Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Pharmacol ; 93(6): 581-591, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29572336

RESUMO

GPR40 is a clinically validated molecular target for the treatment of diabetes. Many GPR40 agonists have been identified to date, with the partial agonist fasiglifam (TAK-875) reaching phase III clinical trials before its development was terminated due to off-target liver toxicity. Since then, attention has shifted toward the development of full agonists that exhibit superior efficacy in preclinical models. Full agonists bind to a distinct binding site, suggesting conformational plasticity and a potential for biased agonism. Indeed, it has been suggested that alternative pharmacology may be required for meaningful efficacy. In this study, we described the discovery and characterization of Compound A, a newly identified GPR40 allosteric full agonist highly efficacious in human islets at potentiating glucose-stimulated insulin secretion. We compared Compound A-induced GPR40 activity to that induced by both fasiglifam and AM-1638, another allosteric full agonist previously reported to be highly efficacious in preclinical models, at a panel of G proteins. Compound A was a full agonist at both the Gαq and Gαi2 pathways, and in contrast to fasiglifam Compound A also induced Gα12 coupling. Compound A and AM-1638 displayed similar activity at all pathways tested. The Gα12/Gα13-mediated signaling pathway has been linked to protein kinase D activation as well as actin remodeling, well known to contribute to the release of insulin vesicles. Our data suggest that the pharmacology of GPR40 is complex and that Gα12/Gα13-mediated signaling, which may contribute to GPR40 agonists therapeutic efficacy, is a specific property of GPR40 allosteric full agonists.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Glucose/metabolismo , Secreção de Insulina/fisiologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Benzofuranos/farmacologia , Células CHO , Linhagem Celular , Cricetulus , Células HEK293 , Humanos , Hipoglicemiantes/farmacologia , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sulfonas/farmacologia
2.
J Biol Chem ; 290(34): 21131-21140, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26157145

RESUMO

FFAR1/GPR40 is a seven-transmembrane domain receptor (7TMR) expressed in pancreatic ß cells and activated by FFAs. Pharmacological activation of GPR40 is a strategy under consideration to increase insulin secretion in type 2 diabetes. GPR40 is known to signal predominantly via the heterotrimeric G proteins Gq/11. However, 7TMRs can also activate functionally distinct G protein-independent signaling via ß-arrestins. Further, G protein- and ß-arrestin-based signaling can be differentially modulated by different ligands, thus eliciting ligand-specific responses ("biased agonism"). Whether GPR40 engages ß-arrestin-dependent mechanisms and is subject to biased agonism is unknown. Using bioluminescence resonance energy transfer-based biosensors for real-time monitoring of cell signaling in living cells, we detected a ligand-induced GPR40-ß-arrestin interaction, with the synthetic GPR40 agonist TAK-875 being more effective than palmitate or oleate in recruiting ß-arrestins 1 and 2. Conversely, TAK-875 acted as a partial agonist of Gq/11-dependent GPR40 signaling relative to both FFAs. Pharmacological blockade of Gq activity decreased FFA-induced insulin secretion. In contrast, knockdown or genetic ablation of ß-arrestin 2 in an insulin-secreting cell line and mouse pancreatic islets, respectively, uniquely attenuated the insulinotropic activity of TAK-875, thus providing functional validation of the biosensor data. Collectively, these data reveal that in addition to coupling to Gq/11, GPR40 is functionally linked to a ß-arrestin 2-mediated insulinotropic signaling axis. These observations expose previously unrecognized complexity for GPR40 signal transduction and may guide the development of biased agonists showing improved clinical profile in type 2 diabetes.


Assuntos
Arrestinas/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Animais , Arrestinas/antagonistas & inibidores , Arrestinas/metabolismo , Benzofuranos/farmacologia , Técnicas Biossensoriais , Linhagem Celular Tumoral , Espectroscopia de Ressonância de Spin Eletrônica , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Insulina/agonistas , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Cinética , Camundongos , Ácido Oleico/farmacologia , Ácido Palmítico/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Sulfonas/farmacologia , Técnicas de Cultura de Tecidos , beta-Arrestina 2 , beta-Arrestinas
3.
Proc Natl Acad Sci U S A ; 109(7): 2376-81, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308370

RESUMO

The G protein-coupled free fatty acid receptor-1 (FFA1/GPR40) plays a major role in the regulation of insulin secretion by fatty acids. GPR40 is considered a potential therapeutic target to enhance insulin secretion in type 2 diabetes; however, its mode of regulation is essentially unknown. The aims of this study were to test the hypothesis that glucose regulates GPR40 gene expression in pancreatic ß-cells and to determine the mechanisms of this regulation. We observed that glucose stimulates GPR40 gene transcription in pancreatic ß-cells via increased binding of pancreas-duodenum homeobox-1 (Pdx-1) to the A-box in the HR2 region of the GPR40 promoter. Mutation of the Pdx-1 binding site within the HR2 abolishes glucose activation of GPR40 promoter activity. The stimulation of GPR40 expression and Pdx-1 binding to the HR2 in response to glucose are mimicked by N-acetyl glucosamine, an intermediate of the hexosamine biosynthesis pathway, and involve PI3K-dependent O-GlcNAcylation of Pdx-1 in the nucleus. We demonstrate that O-GlcNAc transferase (OGT) interacts with the product of the PI3K reaction, phosphatidylinositol 3,4,5-trisphosphate (PIP(3)), in the nucleus. This interaction enables OGT to catalyze O-GlcNAcylation of nuclear proteins, including Pdx-1. We conclude that glucose stimulates GPR40 gene expression at the transcriptional level through Pdx-1 binding to the HR2 region and via a signaling cascade that involves an interaction between OGT and PIP(3) at the nuclear membrane. These observations reveal a unique mechanism by which glucose metabolism regulates the function of transcription factors in the nucleus to induce gene expression.


Assuntos
Acetilglucosamina/metabolismo , Duodeno/metabolismo , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Pâncreas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Acoplados a Proteínas G/genética , Transcrição Gênica , Animais , Hexosaminas/biossíntese , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , Camundongos Endogâmicos C57BL
4.
Commun Biol ; 7(1): 250, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429428

RESUMO

Mutations of receptor tyrosine kinases (RTKs) are associated with the development of many cancers by modifying receptor signaling and contributing to drug resistance in clinical settings. We present enhanced bystander bioluminescence resonance energy transfer-based biosensors providing new insights into RTK biology and pharmacology critical for the development of more effective RTK-targeting drugs. Distinct SH2-specific effector biosensors allow for real-time and spatiotemporal monitoring of signal transduction pathways engaged upon RTK activation. Using EGFR as a model, we demonstrate the capacity of these biosensors to differentiate unique signaling signatures, with EGF and Epiregulin ligands displaying differences in efficacy, potency, and responses within different cellular compartments. We further demonstrate that EGFR single point mutations found in Glioblastoma or non-small cell lung cancer, impact the constitutive activity of EGFR and response to tyrosine kinase inhibitor. The BRET-based biosensors are compatible with microscopy, and more importantly characterize the next generation of therapeutics directed against RTKs.


Assuntos
Técnicas Biossensoriais , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Transdução de Sinais , Receptores Proteína Tirosina Quinases/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo
5.
Med Sci (Paris) ; 29(8-9): 715-21, 2013.
Artigo em Francês | MEDLINE | ID: mdl-24005625

RESUMO

Glucose homeostasis requires a tight regulation of insulin secretion from pancreatic ß-cells. Insulin release is chiefly stimulated by glucose, but also modulated by other nutrients, including long-chain fatty acids which potentiate glucose-induced insulin secretion. The discovery of G-protein coupled receptors activated by fatty acids (and other lipid derivatives) and expressed at the surface of various cell types, including ß-cells, has added a new dimension to our understanding of the control of glucose homeostasis by fatty acids. Amongst these receptors, GPR40 and GPR119 have generated great interest as potential therapeutic targets to augment insulin secretion in type 2 diabetes. In fact, the promising results of a phase 2 clinical trial with a GPR40 agonist have provided a proof of concept for this therapeutic strategy. However, our understanding of the biology and pharmacology of these receptors remains incomplete.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Ácidos Graxos/fisiologia , Células Secretoras de Insulina/química , Receptores Acoplados a Proteínas G/fisiologia , Humanos , Células Secretoras de Insulina/fisiologia , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G/análise
6.
Nat Commun ; 14(1): 3953, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37402735

RESUMO

Urocortin 2 (UCN2) acts as a ligand for the G protein-coupled receptor corticotropin-releasing hormone receptor 2 (CRHR2). UCN2 has been reported to improve or worsen insulin sensitivity and glucose tolerance in vivo. Here we show that acute dosing of UCN2 induces systemic insulin resistance in male mice and skeletal muscle. Inversely, chronic elevation of UCN2 by injection with adenovirus encoding UCN2 resolves metabolic complications, improving glucose tolerance. CRHR2 recruits Gs in response to low concentrations of UCN2, as well as Gi and ß-Arrestin at high concentrations of UCN2. Pre-treating cells and skeletal muscle ex vivo with UCN2 leads to internalization of CRHR2, dampened ligand-dependent increases in cAMP, and blunted reductions in insulin signaling. These results provide mechanistic insights into how UCN2 regulates insulin sensitivity and glucose metabolism in skeletal muscle and in vivo. Importantly, a working model was derived from these results that unifies the contradictory metabolic effects of UCN2.


Assuntos
Resistência à Insulina , Animais , Masculino , Camundongos , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Glucose/metabolismo , Insulina , Ligantes , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Urocortinas/genética , Urocortinas/metabolismo
7.
Eur J Pharmacol ; 927: 175043, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35598847

RESUMO

Prostaglandins are bioactive lipids involved in many physiological and pathophysiological conditions, such as pain, atherosclerosis, type II diabetes, and parturition. Prostaglandin E2 (PGE2) activates four G protein-coupled receptors (GPCRs), named the PGE2 types 1-4 receptors (EP1-4), to elicit the intracellular signaling responsible for their physiological actions. There are more than twelve EP3 isoforms in humans that differ only by the sequence of their C-termini. However, the signaling mechanisms engaged by the various isoforms have never been clearly defined. In this study, we used a recently described BRET-based biosensor technology to define the signaling profiles for each of the human isoforms on a selection of signaling pathways using the agonists, PGE2 and sulprostone, and the purportedly EP3-specific antagonist L798106. We found that L798106 is a biased agonist of the Gαz pathway for some human EP3 isoforms, an effect that is not detected in the close ortholog mouse EP3 isoform α. We also found that the presence of a threonine residue at position 107 in the binding site of human EP3, which is a serine in most other species including mice, is important for L798106-mediated Gαz efficacy. Given the reported importance of EP3-Gαz signaling on the potential therapeutic efficacy of EP3 and since many preclinical studies for these mechanisms have been performed in rodents, this finding demonstrates the importance of determining a detailed signaling profile of ligands for different species and receptor isoforms, which constitutes an important step to better understand the therapeutic potential of the EP3.


Assuntos
Diabetes Mellitus Tipo 2 , Animais , Camundongos , Dor , Isoformas de Proteínas/metabolismo , Receptores de Prostaglandina E/metabolismo , Transdução de Sinais
8.
Elife ; 112022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35302494

RESUMO

Two-thirds of human hormones and one-third of clinical drugs act on membrane receptors that couple to G proteins to achieve appropriate functional responses. While G protein transducers from literature are annotated in the Guide to Pharmacology database, two recent large-scale datasets now expand the receptor-G protein 'couplome'. However, these three datasets differ in scope and reported G protein couplings giving different coverage and conclusions on G protein-coupled receptor (GPCR)-G protein signaling. Here, we report a common coupling map uncovering novel couplings supported by both large-scale studies, the selectivity/promiscuity of GPCRs and G proteins, and how the co-coupling and co-expression of G proteins compare to the families from phylogenetic relationships. The coupling map and insights on GPCR-G protein selectivity will catalyze advances in receptor research and cellular signaling toward the exploitation of G protein signaling bias in design of safer drugs.


Assuntos
Receptores Acoplados a Proteínas G , Transdução de Sinais , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Humanos , Filogenia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia
9.
Elife ; 112022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35302493

RESUMO

The recognition that individual GPCRs can activate multiple signaling pathways has raised the possibility of developing drugs selectively targeting therapeutically relevant ones. This requires tools to determine which G proteins and ßarrestins are activated by a given receptor. Here, we present a set of BRET sensors monitoring the activation of the 12 G protein subtypes based on the translocation of their effectors to the plasma membrane (EMTA). Unlike most of the existing detection systems, EMTA does not require modification of receptors or G proteins (except for Gs). EMTA was found to be suitable for the detection of constitutive activity, inverse agonism, biased signaling and polypharmacology. Profiling of 100 therapeutically relevant human GPCRs resulted in 1500 pathway-specific concentration-response curves and revealed a great diversity of coupling profiles ranging from exquisite selectivity to broad promiscuity. Overall, this work describes unique resources for studying the complexities underlying GPCR signaling and pharmacology.


Assuntos
Técnicas Biossensoriais , Proteínas de Ligação ao GTP , Técnicas Biossensoriais/métodos , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo
10.
J Biol Chem ; 285(31): 23568-80, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20489206

RESUMO

Recent studies suggest that active resolution of the inflammatory response in animal models of arthritis may involve leukotriene B(4) (LTB(4))-dependent stimulation of "intermediate" prostaglandin production, which in turn favors the synthesis of "downstream" anti-inflammatory and pro-resolving lipoxins, resolvins, and protectins. We explored a putative mechanism involving LTB(4)-dependent control of cyclooxygenase-2 (COX-2) expression, the rate-limiting step in inflammatory prostaglandin biosynthesis. Indeed, LTB(4) potently up-regulated/stabilized interleukin-1beta-induced COX-2 mRNA and protein expression under conditions of COX-2 inhibitor-dependent blockade of PGE(2) release in human synovial fibroblasts (EC(50) = 16.5 + or - 1.7 nm for mRNA; 19 + or - 2.4 nm for protein, n = 4). The latter response was pertussis toxin-sensitive, and semi-quantitative reverse transcription-PCR confirmed the quantitative predominance of the BLT2 receptor. Transfection experiments, using human COX-2 promoter plasmids and chimeric luciferase-COX-2 mRNA 3'-untranslated region (3'-UTR) reporter constructs, revealed that LTB(4) exerted its stabilizing effect at the post-transcriptional level through a 116-bp adenylate/uridylate-rich sequence in the proximal region of the COX-2 3'-UTR. Using luciferase-COX-2 mRNA 3'-UTR reporter constructs and Ras/c-Raf expression and mutant constructs, we showed that the Ras/c-Raf/MEK1/2/ERK1/2 signaling pathway mediated LTB(4)-dependent COX-2 mRNA stabilization. Knockdown experiments with specific short hairpin RNAs confirmed that LTB(4) stabilization of COX-2 mRNA was apparently mediated through the RNA-binding protein, p42 AUF1. The nuclear export of p42 AUF1 was driven by c-Raf/MEK1/2/ERK1/2 signaling and sensitive to leptomycin B treatment, suggesting a CRM1-dependent mechanism. We conclude that LTB(4) may support the resolution phase of the inflammatory response by stabilizing COX-2, ensuring a reservoir of ambient pro-resolution lipid mediators.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , Leucotrieno B4/química , Membrana Sinovial/citologia , Quinases raf/metabolismo , Proteínas ras/metabolismo , Regiões 3' não Traduzidas , Quinases Ciclina-Dependentes/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/metabolismo , Células HeLa , Ribonucleoproteína Nuclear Heterogênea D0 , Humanos , Lipídeos/química , Receptores do Leucotrieno B4/metabolismo , Transdução de Sinais , Quinase Ativadora de Quinase Dependente de Ciclina
11.
Chem Sci ; 12(33): 10990-11003, 2021 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-34522296

RESUMO

Brain functions rely on neurotransmitters that mediate communication between billions of neurons. Disruption of this communication can result in a plethora of psychiatric and neurological disorders. In this work, we combine molecular dynamics simulations, live-cell biosensor and electrophysiological assays to investigate the action of the neurotransmitter dopamine at the dopaminergic D2 receptor (D2R). The study of dopamine and closely related chemical probes reveals how neurotransmitter binding translates into the activation of distinct subsets of D2R effectors (i.e.: Gi2, GoB, Gz and ß-arrestin 2). Ligand interactions with key residues in TM5 (S5.42) and TM6 (H6.55) in the D2R binding pocket yield a dopamine-like coupling signature, whereas exclusive TM5 interaction is typically linked to preferential G protein coupling (in particular GoB) over ß-arrestin. Further experiments for serotonin receptors indicate that the reported molecular mechanism is shared by other monoaminergic neurotransmitter receptors. Ultimately, our study highlights how sequence variation in position 6.55 is used by nature to fine-tune ß-arrestin recruitment and in turn receptor signaling and internalization of neurotransmitter receptors.

12.
Front Biosci ; 11: 423-46, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16146743

RESUMO

The mammalian extracellular matrix (ECM) is a complex network of collagens, proteoglycans, glycoproteins, polysaccharides and other secreted proteins that plays fundamental structural and functional roles. In addition to its key function as an extracellular space-filling scaffold, the ECM is also implicated in the formation of important cell-cell and cell-ECM (i.e. juxtacrine) interactions that subsequently provide key regulatory signals that influence cellular proliferation and viability, differentiation, specialization and gene expression. Regulated turnover of the ECM, a process largely mediated by the tightly controlled matrix metalloprotease (MMP) enzyme family, is critical to a number of physiological processes involved in growth and development while aberrant turnover of matrix components is associated with congenital and metabolic diseases. The following review will focus on the transcriptional aspects of MMP gene expression, particularly in diseased states.


Assuntos
Desenvolvimento Ósseo , Regulação Enzimológica da Expressão Gênica , Metaloproteinases da Matriz/química , Transcrição Gênica , Aneurisma/patologia , Animais , Artrite/patologia , Aterosclerose/patologia , Vasos Sanguíneos/patologia , Osso e Ossos/metabolismo , Adesão Celular , Comunicação Celular , Sobrevivência Celular , Epitélio/patologia , Matriz Extracelular/enzimologia , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Mesoderma , Modelos Biológicos , Polissacarídeos/química , Estrutura Terciária de Proteína , Transdução de Sinais , Inibidores Teciduais de Metaloproteinases/metabolismo
13.
Mol Metab ; 5(10): 988-996, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27689011

RESUMO

OBJECTIVE: G protein-coupled receptor (GPCR) signaling regulates insulin secretion and pancreatic ß cell-proliferation. While much knowledge has been gained regarding how GPCRs are activated in ß cells, less is known about the mechanisms controlling their deactivation. In many cell types, termination of GPCR signaling is controlled by the family of Regulators of G-protein Signaling (RGS). RGS proteins are expressed in most eukaryotic cells and ensure a timely return to the GPCR inactive state upon removal of the stimulus. The aims of this study were i) to determine if RGS16, the most highly enriched RGS protein in ß cells, regulates insulin secretion and ß-cell proliferation and, if so, ii) to elucidate the mechanisms underlying such effects. METHODS: Mouse and human islets were infected with recombinant adenoviruses expressing shRNA or cDNA sequences to knock-down or overexpress RGS16, respectively. 60 h post-infection, insulin secretion and cAMP levels were measured in static incubations in the presence of glucose and various secretagogues. ß-cell proliferation was measured in infected islets after 72 h in the presence of 16.7 mM glucose ± somatostatin and various inhibitors. RESULTS: RGS16 mRNA levels are strongly up-regulated in islets of Langerhans under hyperglycemic conditions in vivo and ex vivo. RGS16 overexpression stimulated glucose-induced insulin secretion in isolated mouse and human islets while, conversely, insulin secretion was impaired following RGS16 knock-down. Insulin secretion was no longer affected by RGS16 knock-down when islets were pre-treated with pertussis toxin to inactivate Gαi/o proteins, or in the presence of a somatostatin receptor antagonist. RGS16 overexpression increased intracellular cAMP levels, and its effects were blocked by an adenylyl cyclase inhibitor. Finally, RGS16 overexpression prevented the inhibitory effect of somatostatin on insulin secretion and ß-cell proliferation. CONCLUSIONS: Our results identify RGS16 as a novel regulator of ß-cell function that coordinately controls insulin secretion and proliferation by limiting the tonic inhibitory signal exerted by δ-cell-derived somatostatin in islets.

14.
Curr Top Med Chem ; 15(24): 2528-42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26126903

RESUMO

Seven transmembrane domain receptors (7TMRs) constitute the largest family of transmembrane proteins in vertebrates and are the targets of more than 40% of currently marketed drugs. It is now accepted that these receptors are highly dynamic "microprocessors" that adopt a continuum of functionally distinct active conformations. The novel concept of biased agonism (or functional selectivity) posits that different ligands stabilize unique receptor conformations with each conformation imparting distinct signaling, and thus biological attributes, to a given receptor. The pharmacotherapeutic potential of biased agonism lies in possibility to develop molecules that selectively engage beneficial pathways while inhibiting or remaining inert towards those producing deleterious outcomes. Various strategies are now applied for the discovery of biased ligands. Many assays use second messenger levels (i.e., calcium, inositol trisphosphate, cAMP) as a quantitative readout of G-protein subtype-specific activity. However, due to complex cross-regulation between the various G-protein pathways, second messenger levels alone are not directly reflective of a ligand's activity on a specific pathway. Consequently, direct measurements of receptor-proximal events (such as G-protein activation and ß-arrestin coupling) are required for a more accurate quantification of ligand's efficacy (or bias) towards different pathways. The discovery that various ligands of the same receptor can display different efficacies and potencies towards different receptor-downstream signaling pathways has not only revitalized the process of 7TMR drug discovery, but has significantly transformed the field of pharmacology as a whole. This review will showcase the current pharmacological toolbox available for the discovery and validation of biased ligands.


Assuntos
Descoberta de Drogas , Preparações Farmacêuticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Humanos , Ligantes
15.
Trends Endocrinol Metab ; 24(8): 398-407, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23631851

RESUMO

Glucose homeostasis requires the highly coordinated regulation of insulin secretion by pancreatic ß cells. This is primarily mediated by glucose itself, but other nutrients, including free fatty acids (FFAs), potentiate the insulinotropic capacity of glucose. A decade ago, the seven-transmembrane domain receptor (7TMR) GPR40 was demonstrated to be predominantly expressed in ß cells and activated by long-chain FFAs. This discovery added a new dimension to our understanding of FFA-mediated control of glucose homeostasis. Furthermore, GPR40 has drawn considerable interest as a novel therapeutic target to enhance insulin secretion in type 2 diabetes. However, our understanding of the biology of GPR40 remains incomplete and its physiological role controversial. Here we summarize the current state of knowledge and emerging concepts regarding the role of GPR40 in regulating glucose homeostasis.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores Acoplados a Proteínas G/genética
16.
J Rheumatol ; 38(3): 419-28, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21159825

RESUMO

OBJECTIVE: It was recently reported that CD101 surface expression discriminates potency among CD4+CD25+ FoxP3+ regulatory T cells in the mouse. We investigated whether CD101 may also have a role in the suppressor function of regulatory T cells in humans given that the latter population may affect the autoimmune response in patients with rheumatoid arthritis (RA). METHODS: Sorted T cells and monocyte/macrophage cell populations were analyzed by flow cyto metry using conjugated antibodies specific for cell-surface markers. T cell proliferation assays were conducted by [(3)H]thymidine incorporation and CD8(high) cytotoxicity measurements by Cyto-Scan-LDH cytotoxicity assays. ELISA were used to measure cytokines in cell culture supernatants and Western blotting was performed for profiling mitogen-activated protein (MAP) kinase activation using specific antiphospholipid antibodies. RESULTS: CD101 expression coincided with PMA-induced monocyte/leukocyte lineage differentiation. CD8(high)CD101- T cells exhibited greater cytotoxic activity than CD8(high)CD101+ T cells, while no difference was observed between CD4CD25(high)CD101+ and CD4CD25(high)CD101- Treg inhibitory activity through responder T cells. LPS-induced proinflammatory cytokine production and p38 MAP kinase activation were made possible by ligation of CD101 with an anti-CD101 antibody F(ab')(2) fragment. CONCLUSION: These results suggested a modulatory/coregulatory function of CD101 in the human immune system, in contrast to murine models, in which CD101 surface expression discriminates potency among FoxP3+ regulatory T cells. Cytotoxic CD8(high)CD101+ T cells were markedly less cytotoxic than CD8(high) T cells negative for the CD101 antigen and were conspicuously downregulated in patients with RA, suggesting a possible role for CD101 expression and function in the control of certain manifestations of RA pathology.


Assuntos
Antígenos CD/imunologia , Artrite Reumatoide/imunologia , Macrófagos/imunologia , Glicoproteínas de Membrana/imunologia , Monócitos/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular , Proliferação de Células , Citocinas/imunologia , Humanos , Macrófagos/citologia , Camundongos , Monócitos/citologia , Subpopulações de Linfócitos T/citologia , Linfócitos T/citologia , Linfócitos T Reguladores/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
J Cell Biochem ; 101(2): 425-41, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17177291

RESUMO

Cyclooxygenase-2 (COX-2) catalyzes the rate-limiting step in inflammatory prostanoid biosynthesis. Transcriptional, post-transcriptional, and post-translational covalent modifications have been defined as important levels of regulation for COX-2 gene expression. Here, we describe a novel regulatory mechanism in primary human cells involving regulated, sequence-specific proteolysis of COX-2 that correlates with its catalytic activity and ultimately, the biosynthesis of prostaglandin E(2) (PGE(2)). Proinflammatory cytokines induced COX-2 expression and its proteolysis into stable immunoreactive fragments of 66, 42-44, 34-36, and 28 kDa. Increased COX-2 activity (PGE(2) release) was observed coincident with the timing and degree of COX-2 proteolysis with correlation analysis confirming a linear relationship (R(2) = 0.941). Inhibition of induced COX-2 activity with non-steroidal anti-inflammatory drugs (NSAIDs) and COX-2 selective inhibitors also abrogated cleavage. To determine if NSAID inhibition of proteolysis was related to drug-binding-induced conformational changes in COX-2, we assayed COX-inactive NSAID derivatives that fail to bind COX-2. Interestingly, these compounds suppressed COX-2 activity and cleavage in a correlated manner, thus suggesting that the observed NSAID-induced inhibition of COX-2 cleavage occurred through COX-independent mechanisms, presumably through the inhibition of proteases involved in COX-2 processing. Corroborating this observation, COX-2 cleavage and activity were mutually suppressed by calpain/cathepsin protease inhibitors. Our data suggest that the nascent intracellular form of COX-2 may undergo limited proteolysis to attain full catalytic capacity.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprostona/biossíntese , Inflamação/metabolismo , Animais , Anti-Inflamatórios não Esteroides/metabolismo , Artrite Juvenil/imunologia , Células Cultivadas , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/genética , Inibidores de Cisteína Proteinase/metabolismo , Dinoprostona/genética , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Membrana Sinovial/citologia
18.
Am J Physiol Renal Physiol ; 292(2): F845-52, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17003224

RESUMO

Ste20-like kinase, SLK, a germinal center kinase found in kidney epithelial cells, signals to promote apoptosis. Expression of SLK mRNA and protein and kinase activity are increased during kidney development and recovery from ischemic acute renal failure. The 3'-untranslated region (3'-UTR) of SLK mRNA contains multiple adenine and uridine-rich elements, suggesting that 3'-UTR may regulate mRNA stability. This was confirmed in COS cell transient transfection studies, which showed that expression of the SLK open-reading frame plus 3'-UTR mRNA was reduced by 35% relative to the open-reading frame alone. To further characterize the SLK-3'-UTR, this nucleotide sequence was subcloned downstream of enhanced green fluorescent protein (EGFP) cDNA. In COS, 293T, and glomerular epithelial cells, expression of EGFP mRNA and protein was markedly reduced in the presence of the SLK-3'-UTR. After transfection and subsequent addition of actinomycin D, EGFP mRNA remained stable in cells for at least 6 h, whereas EGFP-SLK-3'-UTR mRNA decayed with a half-life of approximately 4 h. A region containing five AUUUA motifs within the SLK-3'-UTR destabilized EGFP mRNA. Deletion of this region from the SLK-3'-UTR, in part, restored mRNA stability. By UV cross-linking and SDS-PAGE, the SLK-3'-UTR bound to protein(s) of approximately 30 kDa in extracts of COS cells, glomerular epithelial cells, and kidney. Cotransfection of HuR (a RNA binding protein of approximately 30 kDa) increased the steady-state mRNA level of EGFP-SLK-3'-UTR but not EGFP. Thus the SLK-3'-UTR may interact with kidney RNA-binding proteins to regulate expression of SLK mRNA during kidney development and after ischemic injury.


Assuntos
Regiões 3' não Traduzidas/fisiologia , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Animais , Células COS , Chlorocebus aethiops , Proteínas de Fluorescência Verde/biossíntese , Humanos , Rim/enzimologia , RNA Mensageiro/metabolismo
19.
J Biol Chem ; 281(29): 19849-60, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16714289

RESUMO

Cyclooxygenase-2 (COX-2) overexpression has been linked to cell survival, transformation, and hyperproliferation. We examined the regulation of the tumor suppressor gene p53 and p53 target genes by prostaglandin E(2) (PGE(2)) in human synovial fibroblasts (HSF). PGE(2) induced a time-dependent increase in p53 Ser(15) phosphorylation, with no discernible change in overall p53 levels. PGE(2)-dependent Ser(15) phosphorylation was apparently mediated by activated p38 MAP kinase as SB202190, a p38 kinase inhibitor, blocked the response. Overexpression of a MKK3 construct, but not MKK1, stimulated SB202190-sensitive p53 Ser(15) phosphorylation. PGE(2)-stimulated [phospho-Ser(15)]p53 transactivated a p53 response element (GADD45)-luciferase reporter in transiently transfected HSF (SN7); the effect was compromised by overexpression of a dominant-negative mutant (dnm) of p53 or excess p53S15A expression plasmid but mimicked by a constitutively active p53S15E expression construct. PGE(2), wtp53 expression in the presence of PGE(2), and p53S15E suppressed steady-state levels of MEKK1-induced MMP-1 mRNA, effects nullified with co-transfection of p53 dnm or p53S15A. MEKK1-induced MMP-1 promoter-driven luciferase activity was largely dependent on a c/EBPbeta-NF-kappaB-like enhancer site at -2008 to -1972 bp, as judged by deletion and point mutation analyses. PGE(2), overexpression of p53wt with PGE(2), or p53S15E abolished the MEKK1-induced MMP-1 promoter luciferase activity. Gel-shift/super gel-shift analyses identified c/EBPbeta dimers and c/EBPbeta/NF-kappaB p65 heterodimers as binding species at the apparent site of MEKK1-dependent transactivation. PGE(2)-stimulated [phospho-Ser(15)]p53 abrogated the DNA binding of c/EBPbeta dimers and c/EBPbeta/NF-kappaB p65 heterodimers. Our data suggest that COX-2 prostaglandins may be implicated in p53 function and p53 target gene expression.


Assuntos
Dinoprostona/farmacologia , MAP Quinase Quinase Quinase 1/metabolismo , Metaloproteinase 1 da Matriz/genética , Fosfosserina/metabolismo , Membrana Sinovial/metabolismo , Proteína Supressora de Tumor p53/genética , Artrite Reumatoide/metabolismo , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Primers do DNA , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Humanos , Luciferases/genética , MAP Quinase Quinase Quinase 1/genética , NF-kappa B/metabolismo , Osteoartrite/metabolismo , Fosforilação , Plasmídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membrana Sinovial/efeitos dos fármacos , Ativação Transcricional
20.
J Biol Chem ; 280(10): 9536-46, 2005 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-15640148

RESUMO

Tumor necrosis factor-alpha (TNF-alpha) is a pleiotropic proinflammatory cytokine that modulates a broad range of inflammatory and immunological processes. We have investigated the potential immunomodulatory properties of prostaglandin E2 (PGE2) by examining the molecular mechanism by which the eicosanoid suppresses T-cell-derived interleukin-17 (IL-17)-induced TNF-alpha mRNA expression and protein synthesis in human macrophages and rheumatoid arthritis-affected synovial fibroblasts. Initial studies confirmed that PGE2 induces egr-1 mRNA expression and protein synthesis by restricted SAPK2/p38 MAPK-dependent activating transcription factor-2 (ATF-2) dimer transactivation of the egr-1 promoter as judged by studies using wild-type (WT) and deletion mutant egr-1 promoter constructs, Northern and Western blotting, and standard and supershift electrophoretic mobility shift analyses. Using human leukemic monocytic THP-1 cells stably transfected with WT and dominant-negative mutant expression constructs of Egr-1, cotransfected or not with a WT pTNF-615SVOCAT construct, we observed that PGE2 inhibition of IL-17-stimulated TNF-alpha mRNA expression and promoter activity was dependent on Egr-1 expression, as mutants of Egr-1, alone or in combination, markedly abrogated any inhibitory effect of PGE2. Standard and supershift electrophoretic mobility shift analysis, signaling "decoy" overexpression studies, and pTNF-615SVOCAT promoter assays using WT and mutant promoter constructs revealed that IL-17-up-regulated promoter activity was largely dependent on ATF-2/c-Jun transactivation. PGE2 suppression of IL-17-induced ATF-2/c-Jun transactivation and DNA binding was dependent on Egr-1-mediated inhibition of induced c-Jun expression. We suggest that egr-1 is an immediate-early PGE2 target gene that may be a key regulatory factor in mediating eicosanoid control of genes involved in the immune and inflammatory responses.


Assuntos
Artrite Reumatoide/imunologia , Proteínas de Ligação a DNA/genética , Dinoprostona/farmacologia , Proteínas Imediatamente Precoces/genética , Fatores de Transcrição/genética , Células 3T3 , Animais , Artrite Reumatoide/sangue , Células Cultivadas , Proteína 1 de Resposta de Crescimento Precoce , Fibroblastos/imunologia , Fibroblastos/fisiologia , Células HeLa , Humanos , Interleucina-17/farmacologia , Macrófagos/imunologia , Camundongos , Monócitos/imunologia , RNA Mensageiro/genética , Proteínas Recombinantes/farmacologia , Membrana Sinovial/patologia , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA