Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
2.
Mol Pharmacol ; 103(2): 89-99, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36351797

RESUMO

Known off-target interactions frequently cause predictable drug side-effects (e.g., ß1-antagonists used for heart disease, risk ß2-mediated bronchospasm). Computer-aided drug design would improve if the structural basis of existing drug selectivity was understood. A mutagenesis approach determined the ligand-amino acid interactions required for ß1-selective affinity of xamoterol and nebivolol, followed by computer-based modeling to provide possible structural explanations. 3H-CGP12177 whole cell binding was conducted in Chinese hamster ovary cells stably expressing human ß1, ß2, and chimeric ß1/ß2-adrenoceptors (ARs). Single point mutations were investigated in transiently transfected cells. Modeling studies involved docking ligands into three-dimensional receptor structures and performing molecular dynamics simulations, comparing interaction frequencies between apo and holo structures of ß1 and ß2-ARs. From these observations, an ICI89406 derivative was investigated that gave further insights into selectivity. Stable cell line studies determined that transmembrane 2 was crucial for the ß1-selective affinity of xamoterol and nebivolol. Single point mutations determined that the ß1-AR isoleucine (I118) rather than the ß2 histidine (H93) explained selectivity. Studies of other ß1-ligands found I118 was important for ICI89406 selective affinity but not that for betaxolol, bisoprolol, or esmolol. Modeling studies suggested that the interaction energies and solvation of ß1-I118 and ß2-H93 are factors determining selectivity of xamoterol and ICI89406. ICI89406 without its phenyl group loses its high ß1-AR affinity, resulting in the same affinity as for the ß2-AR. The human ß1-AR residue I118 is crucial for the ß1-selective affinity of xamoterol, nebivolol, and ICI89406 but not all ß1-selective compounds. SIGNIFICANCE STATEMENT: Some ligands have selective binding affinity for the human ß1 versus the ß2-adrenoceptor; however, the molecular/structural reason for this is not known. The transmembrane 2 residue isoleucine I118 is responsible for the selective ß1-binding of xamoterol, nebivolol, and ICI89406 but does not explain the selective ß1-binding of betaxolol, bisoprolol, or esmolol. Understanding the structural basis of selectivity is important to improve computer-aided ligand design, and targeting I118 in ß1-adrenoceptors is likely to increase ß1-selectivity of drugs.


Assuntos
Antagonistas Adrenérgicos beta , Bisoprolol , Animais , Cricetinae , Humanos , Xamoterol , Nebivolol/farmacologia , Antagonistas Adrenérgicos beta/metabolismo , Isoleucina , Agonistas Adrenérgicos beta , Betaxolol , Células CHO , Ligantes , Cricetulus , Receptores Adrenérgicos , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 1/química
3.
J Mol Biol ; 434(16): 167687, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35717996

RESUMO

Cell-free expression enables direct cotranslational insertion of G protein coupled receptors (GPCRs) and other membrane proteins into the defined membrane environments of nanodiscs. This technique avoids GPCR contacts with detergents and allows rapid identification of lipid effects on GPCR function as well as fast screening of receptor derivatives. Critical steps of conventional GPCR preparation from cellular membranes followed by detergent-based reconstitution into nanodisc membranes are thus eliminated. We report the efficient cotranslational insertion of full-length human ß1-adrenergic receptor and of a truncated derivative into preformed nanodisc membranes. Their biochemical characterization revealed significant differences in lipid requirements, dimer formation and ligand binding activity. The truncated receptor showed a higher affinity to most tested ligands, in particular in presence of choline-containing lipids. However, introducing the naturally occurring G389R polymorphism in the full-length receptor resulted into an increased affinity to the antagonists alprenolol and carvedilol. Receptor quality was generally improved by coexpression with the agonist isoproterenol and the percentage of the ligand binding active fraction was twofold increased. Specific coupling of full-length and truncated human receptors in nanodisc membranes to Mini-Gαs protein as well as to purified Gs heterotrimer could be demonstrated and homogeneity of purified GPCR/Gs protein complexes in nanodiscs was demonstrated by negative stain single particle analysis.


Assuntos
Nanoestruturas , Receptores Adrenérgicos beta 1 , Sistema Livre de Células , Humanos , Ligantes , Lipídeos/química , Nanoestruturas/química , Polimorfismo Genético , Ligação Proteica , Biossíntese de Proteínas , Multimerização Proteica , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
4.
Int J Mol Sci ; 22(20)2021 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-34681845

RESUMO

Cardiovascular diseases (CDs) are a major concern in the human race and one of the leading causes of death worldwide. ß-Adrenergic receptors (ß1-AR and ß2-AR) play a crucial role in the overall regulation of cardiac function. In the present study, structure-based virtual screening, machine learning (ML), and a ligand-based similarity search were conducted for the PubChem database against both ß1- and ß2-AR. Initially, all docked molecules were screened using the threshold binding energy value. Molecules with a better binding affinity were further used for segregation as active and inactive through ML. The pharmacokinetic assessment was carried out on molecules retained in the above step. Further, similarity searching of the ChEMBL and DrugBank databases was performed. From detailed analysis of the above data, four compounds for each of ß1- and ß2-AR were found to be promising in nature. A number of critical ligand-binding amino acids formed potential hydrogen bonds and hydrophobic interactions. Finally, a molecular dynamics (MD) simulation study of each molecule bound with the respective target was performed. A number of parameters obtained from the MD simulation trajectories were calculated and substantiated the stability between the protein-ligand complex. Hence, it can be postulated that the final molecules might be crucial for CDs subjected to experimental validation.


Assuntos
Descoberta de Drogas , Simulação de Dinâmica Molecular , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 2/química , Humanos , Ligantes , Aprendizado de Máquina , Ligação Proteica
5.
J Mol Cell Cardiol ; 154: 70-79, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33556394

RESUMO

ß1-adrenergic receptors (ß1ARs) are the principle mediators of catecholamine action in cardiomyocytes. We previously showed that the ß1AR extracellular N-terminus is a target for post-translational modifications that impact on signaling responses. Specifically, we showed that the ß1AR N-terminus carries O-glycan modifications at Ser37/Ser41, that O-glycosylation prevents ß1AR N-terminal cleavage, and that N-terminal truncation influences ß1AR signaling to downstream effectors. However, the site(s) and mechanism for ß1AR N-terminal cleavage in cells was not identified. This study shows that ß1ARs are expressed in cardiomyocytes and other cells types as both full-length and N-terminally truncated species and that the truncated ß1AR species is formed as a result of an O-glycan regulated N-terminal cleavage by ADAM17 at R31↓L32. We identify Ser41 as the major O-glycosylation site on the ß1AR N-terminus and show that an O-glycan modification at Ser41 prevents ADAM17-dependent cleavage of the ß1-AR N-terminus at S41↓L42, a second N-terminal cleavage site adjacent to this O-glycan modification (and it attenuates ß1-AR N-terminal cleavage at R31↓L32). We previously reported that oxidative stress leads to a decrease in ß1AR expression and catecholamine responsiveness in cardiomyocytes. This study shows that redox-inactivation of cardiomyocyte ß1ARs is via a mechanism involving N-terminal truncation at R31↓L32 by ADAM17. In keeping with the previous observation that N-terminally truncated ß1ARs constitutively activate an AKT pathway that affords protection against doxorubicin-dependent apoptosis, overexpression of a cleavage resistant ß1AR mutant exacerbates doxorubicin-dependent apoptosis. These studies identify the ß1AR N-terminus as a structural determinant of ß1AR responses that can be targeted for therapeutic advantage.


Assuntos
Proteína ADAM17/metabolismo , Miócitos Cardíacos/metabolismo , Oxirredução , Receptores Adrenérgicos beta 1/metabolismo , Expressão Gênica , Glicosilação , Humanos , Estresse Oxidativo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Proteólise , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
6.
J Mol Cell Cardiol ; 153: 14-25, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33326834

RESUMO

ß-adrenergic receptor antagonists (ß-blockers) are extensively used to improve cardiac performance in heart failure (HF), but the electrical improvements with these clinical treatments are not fully understood. The aim of this study was to analyze the electrophysiological effects of ß-adrenergic system remodeling in heart failure with reduced ejection fraction and the underlying mechanisms. We used a combined mathematical model that integrated ß-adrenergic signaling with electrophysiology and calcium cycling in human ventricular myocytes. HF remodeling, both in the electrophysiological and signaling systems, was introduced to quantitatively analyze changes in electrophysiological properties due to the stimulation of ß-adrenergic receptors in failing myocytes. We found that the inotropic effect of ß-adrenergic stimulation was reduced in HF due to the altered Ca2+ dynamics resulting from the combination of structural, electrophysiological and signaling remodeling. Isolated cells showed proarrhythmic risk after sympathetic stimulation because early afterdepolarizations appeared, and the vulnerability was greater in failing myocytes. When analyzing coupled cells, ß-adrenergic stimulation reduced transmural repolarization gradients between endocardium and epicardium in normal tissue, but was less effective at reducing these gradients after HF remodeling. The comparison of the selective activation of ß-adrenergic isoforms revealed that the response to ß2-adrenergic receptors stimulation was blunted in HF while ß1-adrenergic receptors downstream effectors regulated most of the changes observed after sympathetic stimulation. In conclusion, this study was able to reproduce an altered ß-adrenergic activity on failing myocytes and to explain the mechanisms involved. The derived predictions could help in the treatment of HF and guide in the design of future experiments.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Simulação por Computador , Insuficiência Cardíaca/fisiopatologia , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Remodelação Ventricular , Potenciais de Ação , Cálcio/metabolismo , Humanos , Modelos Cardiovasculares , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 2/química
7.
Cell Signal ; 78: 109846, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33238186

RESUMO

ß1-adrenergic receptor (ß1AR)-mediated transactivation of epidermal growth factor receptor (EGFR) engages downstream signaling events that impact numerous cellular processes including growth and survival. While association of these receptors has been shown to occur basally and be important for relaying transactivation-specific intracellular events, the mechanism by which they do so is unclear and elucidation of which would aid in understanding the consequence of disrupting their interaction. Using fluorescence resonance energy transfer (FRET) and immunoprecipitation (IP) analyses, we evaluated the impact of C-terminal truncations of EGFR on its ability to associate with ß1AR. While loss of the last 230 amino acid C-terminal phosphotyrosine-rich domain did not disrupt the ability of EGFR to associate with ß1AR, truncation of the entire intracellular domain of EGFR resulted in almost complete loss of its interaction with ß1AR, suggesting that either the kinase domain or juxtamembrane domain (JMD) may be required for this association. Treatment with the EGFR antagonist gefitinib did not prevent ß1AR-EGFR association, however, treatment with a palmitoylated peptide encoding the first 20 amino acids of the JMD domain (JMD-A) disrupted ß1AR-EGFR association over time and prevented ß1AR-mediated ERK1/2 phosphorylation, both in general and specifically in association with EGFR. Conversely, neither a mutated JMD-A peptide nor a palmitoylated peptide fragment consisting of the subsequent 18 amino acids of the JMD domain (JMD-B) were capable of doing so. Altogether, the proximal region of the JMD of EGFR is responsible for its association with ß1AR, and its disruption prevents ß1AR-mediated transactivation, thus providing a new tool to study the functional consequences of disrupting ß1AR-EGFR downstream signaling.


Assuntos
Receptores Adrenérgicos beta 1/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Receptores ErbB/química , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Peptídeos/química , Peptídeos/genética , Peptídeos/farmacologia , Domínios Proteicos , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
8.
Mol Cell ; 80(1): 59-71.e4, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32818430

RESUMO

Cardiac disease remains the leading cause of morbidity and mortality worldwide. The ß1-adrenergic receptor (ß1-AR) is a major regulator of cardiac functions and is downregulated in the majority of heart failure cases. A key physiological process is the activation of heterotrimeric G-protein Gs by ß1-ARs, leading to increased heart rate and contractility. Here, we use cryo-electron microscopy and functional studies to investigate the molecular mechanism by which ß1-AR activates Gs. We find that the tilting of α5-helix breaks a hydrogen bond between the sidechain of His373 in the C-terminal α5-helix and the backbone carbonyl of Arg38 in the N-terminal αN-helix of Gαs. Together with the disruption of another interacting network involving Gln59 in the α1-helix, Ala352 in the ß6-α5 loop, and Thr355 in the α5-helix, these conformational changes might lead to the deformation of the GDP-binding pocket. Our data provide molecular insights into the activation of G-proteins by G-protein-coupled receptors.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Isoproterenol/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/metabolismo , Animais , Sítios de Ligação , Bovinos , Linhagem Celular , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína
9.
Nature ; 583(7818): 862-866, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32555462

RESUMO

The ß1-adrenoceptor (ß1AR) is a G-protein-coupled receptor (GPCR) that couples1 to the heterotrimeric G protein Gs. G-protein-mediated signalling is terminated by phosphorylation of the C terminus of the receptor by GPCR kinases (GRKs) and by coupling of ß-arrestin 1 (ßarr1, also known as arrestin 2), which displaces Gs and induces signalling through the MAP kinase pathway2. The ability of synthetic agonists to induce signalling preferentially through either G proteins or arrestins-known as biased agonism3-is important in drug development, because the therapeutic effect may arise from only one signalling cascade, whereas the other pathway may mediate undesirable side effects4. To understand the molecular basis for arrestin coupling, here we determined the cryo-electron microscopy structure of the ß1AR-ßarr1 complex in lipid nanodiscs bound to the biased agonist formoterol5, and the crystal structure of formoterol-bound ß1AR coupled to the G-protein-mimetic nanobody6 Nb80. ßarr1 couples to ß1AR in a manner distinct to that7 of Gs coupling to ß2AR-the finger loop of ßarr1 occupies a narrower cleft on the intracellular surface, and is closer to transmembrane helix H7 of the receptor when compared with the C-terminal α5 helix of Gs. The conformation of the finger loop in ßarr1 is different from that adopted by the finger loop of visual arrestin when it couples to rhodopsin8. ß1AR coupled to ßarr1 shows considerable differences in structure compared with ß1AR coupled to Nb80, including an inward movement of extracellular loop 3 and the cytoplasmic ends of H5 and H6. We observe weakened interactions between formoterol and two serine residues in H5 at the orthosteric binding site of ß1AR, and find that formoterol has a lower affinity for the ß1AR-ßarr1 complex than for the ß1AR-Gs complex. The structural differences between these complexes of ß1AR provide a foundation for the design of small molecules that could bias signalling in the ß-adrenoceptors.


Assuntos
Microscopia Crioeletrônica , Fumarato de Formoterol/química , Fumarato de Formoterol/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/ultraestrutura , beta-Arrestina 1/química , beta-Arrestina 1/ultraestrutura , Sequência de Aminoácidos , Animais , Sítios de Ligação , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/ultraestrutura , Células HEK293 , Humanos , Modelos Moleculares , Complexos Multiproteicos , Receptores Adrenérgicos beta 1/metabolismo , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo , Anticorpos de Cadeia Única/ultraestrutura , Peixe-Zebra , beta-Arrestina 1/metabolismo
10.
Nat Commun ; 11(1): 2216, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32371991

RESUMO

Signal transmission and regulation of G-protein-coupled receptors (GPCRs) by extra- and intracellular ligands occurs via modulation of complex conformational equilibria, but their exact kinetic details and underlying atomic mechanisms are unknown. Here we quantified these dynamic equilibria in the ß1-adrenergic receptor in its apo form and seven ligand complexes using 1H/15N NMR spectroscopy. We observe three major exchanging conformations: an inactive conformation (Ci), a preactive conformation (Cp) and an active conformation (Ca), which becomes fully populated in a ternary complex with a G protein mimicking nanobody. The Ci ↔ Cp exchange occurs on the microsecond scale, the Cp ↔ Ca exchange is slower than ~5 ms and only occurs in the presence of two highly conserved tyrosines (Y5.58, Y7.53), which stabilize the active conformation of TM6. The Cp→Ca chemical shift changes indicate a pivoting motion of the entire TM6 that couples the effector site to the orthosteric ligand pocket.


Assuntos
Regulação Alostérica , Espectroscopia de Ressonância Magnética/métodos , Conformação Proteica , Receptores Adrenérgicos beta 1/química , Receptores Acoplados a Proteínas G/química , Algoritmos , Animais , Humanos , Ligantes , Modelos Moleculares , Modelos Teóricos , Ligação Proteica , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Sf9 , Spodoptera
11.
Trends Pharmacol Sci ; 41(6): 387-389, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32362341

RESUMO

The ability to design 'biased' drugs that selectively activate G protein-coupled receptor (GPCR) signaling pathways beneficial in treating a disease, while limiting their side effects, is of broad significance. Lee et al. move us a step closer to this important goal by identifying structural differences in the ß1-adrenoceptor in complex with ß-arrestin 1 versus a G protein-mimicking nanobody.


Assuntos
Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , beta-Arrestina 1/química , Animais , Materiais Biomiméticos/química , Humanos , Modelos Moleculares , Terapia de Alvo Molecular , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anticorpos de Domínio Único/química , beta-Arrestina 1/metabolismo
12.
Nat Commun ; 11(1): 669, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32015348

RESUMO

G-protein-coupled receptors (GPCRs) are allosteric signaling proteins that transmit an extracellular stimulus across the cell membrane. Using 19F NMR and site-specific labelling, we investigate the response of the cytoplasmic region of transmembrane helices 6 and 7 of the ß1-adrenergic receptor to agonist stimulation and coupling to a Gs-protein-mimetic nanobody. Agonist binding shows the receptor in equilibrium between two inactive states and a pre-active form, increasingly populated with higher ligand efficacy. Nanobody coupling leads to a fully active ternary receptor complex present in amounts correlating directly with agonist efficacy, consistent with partial agonism. While for different agonists the helix 6 environment in the active-state ternary complexes resides in a well-defined conformation, showing little conformational mobility, the environment of the highly conserved NPxxY motif on helix 7 remains dynamic adopting diverse, agonist-specific conformations, implying a further role of this region in receptor function. An inactive nanobody-coupled ternary receptor form is also observed.


Assuntos
Imagem por Ressonância Magnética de Flúor-19 , Receptores Adrenérgicos beta 1/química , Receptores Acoplados a Proteínas G/química , Sequência de Aminoácidos , Membrana Celular/metabolismo , Humanos , Ligantes , Proteínas de Membrana/química , Modelos Moleculares , Conformação Proteica , Receptores Adrenérgicos beta 1/isolamento & purificação , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
13.
J Am Chem Soc ; 141(42): 16663-16670, 2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31564099

RESUMO

G protein-coupled receptors (GPCRs) are versatile chemical sensors, which transmit the signal of an extracellular binding event across the plasma membrane to the intracellular side. This function is achieved via the modulation of highly dynamical equilibria of various conformational receptor states. Here we have probed the effect of pressure on the conformational equilibria of a functional thermostabilized ß1-adrenergic GPCR (ß1AR) by solution NMR. High pressure induces a large shift in the conformational equilibrium (midpoint ∼600 bar) from the preactive conformation of agonist-bound ß1AR to the fully active conformation, which under normal pressure is only populated when a G protein or a G protein-mimicking nanobody (Nb) binds to the intracellular side of the ß1AR·agonist complex. No such large effects are observed for an antagonist-bound ß1AR or the ternary ß1AR·agonist·Nb80 complex. The detected structural changes of agonist-bound ß1AR around the orthosteric ligand binding pocket indicate that the fully active receptor occupies an ∼100 Å3 smaller volume than that of its preactive form. Most likely, this volume reduction is caused by the compression of empty (nonhydrated) cavities in the ligand binding pocket and the center of the receptor, which increases the ligand receptor interactions and explains the ∼100-fold affinity increase of agonists in the presence of G protein. The finding that isotropic pressure induces a directed motion from the preactive to the fully active GPCR conformation provides evidence of the high mechanical robustness of this important functional switch.


Assuntos
Modelos Moleculares , Pressão , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/metabolismo , Regulação Alostérica , Membrana Celular/metabolismo , Conformação Proteica
14.
Science ; 364(6442): 775-778, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-31072904

RESUMO

G protein-coupled receptors (GPCRs) in the G protein-coupled active state have higher affinity for agonists as compared with when they are in the inactive state, but the molecular basis for this is unclear. We have determined four active-state structures of the ß1-adrenoceptor (ß1AR) bound to conformation-specific nanobodies in the presence of agonists of varying efficacy. Comparison with inactive-state structures of ß1AR bound to the identical ligands showed a 24 to 42% reduction in the volume of the orthosteric binding site. Potential hydrogen bonds were also shorter, and there was up to a 30% increase in the number of atomic contacts between the receptor and ligand. This explains the increase in agonist affinity of GPCRs in the active state for a wide range of structurally distinct agonists.


Assuntos
Agonistas de Receptores Adrenérgicos beta 1/química , Desenho de Fármacos , Receptores Acoplados a Proteínas G/agonistas , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Sítio Alostérico/imunologia , Domínio Catalítico/imunologia , Ligação de Hidrogênio , Ligantes , Ligação Proteica , Estrutura Secundária de Proteína , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/imunologia , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/imunologia , Anticorpos de Domínio Único/imunologia
15.
Circ Res ; 124(9): 1350-1359, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30836825

RESUMO

RATIONALE: ßARs (ß-adrenergic receptors) are prototypical GPCRs (G protein-coupled receptors) that play a pivotal role in sympathetic regulation. In heart cells, ß1AR signaling mediates a global response, including both l-type Ca2+ channels in the sarcolemma/T tubules and RyRs (ryanodine receptors) in the SR (sarcoplasmic reticulum). In contrast, ß2AR mediates local signaling with little effect on the function of SR proteins. OBJECTIVE: To investigate the signaling relationship between ß1ARs and ß2ARs. METHOD AND RESULTS: Using whole-cell patch-clamp analyses combined with confocal Ca2+ imaging, we found that the activation of compartmentalized ß2AR signaling was able to convert the ß1AR signaling from global to local mode, preventing ß1ARs from phosphorylating RyRs that were only nanometers away from sarcolemma/T tubules. This offside compartmentalization was eliminated by selective inhibition of ß2AR, GRK2 (GPCR kinase-2), ßarr1 (ß-arrestin-1), and phosphodiesterase-4. A knockin rat model harboring mutations of the last 3 serine residues of the ß1AR C terminus, a component of the putative ßarr1 binding site and GRK2 phosphorylation site, eliminated the offside compartmentalization conferred by ß2AR activation. CONCLUSIONS: ß2AR stimulation compartmentalizes ß1AR signaling into nanoscale local domains in a phosphodiesterase-4-dependent manner by targeting the C terminus of ß1ARs. This finding reveals a fundamental negative feed-forward mechanism that serves to avoid the cytotoxicity of circulating catecholamine and to sharpen the transient ß1AR response of sympathetic excitation.


Assuntos
Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Adrenérgicos/farmacologia , Animais , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Masculino , Mutação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Transgênicos , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
PLoS One ; 13(8): e0201160, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30125285

RESUMO

RATIONALE: Despite advances in pharmacotherapy, heart failure still incurs significant morbidity and mortality. Stimulating antibodies directed against the secondextracellular loop of the human ß1-adrenergic receptor (anti-ß1EC2) cause myocyte damage and heart failure in rats. This receptor domain is 100% homologous between rats and humans. OBJECTIVE: ß1EC2-mimicking cyclopeptides (25-meric) markedly improved the development and/or course of anti-ß1EC2-mediated cardiomyopathy. Further developments should be investigated. METHODS AND RESULTS: The shortened 18-meric cyclic peptide COR-1, in which one of the two disulphide bonds was removed to enable reproducible GMP production, can also be used to treat cardiomyopathic rats. Echocardiography, catheterization and histopathology of the rat hearts revealed that monthly intravenous administrations of COR-1 almost fully reversed the cardiomyopathic phenotype within 6 months at doses of 1 to 4 mg/kg body weight. Administration of COR-1 resulted in markedly reduced anti-ß1EC2-expressing memory B lymphocytes in the spleen despite continued antigenic boosts, but did not significantly decrease overall peripheral anti-ß1EC2 titers. COR-1 did not induce any anti-ß1EC2 or other immune response in naïve rats (corresponding to findings in healthy human volunteers). It did not cause any toxic side effects in GLP studies in dogs, rats or mice, and the "no observed adverse effect level" (NOAEL) exceeded the therapeutic doses by 100-fold. CONCLUSION: The second generation immunomodulating epitope-mimicking cyclopeptide COR-1 (also termed JNJ-5442840) offers promise to treat immune-mediated cardiac diseases.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Receptores Adrenérgicos beta 1/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/administração & dosagem , Especificidade de Anticorpos , Modelos Animais de Doenças , Feminino , Cobaias , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Mimetismo Molecular/imunologia , Miocárdio/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Peptídeos Cíclicos/química , Peptídeos Cíclicos/toxicidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
17.
Nature ; 559(7714): 423-427, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29995853

RESUMO

G-protein-coupled receptors (GPCRs) are involved in many physiological processes and are therefore key drug targets1. Although detailed structural information is available for GPCRs, the effects of lipids on the receptors, and on downstream coupling of GPCRs to G proteins are largely unknown. Here we use native mass spectrometry to identify endogenous lipids bound to three class A GPCRs. We observed preferential binding of phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2) over related lipids and confirm that the intracellular surface of the receptors contain hotspots for PtdIns(4,5)P2 binding. Endogenous lipids were also observed bound directly to the trimeric Gαsßγ protein complex of the adenosine A2A receptor (A2AR) in the gas phase. Using engineered Gα subunits (mini-Gαs, mini-Gαi and mini-Gα12)2, we demonstrate that the complex of mini-Gαs with the ß1 adrenergic receptor (ß1AR) is stabilized by the binding of two PtdIns(4,5)P2 molecules. By contrast, PtdIns(4,5)P2 does not stabilize coupling between ß1AR and other Gα subunits (mini-Gαi or mini-Gα12) or a high-affinity nanobody. Other endogenous lipids that bind to these receptors have no effect on coupling, highlighting the specificity of PtdIns(4,5)P2. Calculations of potential of mean force and increased GTP turnover by the activated neurotensin receptor when coupled to trimeric Gαißγ complex in the presence of PtdIns(4,5)P2 provide further evidence for a specific effect of PtdIns(4,5)P2 on coupling. We identify key residues on cognate Gα subunits through which PtdIns(4,5)P2 forms bridging interactions with basic residues on class A GPCRs. These modulating effects of lipids on receptors suggest consequences for understanding function, G-protein selectivity and drug targeting of class A GPCRs.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Simulação de Dinâmica Molecular , Estabilidade Proteica , Ratos , Receptores Adrenérgicos alfa 2/química , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Neurotensina/química , Receptores de Neurotensina/genética , Receptores de Neurotensina/metabolismo , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo , Especificidade por Substrato , Perus
18.
Mol Pharmacol ; 94(2): 862-875, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29848777

RESUMO

The ß1-adrenergic receptor (ß1-AR) is a major cardiac G protein-coupled receptor, which mediates cardiac actions of catecholamines and is involved in genesis and treatment of numerous cardiovascular disorders. In mammalian cells, catecholamines induce the internalization of the ß1-AR into endosomes and their removal promotes the recycling of the endosomal ß1-AR back to the plasma membrane; however, whether these redistributive processes occur in terminally differentiated cells is unknown. Compartmentalization of the ß1-AR in response to ß-agonists and antagonists was determined by confocal microscopy in primary adult rat ventricular myocytes (ARVMs), which are terminally differentiated myocytes with unique structures such as transverse tubules (T-tubules) and contractile sarcomeres. In unstimulated ARVMs, the fluorescently labeled ß1-AR was expressed on the external membrane (the sarcolemma) of cardiomyocytes. Exposing ARVMs to isoproterenol redistributed surface ß1-ARs into small (∼225-250 nm) regularly spaced internal punctate structures that overlapped with puncta stained by Di-8 ANEPPS, a membrane-impermeant T-tubule-specific dye. Replacing the ß-agonist with the ß-blocker alprenolol, induced the translocation of the wild-type ß1-AR from these punctate structures back to the plasma membrane. This step was dependent on two barcodes, namely, the type-1 PDZ binding motif and serine at position 312 of the ß1-AR, which is phosphorylated by a pool of cAMP-dependent protein kinases anchored at the type-1 PDZ of the ß1-AR. These data show that redistribution of the ß1-AR in ARVMs from internal structures back to the plasma membrane was mediated by a novel sorting mechanism, which might explain unique aspects of cardiac ß1-AR signaling under normal or pathologic conditions.


Assuntos
Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Miócitos Cardíacos/citologia , Receptores Adrenérgicos beta 1/metabolismo , Alprenolol/farmacologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Isoproterenol/farmacologia , Miócitos Cardíacos/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
19.
J Smooth Muscle Res ; 54(0): 1-12, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29540622

RESUMO

Object We identified the ß-adrenoceptor (ß-AR) subtypes responsible for the relaxant responses to adrenaline (AD) and noradrenaline (NA) in the rat thoracic aorta and examined the role of cAMP which is involved in these relaxant responses. Methods The effects of ß-AR antagonists or the adenylyl cyclase inhibitor SQ 22,536 on AD- and NA-induced relaxant responses in phenylephrine-induced contraction and increases in cAMP levels were examined in isolated, endothelium-denuded rat thoracic aorta segments. Results AD-induced relaxation was completely suppressed by propranolol (10-7 M) or by ICI-118,551 (10-8 M) plus atenolol (10-6 M), and was also very strongly inhibited by ICI-118,551 (10-8 M) alone. AD (10-5 M) increased tissue cAMP levels by approximately 1.9-fold compared with that in non-stimulated aortic tissue, but did not significantly increase cAMP levels in the presence of ICI-118,551 (10-8 M) or SQ 22,536 (10-4 M). AD-induced relaxation was strongly suppressed by SQ 22,536 (10-4 M). NA-induced relaxation was almost completely suppressed by atenolol (10-6 M) plus ICI-118,551 (10-8 M) although it was hardly affected by ICI-118,551 (10-8 M) alone. NA (10-5 M) increased tissue cAMP levels by approximately 2.2-fold compared with that in non-stimulated aortic tissue, but did not significantly increase cAMP levels in the presence of atenolol (10-6 M) or SQ 22,536 (10-4 M). NA-induced relaxation was strongly suppressed by SQ 22,536 (10-4 M). Conclusion In rat thoracic aorta, AD- and NA-induced relaxations, which are both strongly dependent on increased tissue cAMP levels, are mainly mediated through ß2- and ß1-adrenoceptors respectively.


Assuntos
Aorta Torácica/fisiologia , AMP Cíclico/metabolismo , Epinefrina/farmacologia , Relaxamento Muscular/fisiologia , Norepinefrina/farmacologia , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Masculino , Relaxamento Muscular/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 2/química , Transdução de Sinais
20.
J Smooth Muscle Res ; 54(0): 13-27, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29540623

RESUMO

Object We aimed to identify the ß-adrenoceptor (ß-AR) subtypes involved in isoprenaline-induced relaxation of guinea pig colonic longitudinal smooth muscle using pharmacological and biochemical approaches. Methods Longitudinal smooth muscle was prepared from the male guinea pig ascending colon and contracted with histamine prior to comparing the relaxant responses to three catecholamines (isoprenaline, adrenaline, and noradrenaline). The inhibitory effects of subtype-selective ß-AR antagonists on isoprenaline-induced relaxation were then investigated. Results The relaxant potencies of the catecholamines were ranked as: isoprenaline > noradrenaline ≈ adrenaline, whereas the rank order was isoprenaline > noradrenaline > adrenaline in the presence of propranolol (a non-selective ß-AR antagonist; 3 × 10-7 M). Atenolol (a selective ß1-AR antagonist; 3 × 10-7-10-6 M) acted as a competitive antagonist of isoprenaline-induced relaxation, and the pA2 value was calculated to be 6.49 (95% confidence interval: 6.34-6.83). The relaxation to isoprenaline was not affected by ICI-118,551 (a selective ß2-AR antagonist) at 10-9-10-8 M, but was competitively antagonized by 10-7-3 × 10-7 M, with a pA2 value of 7.41 (95% confidence interval: 7.18-8.02). In the presence of propranolol (3 × 10-7 M), the relaxant effect of isoprenaline was competitively antagonized by bupranolol (a non-selective ß-AR antagonist), with a pA2 value of 5.90 (95% confidence interval: 5.73-6.35). Conclusion These findings indicated that the ß-AR subtypes involved in isoprenaline-induced relaxation of colonic longitudinal guinea pig muscles are ß1-AR and ß3-AR.


Assuntos
Colo/fisiologia , Isoproterenol/farmacologia , Relaxamento Muscular/fisiologia , Músculo Liso/fisiologia , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Colo/efeitos dos fármacos , Colo/metabolismo , Cobaias , Masculino , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 2/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...