Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Psychiatry ; 27(5): 2425-2438, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35393556

RESUMEN

Latrophilin-3 (Lphn3; also known as ADGRL3) is a member of the adhesion G Protein Coupled Receptor subfamily, which participates in the stabilization and maintenance of neuronal networks by mediating intercellular adhesion through heterophilic interactions with transmembrane ligands. Polymorphisms modifying the Lphn3 gene are associated with attention-deficit/hyperactivity disorder (ADHD) in children and its persistence into adulthood. How these genetic alterations affect receptor function remains unknown. Here, we conducted the functional validation of distinct ADHD-related Lphn3 variants bearing mutations in the receptor's adhesion motif-containing extracellular region. We found that all variants tested disrupted the ability of Lphn3 to stabilize intercellular adhesion in a manner that was distinct between ligands classes, but which did not depend on ligand-receptor interaction parameters, thus pointing to altered intrinsic receptor signaling properties. Using G protein signaling biosensors, we determined that Lphn3 couples to Gαi1, Gαi2, Gαs, Gαq, and Gα13. However, all ADHD-related receptor variants consistently lacked intrinsic as well as ligand-dependent Gα13 coupling efficiency while maintaining unaltered coupling to Gαi, Gαs, and Gαq. Consistent with these alterations, actin remodeling functions as well as actin-relevant RhoA signaling normally displayed by the constitutively active Lphn3 receptor were impeded by select receptor variants, thus supporting additional signaling defects. Taken together, our data point to Gα13 selective signaling impairments as representing a disease-relevant pathogenicity pathway that can be inherited through Lphn3 gene polymorphisms. This study highlights the intricate interplay between Lphn3 GPCR functions and the actin cytoskeleton in modulating neurodevelopmental cues related to ADHD etiology.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Actinas , Adulto , Trastorno por Déficit de Atención con Hiperactividad/genética , Niño , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Humanos , Ligandos , Receptores Acoplados a Proteínas G/genética , Virulencia
2.
Int J Mol Sci ; 23(5)2022 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-35269644

RESUMEN

Transient receptor potential canonical (TRPC) channels are membrane proteins involved in regulating Ca2+ homeostasis, and whose functions are modulated by G protein-coupled receptors (GPCR). In this study, we developed bioluminescent resonance energy transfer (BRET) biosensors to better study channel conformational changes following receptor activation. For this study, two intramolecular biosensors, GFP10-TRPC7-RLucII and RLucII-TRPC7-GFP10, were constructed and were assessed following the activation of various GPCRs. We first transiently expressed receptors and the biosensors in HEK293 cells, and BRET levels were measured following agonist stimulation of GPCRs. The activation of GPCRs that engage Gαq led to a Gαq-dependent BRET response of the functional TRPC7 biosensor. Focusing on the Angiotensin II type-1 receptor (AT1R), GFP10-TRPC7-RLucII was tested in rat neonatal cardiac fibroblasts, expressing endogenous AT1R and TRPC7. We detected similar BRET responses in these cells, thus validating the use of the biosensor in physiological conditions. Taken together, our results suggest that activation of Gαq-coupled receptors induce conformational changes in a novel and functional TRPC7 BRET biosensor.


Asunto(s)
Transferencia de Energía por Resonancia de Bioluminiscencia , Técnicas Biosensibles , Animales , Transferencia de Energía por Resonancia de Bioluminiscencia/métodos , Técnicas Biosensibles/métodos , Células HEK293 , Humanos , Ratas , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
3.
Mol Pharmacol ; 87(6): 982-95, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25808928

RESUMEN

The octapeptide angiotensin II (AngII) exerts a variety of cardiovascular effects through the activation of the AngII type 1 receptor (AT1), a G protein-coupled receptor. The AT1 receptor engages and activates several signaling pathways, including heterotrimeric G proteins Gq and G12, as well as the extracellular signal-regulated kinases (ERK) 1/2 pathway. Additionally, following stimulation, ßarrestin is recruited to the AT1 receptor, leading to receptor desensitization. It is increasingly recognized that specific ligands selectively bind and favor the activation of some signaling pathways over others, a concept termed ligand bias or functional selectivity. A better understanding of the molecular basis of functional selectivity may lead to the development of better therapeutics with fewer adverse effects. In the present study, we developed assays allowing the measurement of six different signaling modalities of the AT1 receptor. Using a series of AngII peptide analogs that were modified in positions 1, 4, and 8, we sought to better characterize the molecular determinants of AngII that underlie functional selectivity of the AT1 receptor in human embryonic kidney 293 cells. The results reveal that position 1 of AngII does not confer functional selectivity, whereas position 4 confers a bias toward ERK signaling over Gq signaling, and position 8 confers a bias toward ßarrestin recruitment over ERK activation and Gq signaling. Interestingly, the analogs modified in position 8 were also partial agonists of the protein kinase C (PKC)-dependent ERK pathway via atypical PKC isoforms PKCζ and PKCι.


Asunto(s)
Angiotensina II/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina II/química , Arrestinas/metabolismo , Activación Enzimática , Receptores ErbB/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Isoenzimas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Oligopéptidos/síntesis química , Oligopéptidos/química , Oligopéptidos/metabolismo , Proteína Quinasa C/metabolismo , Receptor de Angiotensina Tipo 1/química , Transducción de Señal , beta-Arrestinas
4.
J Biol Chem ; 285(4): 2284-93, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19940150

RESUMEN

The octapeptide hormone angiotensin II (AngII) exerts a wide variety of cardiovascular effects through the activation of the AT(1) receptor, which belongs to the G protein-coupled receptor superfamily. Like other G protein-coupled receptors, the AT(1) receptor possesses seven transmembrane domains that provide structural support for the formation of the ligand-binding pocket. Here, we investigated the role of the first and fourth transmembrane domains (TMDs) in the formation of the binding pocket of the human AT(1) receptor using the substituted-cysteine accessibility method. Each residue within the Phe-28((1.32))-Ile-53((1.57)) fragment of TMD1 and Leu-143((4.40))-Phe-170((4.67)) fragment of TMD4 was mutated, one at a time, to a cysteine. The resulting mutant receptors were expressed in COS-7 cells, which were subsequently treated with the charged sulfhydryl-specific alkylating agent methanethiosulfonate ethylammonium (MTSEA). This treatment led to a significant reduction in the binding affinity of TMD1 mutants M30C((1.34))-AT(1) and T33C((1.37))-AT(1) and TMD4 mutant V169C((4.66))-AT(1). Although this reduction in binding of the TMD1 mutants was maintained when examined in a constitutively active receptor (N111G-AT(1)) background, we found that V169C((4.66))-AT(1) remained unaffected when treated with MTSEA compared with untreated in this context. Moreover, the complete loss of binding observed for R167C((4.64))-AT(1) was restored upon treatment with MTSEA. Our results suggest that the extracellular portion of TMD1, particularly residues Met-30((1.34)) and Thr-33((1.37)), as well as residues Arg-167((4.64)) and Val-169((4.66)) at the junction of TMD4 and the second extracellular loop, are important binding determinants within the AT(1) receptor binding pocket but that these TMDs undergo very little movement, if at all, during the activation process.


Asunto(s)
Angiotensina II/metabolismo , Proteínas de la Membrana , Receptor de Angiotensina Tipo 1 , Animales , Arginina/genética , Arginina/metabolismo , Sitios de Unión , Células COS , Chlorocebus aethiops , Cisteína/genética , Cisteína/metabolismo , Metanosulfonato de Etilo/análogos & derivados , Metanosulfonato de Etilo/farmacología , Humanos , Indicadores y Reactivos/farmacología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutagénesis Sitio-Dirigida/métodos , Estructura Terciaria de Proteína , Receptor de Angiotensina Tipo 1/química , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo
5.
J Biol Chem ; 284(39): 26603-12, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19635801

RESUMEN

The octapeptide hormone angiotensin II (AngII) binds to and activates the human angiotensin II type 1 receptor (hAT(1)) of the G protein-coupled receptor class A family. Several activation mechanisms have been proposed for this family, but they have not yet been experimentally validated. We previously used the methionine proximity assay to show that 11 residues in transmembrane domain (TMD) III, VI, and VII of the hAT(1) receptor reside in close proximity to the C-terminal residue of AngII. With the exception of a single change in TMD VI, the same contacts are present on N111G-hAT(1), a constitutively active mutant; this N111G-hAT(1) is a model for the active form of the receptor. In this study, two series of 53 individual methionine mutations were constructed in TMD I, II, IV, and V on both receptor forms. The mutants were photolabeled with a neutral antagonist, (125)I-[Sar(1),p-benzoyl-L-Phe(8)]AngII, and the resulting complexes were digested with cyanogen bromide. Although no new contacts were found for the hAT(1) mutants, two were found in the constitutively active mutants, Phe-77 in TMD II and Asn-200 in TMD V. To our knowledge, this is the first time that a direct ligand contact with TMD II and TMD V has been reported. These contact point differences were used to identify the structural changes between the WT-hAT(1) and N111G-hAT(1) complexes through homology-based modeling and restrained molecular dynamics. The model generated revealed an important structural rearrangement of several TMDs from the basal to the activated form in the WT-hAT(1) receptor.


Asunto(s)
Angiotensina II/metabolismo , Mutación , Receptor de Angiotensina Tipo 1/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Chlorocebus aethiops , Bromuro de Cianógeno/química , Electroforesis en Gel de Poliacrilamida , Humanos , Fosfatos de Inositol/metabolismo , Ligandos , Metionina/genética , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Etiquetas de Fotoafinidad , Unión Proteica , Receptor de Angiotensina Tipo 1/química , Receptor de Angiotensina Tipo 1/genética , Transfección
6.
J Biol Chem ; 284(46): 31953-61, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19773549

RESUMEN

The octapeptide hormone angiotensin II exerts a wide variety of cardiovascular effects through the activation of the angiotensin II Type 1 (AT(1)) receptor, which belongs to the G protein-coupled receptor superfamily. Like other G protein- coupled receptors, the AT(1) receptor possesses seven transmembrane domains that provide structural support for the formation of the ligand-binding pocket. The role of the fifth transmembrane domain (TMD5) was investigated using the substituted cysteine accessibility method. All of the residues within Thr-190 to Leu-217 region were mutated one at a time to cysteine, and after expression in COS-7 cells, the mutant receptors were treated with the sulfhydryl-specific alkylating agent methanethiosulfonate-ethylammonium (MTSEA). MTSEA reacts selectively with water-accessible, free sulfhydryl groups of endogenous or introduced point mutation cysteines. If a cysteine is found in the binding pocket, the covalent modification will affect the binding kinetics of the ligand. MTSEA substantially decreased the binding affinity of L197C-AT(1), N200C-AT(1), I201C-AT(1), G203C-AT(1), and F204C-AT(1) mutant receptors, which suggests that these residues orient themselves within the water-accessible binding pocket of the AT(1) receptor. Interestingly, this pattern of acquired MTSEA sensitivity was altered for TMD5 reporter cysteines engineered in a constitutively active N111G-AT(1) receptor background. Indeed, mutant I201C-N111G-AT(1) became more sensitive to MTSEA, whereas mutant G203C-N111G-AT(1) lost some sensitivity. Our results suggest that constitutive activation of AT(1) receptor causes an apparent counterclockwise rotation of TMD5 as viewed from the extracellular side.


Asunto(s)
Angiotensina II/farmacología , Receptor de Angiotensina Tipo 1/química , Receptor de Angiotensina Tipo 1/metabolismo , Vasoconstrictores/farmacología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Metanosulfonato de Etilo/análogos & derivados , Metanosulfonato de Etilo/farmacología , Humanos , Indicadores y Reactivos/farmacología , Cinética , Ligandos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Unión Proteica , Conformación Proteica , Receptor de Angiotensina Tipo 1/genética , Transfección , Fosfolipasas de Tipo C/metabolismo
7.
Biochem Pharmacol ; 168: 330-338, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31348898

RESUMEN

The signaling mechanisms of the angiotensin II type 2 receptor (AT2R), a heptahelical receptor, have not yet been clearly and completely defined. In the present contribution, we set out to identify the molecular determinants involved in AT2R activation. Although AT2R has not been shown to engage Gq/11, G12, Gi2, and ß-arrestin (ßarr) pathways as does the AT1R upon angiotensin II (AngII) stimulation, the atypical positioning of helix VIII in the recently published AT2R structure may play a role in the receptor's capacity to couple to downstream effectors. In the AT2R structure, helix VIII points inwards and towards intracellular loop 3 (ICL3) to form tertiary interactions with transmembrane domain 6 (TM6), possibly impeding access to signaling effectors. On the other hand, in most class A GPCRs, helix VIII is found to be engaged in tertiary interactions with ICL1 and away from the effector binding site. Upon closer examination of the AT2R structure, we found that the residues contained within intracellular loop 1 (ICL1) may be involved in driving this unusual conformation of helix VIII. To explore this hypothesis, we designed a series of AT1R/AT2R receptor chimeras to validate the roles of ICL1 and helix VIII in AT2R signaling. Substituting the AT1R ICL1 into AT2R led to a mutant receptor that coupled to Gi2. The substitution of the helix VIII and C-terminal domains of AT2R into the AT1R backbone led to a mutant receptor that retained AT1R-like signaling properties. These results suggest that the C-terminal portion of AT2R is compatible with canonical GPCR signaling and that ICL1 of AT2R is involved in repositioning helix VIII, which impedes engagement of classical GPCR effectors such as G proteins or ßarrs.


Asunto(s)
Secuencias Hélice-Asa-Hélice/fisiología , Membranas Intracelulares/química , Membranas Intracelulares/metabolismo , Receptor de Angiotensina Tipo 2/química , Receptor de Angiotensina Tipo 2/metabolismo , Angiotensina II/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Relación Dosis-Respuesta a Droga , Células HEK293 , Secuencias Hélice-Asa-Hélice/efectos de los fármacos , Humanos , Membranas Intracelulares/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor de Angiotensina Tipo 2/agonistas
8.
ACS Chem Neurosci ; 10(3): 1615-1626, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30614675

RESUMEN

Leu-enkephalin and d-Ala2-Leu-enkephalin were modified at their N- and C-termini with guanidyl and tetrazole groups. The resulting molecules were prepared in solution or by solid phase peptide synthesis. The affinity of the different analogues at mu (MOP) and delta opioid receptors (DOP) was then assessed by competitive binding in stably transfected DOP and MOP HEK293 cells. Inhibition of cAMP production and recruitment of ß-arrestin were also investigated. Finally, lipophilicity (logD7.4) and plasma stability of each compound were measured. Compared to the native ligands, we found that the replacement of the terminal carboxylate by a tetrazole slightly decreased both the affinity at mu and delta opioid receptors as well as the half-life. By contrast, replacing the ammonium at the N-terminus with a guanidyl significantly improved the affinity, the potency, as well as the lipophilicity and the stability of the resulting peptides. Replacing the glycine residue with a d-alanine in position 2 consistently improved the potency as well as the stability of the analogues. The best peptidomimetic of the whole series, guanidyl-Tyr-d-Ala-Gly-Phe-Leu-tetrazole, displayed sub-nanomolar affinity and an increased lipophilicity. Moreover, it proved to be stable in plasma for up to 24 h, suggesting that the modifications are protecting the compound against protease degradation.


Asunto(s)
Encefalina Leucina/análogos & derivados , Oligopéptidos/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Animales , Células HEK293 , Humanos , Péptidos Opioides/efectos de los fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo
9.
Sci Adv ; 5(11): eaax9115, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31807708

RESUMEN

Selective activation of the δ-opioid receptor (DOP) has great potential for the treatment of chronic pain, benefitting from ancillary anxiolytic and antidepressant-like effects. Moreover, DOP agonists show reduced adverse effects as compared to µ-opioid receptor (MOP) agonists that are in the spotlight of the current "opioid crisis." Here, we report the first crystal structures of the DOP in an activated state, in complex with two relevant and structurally diverse agonists: the potent opioid agonist peptide KGCHM07 and the small-molecule agonist DPI-287 at 2.8 and 3.3 Å resolution, respectively. Our study identifies key determinants for agonist recognition, receptor activation, and DOP selectivity, revealing crucial differences between both agonist scaffolds. Our findings provide the first investigation into atomic-scale agonist binding at the DOP, supported by site-directed mutagenesis and pharmacological characterization. These structures will underpin the future structure-based development of DOP agonists for an improved pain treatment with fewer adverse effects.


Asunto(s)
Simulación del Acoplamiento Molecular , Péptidos/química , Receptores Opioides delta/agonistas , Receptores Opioides delta/química , Animales , Cristalografía por Rayos X , Humanos , Dominios Proteicos , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Células Sf9 , Spodoptera
10.
Mol Pharmacol ; 74(3): 552-61, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18509066

RESUMEN

Class A (rhodopsin-like) G protein-coupled receptors possess conserved residues and motifs that are important for their specific activity. In the present study, we examined the role of residue Asp97(2.50) as well as residues Glu147(3.49), Arg148(3.50), and Tyr149(3.51) of the ERY motif on the functionality of the urotensin II receptor (UT). Mutations D97(2.50)A, R148(3.50)A, and R148(3.50)H abolished the ability of UT to activate phospholipase C, whereas mutations E147(3.49)A and Y149(3.51)A reduced the ability to activate PLC by 50%. None of the mutants exhibited constitutive activity. However, R148(3.50)A and R148(3.50)H promoted ERK1/2 activation, which was abolished by 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478), an inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase activity. Both these mutants were capable of directly activating EGFR, which confirmed that they activated the mitogen-activated protein kinase (MAPK) pathway by a Galpha(q/11)-independent transactivation of EGFR. The D97(2.50)A, R148(3.50)A, and R148(3.50)H mutants did not readily internalize and did not promote translocation or colocalize with beta-arrestin2-GFP. Finally, the agonist-induced internalization of the E147(3.49)A mutant receptor was significantly increased compared with wild-type receptor. This study highlights the major contribution of the conserved Asp(2.50) residue to the functionality of the UT receptor. The Arg residue in the ERY motif of UT is an important structural element in signaling crossroads that determine whether Galpha(q/11)-dependent and -independent events can occur.


Asunto(s)
Ácido Aspártico/metabolismo , Secuencia Conservada , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Arrestinas/metabolismo , Células COS , Chlorocebus aethiops , Análisis Mutacional de ADN , Activación Enzimática , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Proteína Quinasa C/metabolismo , Transporte de Proteínas , Ratas , Relación Estructura-Actividad , Activación Transcripcional , beta-Arrestinas
11.
Peptides ; 29(5): 691-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18155322

RESUMEN

The urotensin II receptor (UT) is a member of the G protein-coupled receptor (GPCR) family and binds the cyclic undecapeptide urotensin II (U-II) as well as the octapeptide urotensin II-related peptide (URP). The active UT mediates pleiotropic effects through various signal transduction pathways, including coupling to G proteins and activating the mitogen-activated protein kinase pathway. Several highly conserved residues and motifs of class A GPCRs that are important for activity are found in UT. This review highlights some of the putative roles of these motifs in the binding, activation and desensitization of UT.


Asunto(s)
Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/fisiología , Urotensinas/metabolismo
12.
Biochem J ; 402(1): 51-61, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17064254

RESUMEN

The mechanism by which GPCRs (G-protein-coupled receptors) undergo activation is believed to involve conformational changes following agonist binding. We have used photoaffinity labelling to identify domains within GPCRs that make contact with various photoreactive ligands in order to better understand the activation mechanism. Here, a series of four agonist {[Bpa1]U-II (Bpa is p-benzoyl-L-phenylalanine), [Bpa2]U-II, [Bpa3]U-II and [Bpa4]U-II} and three partial agonist {[Bpa1Pen5D-Trp7Orn8]U-II (Pen is penicillamine), [Bpa2Pen5D-Trp7Orn8]U-II and [Pen5Bpa6D-Trp7Orn8]U-II} photoreactive urotensin II (U-II) analogues were used to identify ligand-binding sites on the UT receptor (U-II receptor). All peptides bound the UT receptor expressed in COS-7 cells with high affinity (Kd of 0.3-17.7 nM). Proteolytic mapping and mutational analysis led to the identification of Met288 of the third extracellular loop of the UT receptor as a binding site for all four agonist peptides. Both partial agonists containing the photoreactive group in positions 1 and 2 also cross-linked to Met288. We found that photolabelling with the partial agonist containing the photoreactive group in position 6 led to the detection of transmembrane domain 5 as a binding site for that ligand. Interestingly, this differs from Met184/Met185 of the fourth transmembrane domain that had been identified previously as a contact site for the full agonist [Bpa6]U-II. These results enable us to better map the binding pocket of the UT receptor. Moreover, the data also suggest that, although structurally related agonists or partial agonists may dock in the same general binding pocket, conformational changes induced by various states of activation may result in slight differences in spatial proximity within the cyclic portion of U-II analogues.


Asunto(s)
Receptores Acoplados a Proteínas G/agonistas , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Humanos , Ligandos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Péptidos/farmacología , Etiquetas de Fotoafinidad , Unión Proteica , Conformación Proteica , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Serina Endopeptidasas/metabolismo , Urotensinas/agonistas , Urotensinas/química , Urotensinas/metabolismo
13.
Biochem Pharmacol ; 154: 104-117, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29684376

RESUMEN

G protein coupled receptors (GPCRs) produce pleiotropic effects by their capacity to engage numerous signaling pathways once activated. Functional selectivity (also called biased signaling), where specific compounds can bring GPCRs to adopt conformations that enable selective receptor coupling to distinct signaling pathways, continues to be significantly investigated. However, an important but often overlooked aspect of functional selectivity is the capability of ligands such as angiotensin II (AngII) to adopt specific conformations that may preferentially bind to selective GPCRs structures. Understanding both receptor and ligand conformation is of the utmost importance for the design of new drugs targeting GPCRs. In this study, we examined the properties of AngII cyclic analogs to impart biased agonism on the angiotensin type 1 receptor (AT1R). Positions 3 and 5 of AngII were substituted for cysteine and homocysteine residues ([Sar1Hcy3,5]AngII, [Sar1Cys3Hcy5]AngII and [Sar1Cys3,5]AngII) and the resulting analogs were evaluated for their capacity to activate the Gq/11, G12, Gi2, Gi3, Gz, ERK and ß-arrestin (ßarr) signaling pathways via AT1R. Interestingly, [Sar1Hcy3,5]AngII exhibited potency and full efficacy on all pathways tested with the exception of the Gq pathway. Molecular dynamic simulations showed that the energy barrier associated with the insertion of residue Phe8 of AngII within the hydrophobic core of AT1R, associated with Gq/11 activation, is increased with [Sar1Hcy3,5]AngII. These results suggest that constraining the movements of molecular determinants within a given ligand by introducing cyclic structures may lead to the generation of novel ligands providing more efficient biased agonism.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Angiotensina II/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Angiotensina II/química , Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/química , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Receptor de Angiotensina Tipo 1/química , Transducción de Señal/fisiología
14.
Sci Rep ; 8(1): 11415, 2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-30061705

RESUMEN

The renin-angiotensin system regulates blood pressure and fluid balance in the body primarily via angiotensin receptor 1 (AT1R). Renal AT1R was found to be primarily responsible for Ang II-mediated hypertension. G protein-coupled receptor kinase 2 (GRK2) modulates AT1R desensitization and increased GRK2 protein expression is reported in hypertensive patients. However, the consequences of GRK2 inhibition on kidney functions remain unknown. We employed shGRK2 knockdown mice (shGRK2 mice) to test the role of GRK2 in kidney development and function that can be ultimately linked to the hypertensive phenotype detected in shGRK2 mice. GRK2 knockdown reduced kidney size, nephrogenesis and glomerular count, and impaired glomerular filtration. Glomerular damage in adult shGRK2 mice was associated with increased renin- and AT1R-mediated production of reactive oxygen species. The AT1R blocker, Losartan, normalized elevated blood pressure and markedly improved glomerular filtration in the shGRK2 knockdown mice. Our findings provide evidence for the crucial role of GRK2 in renal regulation of blood pressure. It also suggests that the detrimental outcomes of GRK2 inhibitors on the kidney should be carefully examined when used as antihypertensive.


Asunto(s)
Presión Sanguínea/fisiología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Técnicas de Silenciamiento del Gen , Riñón/lesiones , Riñón/fisiopatología , Animales , Presión Sanguínea/efectos de los fármacos , Quinasa 2 del Receptor Acoplado a Proteína-G/deficiencia , Tasa de Filtración Glomerular , Riñón/efectos de los fármacos , Riñón/patología , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Losartán/farmacología , Ratones Endogámicos C57BL , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Renina/sangre , Suero/metabolismo
15.
Org Lett ; 19(8): 2018-2021, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28368122

RESUMEN

Given the putative selectivity of the antagonist TIPP (Tyr-Tic-Phe-Phe) for δ-opioid receptors (DOP), this compound was selected for the design of a novel 64Cu-radiolabeled potent and selective DOP positron emission tomography (PET) imaging agent. Ex vivo autoradiography of TIPPD-PEG-K(NOTA/64Cu)-NH2 on rat brain sections produced a distribution pattern consistent with the known expression of DOP. Taken together, the in vitro and ex vivo data indicate that this 64Cu-tracer holds promise for studying the DOP by means of PET.

16.
J Med Chem ; 49(7): 2200-9, 2006 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16570916

RESUMEN

A stereospecific convergent synthesis of N-[(9-fluorenyl)methoxycarbonyl]-p-[3-(trifluoromethyl)-3H-diazirin-3-yl]-l-phenylalanine (Fmoc-12, Fmoc-Tdf) and its incorporation into the C-terminal position of the angiotensin II (AngII) peptide to form (125)I[Sar(1),Tdf(8)]AngII ((125)I-13) is presented. This amino acid photoprobe is a highly reactive carbene-generating diazirine phenylalanine derivative that can be used for photoaffinity labeling. Using model receptors, we compared the reactivity and the Met selectivity of 12 to that of the widely used and reputedly Met-selective p-benzoyl-l-phenylalanine (Bpa) photoprobe. Wild-type and mutant AngII type 2 receptors, a G protein-coupled receptors, were photolabeled with (125)I-13 as well as with (125)I[Sar(1),Bpa(8)]AngII ((125)I-14), and the respective incorporation yields were assessed. The carbene-generating 12 was more reactive toward inert residues and was not Met-selective compared to the biradical ketone-generating Bpa, allowing for more precise determination of ligand contact points in peptidergic receptors.


Asunto(s)
Angiotensina II/análogos & derivados , Angiotensina II/síntesis química , Diazometano/análogos & derivados , Metano/análogos & derivados , Metionina/metabolismo , Fenilalanina/análogos & derivados , Etiquetas de Fotoafinidad/síntesis química , Receptor de Angiotensina Tipo 2/metabolismo , Angiotensina II/química , Angiotensina II/metabolismo , Animales , Células COS , Chlorocebus aethiops , Diazometano/síntesis química , Diazometano/química , Humanos , Hidrocarburos/química , Radioisótopos de Yodo , Marcaje Isotópico , Metano/química , Modelos Moleculares , Mutación , Fenilalanina/síntesis química , Fenilalanina/química , Etiquetas de Fotoafinidad/química , Etiquetas de Fotoafinidad/metabolismo , Ensayo de Unión Radioligante , Receptor de Angiotensina Tipo 2/genética , Estereoisomerismo , Relación Estructura-Actividad
17.
J Leukoc Biol ; 73(2): 243-52, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12554801

RESUMEN

Collagen type IV (CnIV) and fibronectin (Fn) were used as ligands to study the distribution of alpha(2)beta(1) and alpha(4)beta(1) integrins in low-density, detergent-resistant microdomains (DRM) of Jurkat lymphocytes. CnIV-coated microspheres induced (optical trapping) the redistribution of GM(1)-associated fluorescence from the cell periphery to the area of contact. This was not observed in cells treated with beta-methyl cyclodextrin (MCD). Fn- or bovine serum albumin-coated microspheres did not modify the peripheral distribution of fluorescence. These observations were confirmed by confocal microscopy. Western blot analysis of cells exposed to surfaces coated with CnIV revealed that the alpha(2)-subunit was initially present at low levels in DRM, became strongly associated after 40 min, and returned to basal levels after 75 min. Fn induced a slight recruitment of the beta(1)-integrin alpha(4)-subunit in DRM after 5 and 10 min, followed by a return to basal levels. Neither CnIV nor Fn triggered significant changes in the distribution of the beta(1)-subunit in DRM. Fn- and CnIV-coated microspheres or surfaces coated with these ligands triggered a MCD-sensitive mobilization of Ca(2)(+). MCD did not alter the state of the Ca(2)(+) reserves. The differential distributions of the alpha(2)beta(1) and alpha(4)beta(1) integrins in DRM may provide one additional step in the regulation of outside-in signaling involving these integrins.


Asunto(s)
Colágeno Tipo IV/metabolismo , Fibronectinas/metabolismo , Integrina alfa2beta1/metabolismo , Integrina alfa4beta1/metabolismo , Microdominios de Membrana/metabolismo , Calcio/metabolismo , Fluorescencia , Gangliósido G(M1)/metabolismo , Humanos , Células Jurkat , Microesferas , Factores de Tiempo
18.
Biochem Pharmacol ; 92(2): 280-8, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25175740

RESUMEN

The vasoactive urotensin-II (UII), a cyclic undecapeptide widely distributed in cardiovascular, renal and endocrine systems, specifically binds the UII receptor (UT receptor), a G protein-coupled receptor (GPCR). The involvement of this receptor in numerous pathophysiological conditions including atherosclerosis, heart failure, hypertension, renal impairment and diabetes potentially makes it an interesting therapeutic target. To elucidate how UII binds the UT receptor through the identification of specific residues in transmembrane domains (TM) one (TM1) and two (TM2) that are involved in the formation of the receptor's binding pocket, we used the substituted-cysteine accessibility method (SCAM). Each residue of TM1 (V49((1.30)) to M76((1.57))) and TM2 (V88((2.41)) to H117((2.70))) was mutated, one by one, to a cysteine. The resulting mutants were then expressed in COS-7 cells and subsequently treated with the sulfhydryl-specific alkylating agent methanethiosulfonate-ethylammonium (MTSEA). MTSEA treatment resulted in a significant binding inhibition of (125)I-UII to mutant I54C((1.35)) in TM1 and mutants Y100C((2.53)), S103C((2.56)), F106C((2.59)), I107C((2.60)), T110C((2.63)) and Y111C((2.64)) in TM2. These results identify key structural residues in TM1 and TM2 that participate in the formation of the UT receptor binding pocket. Together with previous SCAM analysis of TM3, TM4, TM5, TM6 and TM7, these results have led us to identify residues within all 7 TMs that participate in UT's binding pocket and have enabled us to propose a model of this receptor's orthosteric binding site.


Asunto(s)
Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Células COS , Chlorocebus aethiops , Datos de Secuencia Molecular , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína/fisiología , Ratas
19.
Biochem Pharmacol ; 86(11): 1584-93, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24084430

RESUMEN

Urotensin-II (UII), a cyclic undecapeptide, selectively binds the urotensin-II receptor (UT receptor), a G protein-coupled receptor (GPCR) involved in cardiovascular effects and associated with numerous pathophysiological conditions including hypertension, atherosclerosis, heart failure, pulmonary hypertension and others. In order to identify specific residues in transmembrane domains (TM) three (TM3), four (TM4) and five (TM5) that are involved in the formation of the UT receptor binding pocket, we used the substituted-cysteine accessibility method (SCAM). Each residue in the F118((3.20)) to S146((3.48)) fragment of TM3, the L168((4.44)) to G194((4.70)) fragment of TM4 and the W203((5.30)) to V232((5.59)) fragment of TM5, was mutated, individually, to a cysteine. The resulting mutants were then expressed in COS-7 cells and subsequently treated with the positively charged sulfhydryl-specific alkylating agent methanethiosulfonate-ethylammonium (MTSEA). MTSEA treatment resulted in a significant reduction in the binding of (125)I-UII to TM3 mutants L126C((3.28)), F127C((3.29)), F131C((3.33)) and M134C((3.36)) and TM4 mutants M184C((4.60)) and I188C((4.64)). No loss of binding was detected following treatment by MTSEA for all TM5 mutants tested. In absence of a crystal structure of UT receptor, these results identify key determinants in TM3, TM4 and TM5 that participate in the formation of the UT receptor binding pocket and has led us to propose a homology model of the UT receptor.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Urotensinas/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células COS , Técnicas de Cultivo de Célula , Chlorocebus aethiops , Cisteína/genética , Metanosulfonato de Etilo/análogos & derivados , Metanosulfonato de Etilo/farmacología , Ligandos , Modelos Moleculares , Mutación , Ratas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Transfección
20.
Biochem Pharmacol ; 77(8): 1374-82, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19426676

RESUMEN

Urotensin II (U-II), a cyclic undecapeptide, is the natural ligand of the urotensin II (UT) receptor, a G protein-coupled receptor. In the present study, we used the substituted-cysteine accessibility method to identify specific residues in transmembrane domains (TMDs) six and seven of the rat urotensin II receptor (rUT) that contribute to the formation of the binding pocket of the receptor. Each residue in the R256(6.32)-Q283(6.59) fragment of TMD6 and the A295(7.31)-T321(7.57) fragment of TMD7 was mutated, individually, to a cysteine. The resulting mutants were expressed in COS-7 cells, which were subsequently treated with the positively charged methanethiosulfonate-ethylammonium (MTSEA) or the negatively charged methanethiosulfonate-ethylsulfonate (MTSES) sulfhydryl-specific alkylating agents. MTSEA treatment resulted in a significant reduction in the binding of TMD6 mutants F268C(6.44) and W278C(6.54) and TMD7 mutants L298C(7.34), T302C(7.38), and T303C(7.39) to (125)I-U-II. MTSES treatment resulted in a significant reduction in the binding of two additional mutants, namely L282C(6.58) in TMD6 and Y300C(7.36) in TMD7. These results suggest that specific residues orient themselves within the water-accessible binding pocket of the rUT receptor. This approach, which allowed us to identify key determinants in TMD6 and TMD7 that contribute to the UT receptor binding pocket, enabled us to further refine our homology-based model of how U-II interacts with its cognate receptor.


Asunto(s)
Cisteína/metabolismo , Mutación , Receptores Acoplados a Proteínas G/metabolismo , Alquilantes/farmacología , Animales , Células COS , Chlorocebus aethiops , Cisteína/genética , Metanosulfonato de Etilo/análogos & derivados , Metanosulfonato de Etilo/farmacología , Ligandos , Mesilatos/farmacología , Modelos Moleculares , Reacción en Cadena de la Polimerasa , Unión Proteica , Estructura Terciaria de Proteína , Ensayo de Unión Radioligante , Ratas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Transfección , Urotensinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA