Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(1): e2307086120, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38147543

RESUMEN

The salt-inducible kinases (SIK) 1-3 are key regulators of pro- versus anti-inflammatory cytokine responses during innate immune activation. The lack of highly SIK-family or SIK isoform-selective inhibitors suitable for repeat, oral dosing has limited the study of the optimal SIK isoform selectivity profile for suppressing inflammation in vivo. To overcome this challenge, we devised a structure-based design strategy for developing potent SIK inhibitors that are highly selective against other kinases by engaging two differentiating features of the SIK catalytic site. This effort resulted in SIK1/2-selective probes that inhibit key intracellular proximal signaling events including reducing phosphorylation of the SIK substrate cAMP response element binding protein (CREB) regulated transcription coactivator 3 (CRTC3) as detected with an internally generated phospho-Ser329-CRTC3-specific antibody. These inhibitors also suppress production of pro-inflammatory cytokines while inducing anti-inflammatory interleukin-10 in activated human and murine myeloid cells and in mice following a lipopolysaccharide challenge. Oral dosing of these compounds ameliorates disease in a murine colitis model. These findings define an approach to generate highly selective SIK1/2 inhibitors and establish that targeting these isoforms may be a useful strategy to suppress pathological inflammation.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Proteínas Serina-Treonina Quinasas , Ratones , Humanos , Animales , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citocinas , Inflamación/tratamiento farmacológico , Isoformas de Proteínas , Antiinflamatorios/farmacología , Inmunidad Innata , Factores de Transcripción
2.
Bioorg Med Chem Lett ; 96: 129492, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37778428

RESUMEN

Natural killer group 2D (NKG2D) is a homodimeric activating immunoreceptor whose function is to detect and eliminate compromised cells upon binding to the NKG2D ligands (NKG2DL) major histocompatibility complex (MHC) molecules class I-related chain A (MICA) and B (MICB) and UL16 binding proteins (ULBP1-6). While typically present at low levels in healthy cells and tissue, NKG2DL expression can be induced by viral infection, cellular stress or transformation. Aberrant activity along the NKG2D/NKG2DL axis has been associated with autoimmune diseases due to the increased expression of NKG2D ligands in human disease tissue, making NKG2D inhibitors an attractive target for immunomodulation. Herein we describe the discovery and optimization of small molecule PPI (protein-protein interaction) inhibitors of NKG2D/NKG2DL. Rapid SAR was guided by structure-based drug design and accomplished by iterative singleton and parallel medicinal chemistry synthesis. These efforts resulted in the identification of several potent analogs (14, 21, 30, 45) with functional activity and improved LLE.


Asunto(s)
Proteínas Portadoras , Subfamilia K de Receptores Similares a Lectina de Células NK , Humanos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Proteínas Portadoras/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Unión Proteica , Células Asesinas Naturales/metabolismo , Ligandos
3.
J Pharmacol Exp Ther ; 386(1): 35-44, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37142444

RESUMEN

Inflammatory bowel disease (IBD) is characterized by chronic mucosal inflammation of the gastrointestinal tract and is associated with extracellular acidification of mucosal tissue. Several extracellular pH-sensing receptors, including G protein-coupled receptor 4 (GPR4), play an important role in the regulation of inflammatory and immune responses, and GPR4 deficiency has been shown to be protective in IBD animal models. To confirm the therapeutic potential of GPR4 antagonism in IBD, we tested Compound 13, a selective GPR4 antagonist, in the interleukin 10-/- mouse model of colitis. Despite good exposures and albeit there was a trend toward improvement for a few readouts, Compound 13 treatment did not improve colitis in this model, and there were no signs of target engagement. Interestingly, Compound 13 behaved as an "orthosteric" antagonist, i.e., its potency was pH dependent and mostly inactive at pH levels lower than 6.8 with preferential binding to the inactive conformation of GPR4. Mutagenesis studies confirmed Compound 13 likely binds to the conserved orthosteric binding site in G protein-coupled receptors, where a histidine sits in GPR4 likely preventing Compound 13 binding when protonated in acidic conditions. While the exact mucosal pH in the human disease and relevant IBD mice models is unknown, it is well established that the degree of acidosis is positively correlated with the degree of inflammation, suggesting Compound 13 is not an ideal tool to study the role of GPR4 in moderate to severe inflammatory conditions. SIGNIFICANCE STATEMENT: Compound 13, a reported selective GPR4 antagonist, has been widely used to assess the therapeutic potential of GPR4, a pH-sensing receptor, for numerous indications. Its pH dependence and mechanism of inhibition identified in this study clearly highlights the limitations of this chemotype for target validation.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Humanos , Animales , Ratones , Receptores Acoplados a Proteínas G/metabolismo , Colitis/metabolismo , Inflamación , Concentración de Iones de Hidrógeno , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico
4.
Proc Natl Acad Sci U S A ; 120(18): e2216342120, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-37098070

RESUMEN

NKG2D (natural-killer group 2, member D) is a homodimeric transmembrane receptor that plays an important role in NK, γδ+, and CD8+ T cell-mediated immune responses to environmental stressors such as viral or bacterial infections and oxidative stress. However, aberrant NKG2D signaling has also been associated with chronic inflammatory and autoimmune diseases, and as such NKG2D is thought to be an attractive target for immune intervention. Here, we describe a comprehensive small-molecule hit identification strategy and two distinct series of protein-protein interaction inhibitors of NKG2D. Although the hits are chemically distinct, they share a unique allosteric mechanism of disrupting ligand binding by accessing a cryptic pocket and causing the two monomers of the NKG2D dimer to open apart and twist relative to one another. Leveraging a suite of biochemical and cell-based assays coupled with structure-based drug design, we established tractable structure-activity relationships with one of the chemical series and successfully improved both the potency and physicochemical properties. Together, we demonstrate that it is possible, albeit challenging, to disrupt the interaction between NKG2D and multiple protein ligands with a single molecule through allosteric modulation of the NKG2D receptor dimer/ligand interface.


Asunto(s)
Células Asesinas Naturales , Subfamilia K de Receptores Similares a Lectina de Células NK , Ligandos , Linfocitos T CD8-positivos , Unión Proteica
5.
Eur J Med Chem ; 236: 114330, 2022 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-35436670

RESUMEN

Pramlintide is an equipotent amylin analogue that reduces food intake and body weight in obese subjects and has been clinically approved as an adjunctive therapy for the treatment of adult diabetic patients. However, due to its extremely short half-life in vivo, a regimen of multiple daily administrations is required for achieving clinical effectiveness. Herein is described the development of prototypical long-acting pramlintide bioconjugates, in which pramlintide's disulfide-linked macrocycle was replaced by a cyclic thioether motif. This modification enabled stable chemical conjugation to a half-life extending antibody. In contrast to pramlintide (t1/2 < 0.75 h), bioconjugates 35 and 38 have terminal half-lives of ∼2 days in mice and attain significant exposure levels that are maintained up to 7 days. Single dose subcutaneous administration of 35 in lean mice, given 18-20 h prior to oral acetaminophen (AAP) administration, significantly reduced gastric emptying (as determined by plasma AAP levels). In a separate study, similar administration of 35 in fasted lean mice effected a reduction in food intake for up to 48 h. These data are consistent with durable amylinomimetic responses and provide the basis for further development of such long-acting amylinomimetic conjugates for the potential treatment of obesity and associated pathologies.


Asunto(s)
Agonistas de los Receptores de Amilina , Agonistas de los Receptores de Amilina/farmacología , Agonistas de los Receptores de Amilina/uso terapéutico , Amiloide , Animales , Peso Corporal , Humanos , Hipoglucemiantes/uso terapéutico , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Ratones , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico
6.
J Biol Chem ; 295(13): 4359-4366, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32079674

RESUMEN

Excitatory amino acid transporters (EAATs) represent a protein family that is an emerging drug target with great therapeutic potential for managing central nervous system disorders characterized by dysregulation of glutamatergic neurotransmission. As such, it is of significant interest to discover selective modulators of EAAT2 function. Here, we applied computational methods to identify specific EAAT2 inhibitors. Utilizing a homology model of human EAAT2, we identified a binding pocket at the interface of the transport and trimerization domain. We next conducted a high-throughput virtual screen against this site and identified a selective class of EAAT2 inhibitors that were tested in glutamate uptake and whole-cell electrophysiology assays. These compounds represent potentially useful pharmacological tools suitable for further exploration of the therapeutic potential of EAAT2 and may provide molecular insights into mechanisms of allosteric modulation for glutamate transporters.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/antagonistas & inhibidores , Sitios de Unión/efectos de los fármacos , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos X-AG/química , Sistema de Transporte de Aminoácidos X-AG/genética , Animales , Sitios de Unión/genética , Transporte Biológico/efectos de los fármacos , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/patología , Biología Computacional , Transportador 2 de Aminoácidos Excitadores/química , Transportador 2 de Aminoácidos Excitadores/genética , Humanos , Unión Proteica/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Interfaz Usuario-Computador
7.
iScience ; 19: 110-118, 2019 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-31369984

RESUMEN

Polymorphism in the microglial receptor CD33 gene has been linked to late-onset Alzheimer disease (AD), and reduced expression of the CD33 sialic acid-binding domain confers protection. Thus, CD33 inhibition might be an effective therapy against disease progression. Progress toward discovery of selective CD33 inhibitors has been hampered by the absence of an atomic resolution structure. We report here the crystal structures of CD33 alone and bound to a subtype-selective sialic acid mimetic called P22 and use them to identify key binding residues by site-directed mutagenesis and binding assays to reveal the molecular basis for its selectivity toward sialylated glycoproteins and glycolipids. We show that P22, when presented on microparticles, increases uptake of the toxic AD peptide, amyloid-ß (Aß), into microglial cells. Thus, the sialic acid-binding site on CD33 is a promising pharmacophore for developing therapeutics that promote clearance of the Aß peptide that is thought to cause AD.

8.
Mol Pharmacol ; 93(6): 581-591, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29572336

RESUMEN

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.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Glucosa/metabolismo , Secreción de Insulina/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Benzofuranos/farmacología , Células CHO , Línea Celular , Cricetulus , Células HEK293 , Humanos , Hipoglucemiantes/farmacología , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Proteína Quinasa C/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sulfonas/farmacología
9.
Biochem Pharmacol ; 135: 1-11, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28214518

RESUMEN

Maintaining the integrity of cellular membranes is critical to protecting metabolic activities and genetic information from the environment. Regulation of transport across membranes of essential chemicals, including water, nutrients, hormones and many drugs, is therefore key to cellular homeostasis and physiological processes. The two main transporter superfamilies are ATP-binding cassette (ABC) transporters that primarily function as efflux transporters, and the solute carrier (SLC) transporters. SLC transporters encompass 52 gene families with almost 400 different human transporter genes. Although long under-explored, SLC transporters are an emerging drug target class and the molecular target of several approved inhibitor drugs, such as selective serotonin reuptake inhibitors (SSRIs) for depression and sodium/glucose co-transporter (SGLT2) inhibitors for diabetes. Interestingly though, although loss-of-function mutations in numerous human SLC transporters are linked to Mendelian diseases, few reports of SLC transporter activators have appeared, and only inhibitors have been advanced to clinical studies. In this commentary, we discuss several strategies for potentiating SLC transporter function, from direct acting potentiators to modulators of transcription, translation or trafficking. We review the progress made in recent years toward the understanding of the structural and molecular basis of SLC transporter function and the pathways and mechanisms that regulate SLC expression, and describe the opportunities these new insights present for discovery of SLC transporter potentiators. Finally, we highlight the challenges associated with the various approaches and provide some thoughts on future directions that might facilitate the search for SLC potentiators with therapeutic potential.


Asunto(s)
Descubrimiento de Drogas/métodos , Proteínas de Transporte de Membrana/metabolismo , Proteínas Transportadoras de Solutos/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Descubrimiento de Drogas/tendencias , Humanos , Proteínas de Transporte de Membrana/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Inhibidores Selectivos de la Recaptación de Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Proteínas Transportadoras de Solutos/química
10.
Mol Pharmacol ; 90(6): 766-774, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27754898

RESUMEN

In the liver, citrate is a key metabolic intermediate involved in the regulation of glycolysis and lipid synthesis and reduced expression of the hepatic citrate SLC13A5 transporter has been shown to improve metabolic outcomes in various animal models. Although inhibition of hepatic extracellular citrate uptake through SLC13A5 has been suggested as a potential therapeutic approach for Type-2 diabetes and/or fatty liver disease, so far, only a few SLC13A5 inhibitors have been identified. Moreover, their mechanism of action still remains unclear, potentially limiting their utility for in vivo proof-of-concept studies. In this study, we characterized the pharmacology of the recently identified hydroxysuccinic acid SLC13A5 inhibitors, PF-06649298 and PF-06761281, using a combination of 14C-citrate uptake, a membrane potential assay and electrophysiology. In contrast to their previously proposed mechanism of action, our data suggest that both PF-06649298 and PF-06761281 are allosteric, state-dependent SLC13A5 inhibitors, with low-affinity substrate activity in the absence of citrate. As allosteric state-dependent modulators, the inhibitory potency of both compounds is highly dependent on the ambient citrate concentration and our detailed mechanism of action studies therefore, may be of value in interpreting the in vivo effects of these compounds.


Asunto(s)
Malatos/farmacología , Fenilbutiratos/farmacología , Piridinas/farmacología , Succinatos/farmacología , Simportadores/antagonistas & inhibidores , Regulación Alostérica/efectos de los fármacos , Radioisótopos de Carbono , Ácido Cítrico/metabolismo , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Malatos/química , Modelos Biológicos , Técnicas de Placa-Clamp , Fenilbutiratos/química , Piridinas/química , Especificidad por Sustrato/efectos de los fármacos , Succinatos/química , Simportadores/metabolismo
11.
Expert Opin Drug Discov ; 10(8): 825-39, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25967138

RESUMEN

INTRODUCTION: Obesity is a body weight disorder characterized by excess adiposity that increases the risk for developing co-morbidities such as type 2 diabetes. A large medical need exists for new anti-obesity treatments capable of promoting 10% or greater weight loss, with minimal side effects. AREAS COVERED: The authors describe the application of monogenic forms of rare obesity and genome-wide association studies in selecting critical pathways for drug discovery. Furthermore, they review in detail several pathways and pharmacological targets in the central nervous system (e.g., the leptin-melanocortin axis, the opioid system, GLP-1/GLP-1 system, and FGF21/FGFR1c/ß-Klotho axis) that play an important role in the regulation of feeding behavior and energy homeostasis. Special focus is given to new strategies that engage well-known targets via novel mechanisms in order to circumvent issues seen with previous drug candidates that failed in the clinic. Finally, the authors discuss the recent developments around fixed-dose combinations, targeted polypharmacology, and non-traditional combinations of drugs and devices. EXPERT OPINION: The future for new weight-loss approaches to treat obesity looks promising. Current therapies have shown modest effects on weight loss in the general obese population but will have greater impact in smaller homogeneous sub-populations of obese subjects using personalized medicine. Drug combinations that target multiple, complementary pathways have the potential to promote double-digit weight loss in a broader, heterogeneous patient population. Furthermore, the development of advanced subcutaneous delivery technologies has opened up opportunities to develop breakthrough peptide and biologic agents for the treatment of obesity.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Animales , Fármacos Antiobesidad/efectos adversos , Fármacos Antiobesidad/farmacología , Diseño de Fármacos , Quimioterapia Combinada , Humanos , Terapia Molecular Dirigida , Obesidad/complicaciones , Obesidad/fisiopatología
12.
Mol Pharmacol ; 85(1): 83-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24113749

RESUMEN

The κ-opioid receptor (KOR)-dynorphin system has been implicated in the control of affect, cognition, and motivation, and is thought to be dysregulated in mood and psychotic disorders, as well as in various phases of opioid dependence. KOR agonists exhibit analgesic effects, although the adverse effects produced by some KOR agonists, including sedation, dysphoria, and hallucinations, have limited their clinical use. Interestingly, KOR-mediated dysphoria, assessed in rodents as aversion, has recently been attributed to the activation of the p38 mitogen-activated protein kinase pathway following arrestin recruitment to the activated KOR. Therefore, KOR-selective G protein-biased agonists, which do not recruit arrestin, have been proposed to be more effective analgesics, without the adverse effects triggered by the arrestin pathway. As an initial step toward identifying novel biased KOR agonists, we applied a multifaceted screening strategy utilizing both in silico and parallel screening approaches. We identified several KOR-selective ligand scaffolds with a range of signaling bias in vitro. The arylacetamide-based scaffold includes both G protein- and ß-arrestin-biased ligands, while the endogenous peptides and the diterpene scaffolds are G protein biased. Interestingly, we found scaffold screening to be more successful than library screening in identifying biased ligands. Many of the identified functionally selective ligands are potent selective KOR agonists that are reported to be active in the central nervous system. They therefore represent excellent candidates for in vivo studies aiming at determining the behavioral effects mediated by specific KOR-mediated signaling cascades.


Asunto(s)
Analgésicos Opioides/química , Receptores Opioides kappa/agonistas , Acetamidas/química , Acetamidas/farmacología , Analgésicos Opioides/farmacología , Arrestinas/metabolismo , Simulación por Computador , Bases de Datos de Compuestos Químicos , Diterpenos/química , Diterpenos/farmacología , Dinorfinas/química , Dinorfinas/farmacología , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Transporte de Proteínas , Receptores Opioides kappa/química , Receptores Opioides kappa/metabolismo , Transducción de Señal , Relación Estructura-Actividad , beta-Arrestinas
14.
J Chem Inf Model ; 53(3): 521-6, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23461591

RESUMEN

Kappa-opioid (KOP) receptor agonists exhibit analgesic effects without activating reward pathways. In the search for nonaddictive opioid therapeutics and novel chemical tools to study physiological functions regulated by the KOP receptor, we screened in silico its recently released inactive crystal structure. A selective novel KOP receptor agonist emerged as a notable result and is proposed as a new chemotype for the study of the KOP receptor in the etiology of drug addiction, depression, and/or pain.


Asunto(s)
Descubrimiento de Drogas/métodos , Receptores Opioides kappa/agonistas , Arrestina/metabolismo , Cristalografía por Rayos X , AMP Cíclico/metabolismo , ADN Complementario/biosíntesis , ADN Complementario/genética , Bases de Datos Factuales , Proteínas de Unión al GTP/metabolismo , Vectores Genéticos , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Conformación Molecular , Receptores Opioides kappa/genética , Estereoisomerismo , Relación Estructura-Actividad , Transfección , Interfaz Usuario-Computador
15.
J Biol Chem ; 287(32): 27050-4, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22736766

RESUMEN

κ-Opioid receptor (KOR) agonists do not activate the reward pathway stimulated by morphine-like µ-opioid receptor (MOR) agonists and thus have been considered to be promising nonaddictive analgesics. However, KOR agonists produce other adverse effects, including dysphoria, diuresis, and constipation. The therapeutic promise of KOR agonists has nonetheless recently been revived by studies showing that their dysphoric effects require arrestin recruitment, whereas their analgesic effects do not. Moreover, KOR agonist-induced antinociceptive tolerance observed in vivo has also been proposed to be correlated to the ability to induce arrestin-dependent phosphorylation, desensitization, and internalization of the receptor. The discovery of functionally selective drugs that are therapeutically effective without the adverse effects triggered by the arrestin pathway is thus an important goal. We have identified such an extreme G protein-biased KOR compound, 6'-guanidinonaltrindole (6'-GNTI), a potent partial agonist at the KOR receptor for the G protein activation pathway that does not recruit arrestin. Indeed, 6'-GNTI functions as an antagonist to block the arrestin recruitment and KOR internalization induced by other nonbiased agonists. As an extremely G protein-biased KOR agonist, 6'-GNTI represents a promising lead compound in the search for nonaddictive opioid analgesic as its signaling profile suggests that it will be without the dysphoria and other adverse effects promoted by arrestin recruitment and its downstream signaling.


Asunto(s)
Arrestina/metabolismo , Proteínas de Unión al GTP/metabolismo , Guanidinas/farmacología , Naltrexona/análogos & derivados , Receptores Opioides kappa/antagonistas & inhibidores , Citometría de Flujo , Naltrexona/farmacología
16.
Biotechniques ; 51(2): 111-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21806555

RESUMEN

The existence of G protein-coupled receptor (GPCR) dimers and/or oligomers has been demonstrated in heterologous systems using a variety of biochemical and biophysical assays. While these interactions are the subject of intense research because of their potential role in modulating signaling and altering pharmacology, evidence for the existence of receptor interactions in vivo is still elusive because of a lack of appropriate methods to detect them. Here, we adapted and optimized a proximity ligation assay (PLA) for the detection in brain slices of molecular proximity of two antigens located on either the same or two different GPCRs. Using this approach, we were able to confirm the existence of dopamine D2 and adenosine A2A receptor complexes in the striatum of mice ex vivo.


Asunto(s)
Cuerpo Estriado/química , Immunoblotting/métodos , Inmunohistoquímica/métodos , Receptor de Adenosina A2A/análisis , Receptores de Dopamina D2/análisis , Análisis de Varianza , Animales , Anticuerpos/química , Anticuerpos/metabolismo , Cuerpo Estriado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Receptor de Adenosina A2A/química , Receptor de Adenosina A2A/metabolismo , Receptores de Dopamina D2/química , Receptores de Dopamina D2/metabolismo
17.
Nat Chem Biol ; 6(8): 587-94, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20622858

RESUMEN

G protein-coupled receptor (GPCR) oligomers have been proposed to play critical roles in cell signaling, but confirmation of their existence in a native context remains elusive, as no direct interactions between receptors have been reported. To demonstrate their presence in native tissues, we developed a time-resolved FRET strategy that is based on receptor labeling with selective fluorescent ligands. Specific FRET signals were observed with four different receptors expressed in cell lines, consistent with their dimeric or oligomeric nature in these transfected cells. More notably, the comparison between FRET signals measured with sets of fluorescent agonists and antagonists was consistent with an asymmetric relationship of the two protomers in an activated GPCR dimer. Finally, we applied the strategy to native tissues and succeeded in demonstrating the presence of oxytocin receptor dimers and/or oligomers in mammary gland.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Oligopéptidos/química , Receptores Acoplados a Proteínas G/metabolismo , Algoritmos , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas , Células COS , Línea Celular , Chlorocebus aethiops , Dimerización , Antagonistas de los Receptores de Dopamina D2 , Femenino , Colorantes Fluorescentes , Ligandos , Glándulas Mamarias Animales/metabolismo , Modelos Moleculares , Oligopéptidos/metabolismo , Ensayo de Unión Radioligante , Ratas , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Oxitocina/agonistas , Receptores de Oxitocina/antagonistas & inhibidores , Receptores de Oxitocina/metabolismo , Receptores de Vasopresinas/agonistas , Receptores de Vasopresinas/metabolismo
18.
Curr Opin Pharmacol ; 10(1): 6-13, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19962942

RESUMEN

Communication between cells requires key and specialized signaling systems, like G-protein-coupled receptors (GPCRs). Cells coexpress a large number of different GPCRs, the activation of which generates multiple signals that are integrated via mechanisms still not well understood. The Class C GPCRs like the metabotropic receptors for glutamate (mGlu), GABA (GABA(B)), or calcium ions (CaSR), have been shown to functionally crosstalk with other receptor systems, leading to synergistic or new signaling responses involved in important physiological functions. The Class C GPCRs are well-known dimeric receptors, either homodimeric (mGlu or CaSR) or heterodimeric (GABA(B) or taste T1R1/T1R3 and T1R2/T1R3) receptors. Moreover, they have been reported to form oligomeric complexes themselves or associated to other receptors. As the receptor oligomerization often affect binding, activity, or signaling of GPCRs, the formation of receptor heteromers has been used as an explanation for many of the described crosstalk involving these receptors. Here, we will discuss that crosstalk could result not only from receptor oligomerization, but also from colocalized receptor sharing signaling pathways, or from synergistic regulation of signaling crossroads, independently of oligomerization.


Asunto(s)
Receptor Cross-Talk , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Humanos , Unión Proteica/fisiología , Multimerización de Proteína , Receptores Acoplados a Proteínas G/química
19.
EMBO J ; 28(15): 2195-208, 2009 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-19590495

RESUMEN

G protein-coupled receptors (GPCRs) have critical functions in intercellular communication. Although a wide range of different receptors have been identified in the same cells, the mechanism by which signals are integrated remains elusive. The ability of GPCRs to form dimers or larger hetero-oligomers is thought to generate such signal integration. We examined the molecular mechanisms responsible for the GABA(B) receptor-mediated potentiation of the mGlu receptor signalling reported in Purkinje neurons. We showed that this effect does not require a physical interaction between both receptors. Instead, it is the result of a more general mechanism in which the betagamma subunits produced by the Gi-coupled GABA(B) receptor enhance the mGlu-mediated Gq response. Most importantly, this mechanism could be generally applied to other pairs of Gi- and Gq-coupled receptors and the signal integration varied depending on the time delay between activation of each receptor. Such a mechanism helps explain specific properties of cells expressing two different Gi- and Gq-coupled receptors activated by a single transmitter, or properties of GPCRs naturally coupled to both types of the G protein.


Asunto(s)
Células de Purkinje/fisiología , Receptores de GABA-B/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal , Animales , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos
20.
Nat Methods ; 5(6): 561-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18488035

RESUMEN

Cell-surface proteins are important in cell-cell communication. They assemble into heterocomplexes that include different receptors and effectors. Elucidation and manipulation of such protein complexes offers new therapeutic possibilities. We describe a methodology combining time-resolved fluorescence resonance energy transfer (FRET) with snap-tag technology to quantitatively analyze protein-protein interactions at the surface of living cells, in a high throughput-compatible format. Using this approach, we examined whether G protein-coupled receptors (GPCRs) are monomers or assemble into dimers or larger oligomers--a matter of intense debate. We obtained evidence for the oligomeric state of both class A and class C GPCRs. We also observed different quaternary structure of GPCRs for the neurotransmitters glutamate and gamma-aminobutyric acid (GABA): whereas metabotropic glutamate receptors assembled into strict dimers, the GABA(B) receptors spontaneously formed dimers of heterodimers, offering a way to modulate G-protein coupling efficacy. This approach will be useful in systematic analysis of cell-surface protein interaction in living cells.


Asunto(s)
Biofisica/métodos , Membrana Celular/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Mapeo de Interacción de Proteínas/métodos , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células COS , Chlorocebus aethiops , Dimerización , Humanos , Modelos Biológicos , Estructura Cuaternaria de Proteína , Receptores de GABA-B/química , Propiedades de Superficie , Ácido gamma-Aminobutírico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA