R blockers.


Assuntos
Arrestinas/metabolismo , Cálcio/metabolismo , Cardiomegalia/tratamento farmacológico , Cardiomegalia/fisiopatologia , Ventrículos do Coração/fisiopatologia , Miofibrilas/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Angiotensina II , Animais , Cardiomegalia/induzido quimicamente , Cardiotônicos/administração & dosagem , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Masculino , Contração Miocárdica/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos , beta-Arrestinas
5.
J Pharmacol Exp Ther ; 344(3): 708-17, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23300227

RESUMO

The concept of ligand bias at G protein-coupled receptors broadens the possibilities for agonist activities and provides the opportunity to develop safer, more selective therapeutics. Morphine pharmacology in ß-arrestin-2 knockout mice suggested that a ligand that promotes coupling of the µ-opioid receptor (MOR) to G proteins, but not ß-arrestins, would result in higher analgesic efficacy, less gastrointestinal dysfunction, and less respiratory suppression than morphine. Here we report the discovery of TRV130 ([(3-methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethyl})amine), a novel MOR G protein-biased ligand. In cell-based assays, TRV130 elicits robust G protein signaling, with potency and efficacy similar to morphine, but with far less ß-arrestin recruitment and receptor internalization. In mice and rats, TRV130 is potently analgesic while causing less gastrointestinal dysfunction and respiratory suppression than morphine at equianalgesic doses. TRV130 successfully translates evidence that analgesic and adverse MOR signaling pathways are distinct into a biased ligand with differentiated pharmacology. These preclinical data suggest that TRV130 may be a safer and more tolerable therapeutic for treating severe pain.


Assuntos
Analgésicos/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Trato Gastrointestinal/efeitos dos fármacos , Morfina/farmacologia , Receptores Opioides mu/metabolismo , Sistema Respiratório/efeitos dos fármacos , Animais , Arrestinas/metabolismo , Linhagem Celular , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/tratamento farmacológico , Gastroenteropatias/metabolismo , Células HEK293 , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Doenças Respiratórias/induzido quimicamente , Doenças Respiratórias/tratamento farmacológico , Doenças Respiratórias/metabolismo , Transdução de Sinais/efeitos dos fármacos , beta-Arrestina 2 , beta-Arrestinas
6.
Mol Syst Biol ; 8: 590, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22735336

RESUMO

Seven-transmembrane receptors (7TMRs) are involved in nearly all aspects of chemical communications and represent major drug targets. 7TMRs transmit their signals not only via heterotrimeric G proteins but also through ß-arrestins, whose recruitment to the activated receptor is regulated by G protein-coupled receptor kinases (GRKs). In this paper, we combined experimental approaches with computational modeling to decipher the molecular mechanisms as well as the hidden dynamics governing extracellular signal-regulated kinase (ERK) activation by the angiotensin II type 1A receptor (AT(1A)R) in human embryonic kidney (HEK)293 cells. We built an abstracted ordinary differential equations (ODE)-based model that captured the available knowledge and experimental data. We inferred the unknown parameters by simultaneously fitting experimental data generated in both control and perturbed conditions. We demonstrate that, in addition to its well-established function in the desensitization of G-protein activation, GRK2 exerts a strong negative effect on ß-arrestin-dependent signaling through its competition with GRK5 and 6 for receptor phosphorylation. Importantly, we experimentally confirmed the validity of this novel GRK2-dependent mechanism in both primary vascular smooth muscle cells naturally expressing the AT(1A)R, and HEK293 cells expressing other 7TMRs.


Assuntos
Arrestinas/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinases de Receptores Acoplados a Proteína G/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Modelos Biológicos , Transdução de Sinais , Linhagem Celular , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Humanos , Rim/citologia , Rim/embriologia , Rim/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , beta-Arrestinas
7.
Circ Res ; 109(2): 205-16, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21737816

RESUMO

Drug discovery efforts targeting G-protein-coupled receptors (GPCR) have been immensely successful in creating new cardiovascular medicines. Currently marketed GPCR drugs are broadly classified as either agonists that activate receptors or antagonists that prevent receptor activation by endogenous stimuli. However, GPCR couple to a multitude of intracellular signaling pathways beyond classical G-protein signals, and these signals can be independently activated by biased ligands to vastly expand the potential for new drugs at these classic targets. By selectively engaging only a subset of a receptor's potential intracellular partners, biased ligands may deliver more precise therapeutic benefit with fewer side effects than current GPCR-targeted drugs. In this review, we discuss the history of biased ligand research, the current understanding of how biased ligands exert their unique pharmacology, and how research into GPCR signaling has uncovered previously unappreciated capabilities of receptor pharmacology. We focus on several receptors to illustrate the approaches taken and discoveries made, and how these are steadily illuminating the intricacies of GPCR pharmacology. Discoveries of biased ligands targeting the angiotensin II type 1 receptor and of separable pharmacology suggesting the potential value of biased ligands targeting the ß-adrenergic receptors and nicotinic acid receptor GPR109a highlight the powerful clinical promise of this new category of potential therapeutics.


Assuntos
Fármacos Cardiovasculares/farmacologia , Descoberta de Drogas , Terapia de Alvo Molecular/métodos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Fármacos Cardiovasculares/química , Humanos , Ligantes , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Nicotínicos/efeitos dos fármacos
8.
Am J Physiol Heart Circ Physiol ; 303(8): H1001-10, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22886417

RESUMO

Pharmacological blockade of the ANG II type 1 receptor (AT1R) is a common therapy for treatment of congestive heart failure and hypertension. Increasing evidence suggests that selective engagement of ß-arrestin-mediated AT1R signaling, referred to as biased signaling, promotes cardioprotective signaling. Here, we tested the hypothesis that a ß-arrestin-biased AT1R ligand TRV120023 would confer cardioprotection in response to acute cardiac injury compared with the traditional AT1R blocker (ARB), losartan. TRV120023 promotes cardiac contractility, assessed by pressure-volume loop analyses, while blocking the effects of endogenous ANG II. Compared with losartan, TRV120023 significantly activates MAPK and Akt signaling pathways. These hemodynamic and biochemical effects were lost in ß-arrestin-2 knockout (KO) mice. In response to cardiac injury induced by ischemia reperfusion injury or mechanical stretch, pretreatment with TRV120023 significantly diminishes cell death compared with losartan, which did not appear to be cardioprotective. This cytoprotective effect was lost in ß-arrestin-2 KO mice. The ß-arrestin-biased AT1R ligand, TRV120023, has cardioprotective and functional properties in vivo, which are distinct from losartan. Our data suggest that this novel class of drugs may provide an advantage over conventional ARBs by supporting cardiac function and reducing cellular injury during acute cardiac injury.


Assuntos
Síndrome Coronariana Aguda/tratamento farmacológico , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Arrestinas/metabolismo , Cardiotônicos/farmacologia , Losartan/farmacologia , Oligopeptídeos/farmacologia , Síndrome Coronariana Aguda/metabolismo , Síndrome Coronariana Aguda/patologia , Doença Aguda , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Arrestinas/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , beta-Arrestina 2 , beta-Arrestinas
9.
Proc Natl Acad Sci U S A ; 106(24): 9649-54, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19497875

RESUMO

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.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Arrestinas/metabolismo , Linhagem Celular , Humanos , Ligantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , RNA Interferente Pequeno , Receptores CCR7/metabolismo , Transdução de Sinais , beta-Arrestinas
10.
Mol Pharmacol ; 80(3): 367-77, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21610196

RESUMO

Seven transmembrane receptors (7TMRs), commonly referred to as G protein-coupled receptors, form a large part of the "druggable" genome. 7TMRs can signal through parallel pathways simultaneously, such as through heterotrimeric G proteins from different families, or, as more recently appreciated, through the multifunctional adapters, ß-arrestins. Biased agonists, which signal with different efficacies to a receptor's multiple downstream pathways, are useful tools for deconvoluting this signaling complexity. These compounds may also be of therapeutic use because they have distinct functional and therapeutic profiles from "balanced agonists." Although some methods have been proposed to identify biased ligands, no comparison of these methods applied to the same set of data has been performed. Therefore, at this time, there are no generally accepted methods to quantify the relative bias of different ligands, making studies of biased signaling difficult. Here, we use complementary computational approaches for the quantification of ligand bias and demonstrate their application to two well known drug targets, the ß2 adrenergic and angiotensin II type 1A receptors. The strategy outlined here allows a quantification of ligand bias and the identification of weakly biased compounds. This general method should aid in deciphering complex signaling pathways and may be useful for the development of novel biased therapeutic ligands as drugs.


Assuntos
Receptores de Superfície Celular/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Ligantes , Ensaio Radioligante , Receptor Tipo 2 de Angiotensina/metabolismo
11.
Proc Natl Acad Sci U S A ; 105(38): 14555-60, 2008 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-18787115

RESUMO

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.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Alprenolol/farmacologia , Arrestinas/metabolismo , Carbazóis/farmacologia , Receptores ErbB/metabolismo , Propanolaminas/farmacologia , Ativação Transcricional/efeitos dos fármacos , Animais , Carvedilol , Linhagem Celular Transformada , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Enzimas/metabolismo , Cloridrato de Erlotinib , Genes erbB-1/genética , Coração/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , beta-Arrestinas
12.
Proc Natl Acad Sci U S A ; 105(29): 9988-93, 2008 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-18621717

RESUMO

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.


Assuntos
Arrestinas/química , Receptores Acoplados a Proteínas G/química , Arrestinas/genética , Arrestinas/metabolismo , Fenômenos Biofísicos , Biofísica , Técnicas Biossensoriais , Células , Transferência Ressonante de Energia de Fluorescência , Humanos , Ligantes , Mutagênese Sítio-Dirigida , Fosforilação , Conformação Proteica , Receptor Tipo 1 de Angiotensina/agonistas , Receptor Tipo 1 de Angiotensina/química , Receptor Tipo 1 de Angiotensina/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , beta-Arrestina 2 , beta-Arrestinas
13.
J Clin Invest ; 117(9): 2445-58, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17786238

RESUMO

Deleterious effects on the heart from chronic stimulation of beta-adrenergic receptors (betaARs), members of the 7 transmembrane receptor family, have classically been shown to result from Gs-dependent adenylyl cyclase activation. Here, we identify a new signaling mechanism using both in vitro and in vivo systems whereby beta-arrestins mediate beta1AR signaling to the EGFR. This beta-arrestin-dependent transactivation of the EGFR, which is independent of G protein activation, requires the G protein-coupled receptor kinases 5 and 6. In mice undergoing chronic sympathetic stimulation, this novel signaling pathway is shown to promote activation of cardioprotective pathways that counteract the effects of catecholamine toxicity. These findings suggest that drugs that act as classical antagonists for G protein signaling, but also stimulate signaling via beta-arrestin-mediated cytoprotective pathways, would represent a novel class of agents that could be developed for multiple members of the 7 transmembrane receptor family.


Assuntos
Arrestinas/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Ativação Transcricional/genética , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , beta-Arrestinas
14.
J Pharmacol Exp Ther ; 335(3): 572-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20801892

RESUMO

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.


Assuntos
Angiotensina II/análogos & derivados , Angiotensina II/farmacologia , Arrestinas/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/agonistas , Transdução de Sinais/efeitos dos fármacos , Angiotensina II/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Arrestinas/genética , Ligação Competitiva , Linhagem Celular Tumoral , Interações Medicamentosas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Interferente Pequeno/genética , Ratos , Receptor Tipo 1 de Angiotensina/genética , Transdução de Sinais/fisiologia , Transfecção , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/fisiologia , beta-Arrestinas , Quinases da Família src/metabolismo
15.
J Cell Biol ; 161(5): 899-909, 2003 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-12782683

RESUMO

Signals transduced by kinases depend on the extent and duration of substrate phosphorylation. We generated genetically encoded fluorescent reporters for PKC activity that reversibly respond to stimuli activating PKC. Specifically, phosphorylation of the reporter expressed in mammalian cells causes changes in fluorescence resonance energy transfer (FRET), allowing real time imaging of phosphorylation resulting from PKC activation. Targeting of the reporter to the plasma membrane, where PKC is activated, reveals oscillatory phosphorylation in HeLa cells in response to histamine. Each oscillation in substrate phosphorylation follows a calcium oscillation with a lag of approximately 10 s. Novel FRET-based reporters for PKC translocation, phosphoinositide bisphosphate conversion to IP3, and diacylglycerol show that in HeLa cells the oscillatory phosphorylations correlate with Ca2+-controlled translocation of conventional PKC to the membrane without oscillations of PLC activity or diacylglycerol. However, in MDCK cells stimulated with ATP, PLC and diacylglycerol fluctuate together with Ca2+ and phosphorylation. Thus, specificity of PKC signaling depends on the local second messenger-controlled equilibrium between kinase and phosphatase activities to result in strict calcium-controlled temporal regulation of substrate phosphorylation.


Assuntos
Relógios Biológicos/genética , Membrana Celular/metabolismo , Células Eucarióticas/enzimologia , Genes Reporter/genética , Proteína Quinase C/metabolismo , Transdução de Sinais/genética , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Relógios Biológicos/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Membrana Celular/efeitos dos fármacos , Diglicerídeos/metabolismo , Diglicerídeos/farmacologia , Cães , Células Eucarióticas/efeitos dos fármacos , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes , Células HeLa , Histamina/farmacologia , Humanos , Lipídeos de Membrana/metabolismo , Modelos Biológicos , Fosfatidilinositóis/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína Quinase C/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Sistemas do Segundo Mensageiro/genética , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo
16.
Neuropharmacology ; 148: 77-86, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30553828

RESUMO

Headaches are highly disabling and are among the most common neurological disorders worldwide. Despite the high prevalence of headache, therapeutic options are limited. We recently identified the delta opioid receptor (DOR) as an emerging therapeutic target for migraine. In this study, we examined the effectiveness of a hallmark DOR agonist, SNC80, in disease models reflecting diverse headache disorders including: chronic migraine, post-traumatic headache (PTH), medication overuse headache by triptans (MOH), and opioid-induced hyperalgesia (OIH). To model chronic migraine C57BL/6J mice received chronic intermittent treatment with the known human migraine trigger, nitroglycerin. PTH was modeled by combining the closed head weight drop model with the nitroglycerin model of chronic migraine. For MOH and OIH, mice were chronically treated with sumatriptan or morphine, respectively. The development of periorbital and peripheral allodynia was observed in all four models; and SNC80 significantly inhibited allodynia in all cases. In addition, we also determined if chronic daily treatment with SNC80 would induce MOH/OIH, and we observed limited hyperalgesia relative to sumatriptan or morphine. Together, our results indicate that DOR agonists could be effective in multiple headache disorders, despite their distinct etiology, thus presenting a novel therapeutic target for headache.


Assuntos
Benzamidas/uso terapêutico , Transtornos da Cefaleia/tratamento farmacológico , Piperazinas/uso terapêutico , Receptores Opioides delta/agonistas , Animais , Benzamidas/efeitos adversos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Transtornos da Cefaleia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Morfina , Nitroglicerina , Piperazinas/efeitos adversos , Receptores Opioides delta/uso terapêutico , Sumatriptana
17.
Mol Pharmacol ; 74(3): 585-594, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18524885

RESUMO

Trace amines are neurotransmitters whose role in regulating invertebrate physiology has been appreciated for many decades. Recent studies indicate that trace amines may also play a role in mammalian physiology by binding to a novel family of G protein-coupled receptors (GPCRs) that are found throughout the central nervous system. A major obstacle impeding the careful pharmacological characterization of trace amine associated receptors (TAARs) is their extremely poor membrane expression in model cell systems, and a molecular basis for this phenomenon has not been determined. In the present study, we show that the addition of an asparagine-linked glycosylation site to the N terminus of the human trace amine associated receptor 1 (TAAR1) is sufficient to enable its plasma membrane expression, and thus its pharmacological characterization with a novel cAMP EPAC (exchange protein directly activated by cAMP) protein based bioluminescence resonance energy transfer (BRET) biosensor. We applied this novel cAMP BRET biosensor to evaluate the activity of putative TAAR1 ligands. This study represents the first comprehensive investigation of the membrane-expressed human TAAR1 pharmacology. Our strategy to express TAARs and to identify their ligands using a cAMP BRET assay could provide a foundation for characterizing the functional role of trace amines in vivo and suggests a strategy to apply to groups of poorly expressing GPCRs that have remained difficult to investigate in model systems.


Assuntos
Técnicas Biossensoriais , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Transferência Ressonante de Energia de Fluorescência , Receptores Acoplados a Proteínas G/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dextroanfetamina/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Fenetilaminas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
18.
Trends Pharmacol Sci ; 28(8): 416-22, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17644195

RESUMO

Seven-transmembrane receptors (7TMRs), the most common molecular targets of modern drug therapy, are critically regulated by beta-arrestins, which both inhibit classic G-protein signaling and initiate distinct beta-arrestin signaling. The interplay of G-protein and beta-arrestin signals largely determines the cellular consequences of 7TMR-targeted drugs. Until recently, a drug's efficacy for beta-arrestin recruitment was believed to be proportional to its efficacy for G-protein activities. This paradigm restricts 7TMR drug effects to a linear spectrum of responses, ranging from inhibition of all responses to stimulation of all responses. However, it is now clear that 'biased ligands' can selectively activate G-protein or beta-arrestin functions and thus elicit novel biological effects from even well-studied 7TMRs. Here, we discuss the current state of beta-arrestin-biased ligand research and the prospects for beta-arrestin bias as a therapeutic target. Consideration of ligand bias might have profound influences on the way scientists approach 7TMR-targeted drug discovery.


Assuntos
Arrestinas/farmacologia , Ligantes , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação Alostérica , Animais , Arrestinas/química , Arrestinas/metabolismo , Desenho de Fármacos , Humanos , Modelos Biológicos , Ligação Proteica , Receptores Acoplados a Proteínas G/química , beta-Arrestinas
19.
Curr Opin Pharmacol ; 32: 77-84, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27936408

RESUMO

Opioid chemistry and biology occupy a pivotal place in the history of pharmacology and medicine. Morphine offers unmatched efficacy in alleviating acute pain, but is also associated with a host of adverse side effects. The advent of biased agonism at G protein-coupled receptors has expanded our understanding of intracellular signaling and highlighted the concept that certain ligands are able to differentially modulate downstream pathways. The ability to target one pathway over another has allowed for the development of biased ligands with robust clinical efficacy and fewer adverse events. In this review we summarize these concepts with an emphasis on biased mu opioid receptor pharmacology and highlight how far opioid pharmacology has evolved.


Assuntos
Analgésicos Opioides/farmacologia , Dor/tratamento farmacológico , Receptores Opioides mu/agonistas , Analgésicos Opioides/efeitos adversos , Animais , Desenho de Fármacos , Humanos , Ligantes , Morfina/efeitos adversos , Morfina/farmacologia , Dor/fisiopatologia , Receptores Opioides mu/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Cardiovasc Res ; 107(2): 226-34, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26045475

RESUMO

AIMS: Therapeutic approaches to treat familial dilated cardiomyopathy (DCM), which is characterized by depressed sarcomeric tension and susceptibility to Ca(2+)-related arrhythmias, have been generally unsuccessful. Our objective in the present work was to determine the effect of the angiotensin II type 1 receptor (AT1R) biased ligand, TRV120023, on contractility of hearts of a transgenic mouse model of familial DCM with mutation in tropomyosin at position 54 (TG-E54K). Our rationale is based on previous studies, which have supported the hypothesis that biased G-protein-coupled receptor ligands, signalling via ß-arrestin, increase cardiac contractility with no effect on Ca(2+) transients. Our previous work demonstrated that the biased ligand TRV120023 is able to block angiotensin-induced hypertrophy, while promoting an increase in sarcomere Ca(2+) response. METHODS AND RESULTS: We tested the hypothesis that the depression in cardiac function associated with DCM can be offset by infusion of the AT1R biased ligand, TRV120023. We intravenously infused saline, TRV120023, or the unbiased ligand, losartan, for 15 min in TG-E54K and non-transgenic mice to obtain left ventricular pressure-volume relations. Hearts were analysed for sarcomeric protein phosphorylation. Results showed that the AT1R biased ligand increases cardiac performance in TG-E54K mice in association with increased myosin light chain-2 phosphorylation. CONCLUSION: Treatment of mice with an AT1R biased ligand, acting via ß-arrestin signalling, is able to induce an increase in cardiac contractility associated with an increase in ventricular myosin light chain-2 phosphorylation. AT1R biased ligands may prove to be a novel inotropic approach in familial DCM.


Assuntos
Miosinas Cardíacas/metabolismo , Cardiomiopatia Dilatada/metabolismo , Contração Miocárdica/fisiologia , Cadeias Leves de Miosina/metabolismo , Oligopeptídeos/metabolismo , Animais , Arrestinas/metabolismo , Modelos Animais de Doenças , Feminino , Coração/fisiopatologia , Ligantes , Masculino , Camundongos Transgênicos , Fosforilação , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA