Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Sci Signal ; 16(797): eadf2173, 2023 08 08.
Article En | MEDLINE | ID: mdl-37552769

G protein-coupled receptors engage both G proteins and ß-arrestins, and their coupling can be biased by ligands and mutations. Here, to resolve structural elements and mechanisms underlying effector coupling to the angiotensin II (AngII) type 1 receptor (AT1R), we combined alanine scanning mutagenesis of the entire sequence of the receptor with pharmacological profiling of Gαq and ß-arrestin engagement to mutant receptors and molecular dynamics simulations. We showed that Gαq coupling to AT1R involved a large number of residues spread across the receptor, whereas fewer structural regions of the receptor contributed to ß-arrestin coupling regulation. Residue stretches in transmembrane domain 4 conferred ß-arrestin bias and represented an important structural element in AT1R for functional selectivity. Furthermore, we identified allosteric small-molecule binding sites that were enclosed by communities of residues that produced biased signaling when mutated. Last, we showed that allosteric communication within AT1R emanating from the Gαq coupling site spread beyond the orthosteric AngII-binding site and across different regions of the receptor, including currently unresolved structural regions. Our findings reveal structural elements and mechanisms within AT1R that bias Gαq and ß-arrestin coupling and that could be harnessed to design biased receptors for research purposes and to develop allosteric modulators.


Receptor, Angiotensin, Type 1 , Signal Transduction , beta-Arrestins/genetics , beta-Arrestins/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , beta-Arrestin 1/metabolism , GTP-Binding Proteins/metabolism , Angiotensin II/metabolism
2.
Mol Psychiatry ; 27(5): 2425-2438, 2022 05.
Article En | MEDLINE | ID: mdl-35393556

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.


Attention Deficit Disorder with Hyperactivity , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Actins , Adult , Attention Deficit Disorder with Hyperactivity/genetics , Child , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Humans , Ligands , Receptors, G-Protein-Coupled/genetics , Virulence
3.
Int J Mol Sci ; 23(5)2022 Feb 24.
Article En | MEDLINE | ID: mdl-35269644

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.


Bioluminescence Resonance Energy Transfer Techniques , Biosensing Techniques , Animals , Bioluminescence Resonance Energy Transfer Techniques/methods , Biosensing Techniques/methods , HEK293 Cells , Humans , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
4.
Sci Adv ; 5(11): eaax9115, 2019 11.
Article En | MEDLINE | ID: mdl-31807708

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.


Molecular Docking Simulation , Peptides/chemistry , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/chemistry , Animals , Crystallography, X-Ray , Humans , Protein Domains , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Sf9 Cells , Spodoptera
5.
Biochem Pharmacol ; 168: 330-338, 2019 10.
Article En | MEDLINE | ID: mdl-31348898

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.


Helix-Loop-Helix Motifs/physiology , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Receptor, Angiotensin, Type 2/chemistry , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Dose-Response Relationship, Drug , HEK293 Cells , Helix-Loop-Helix Motifs/drug effects , Humans , Intracellular Membranes/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Angiotensin, Type 2/agonists
6.
ACS Chem Neurosci ; 10(3): 1615-1626, 2019 03 20.
Article En | MEDLINE | ID: mdl-30614675

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.


Enkephalin, Leucine/analogs & derivatives , Oligopeptides/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Animals , HEK293 Cells , Humans , Opioid Peptides/drug effects , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism
8.
Sci Rep ; 8(1): 11415, 2018 07 30.
Article En | MEDLINE | ID: mdl-30061705

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.


Blood Pressure/physiology , G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Knockdown Techniques , Kidney/injuries , Kidney/physiopathology , Animals , Blood Pressure/drug effects , G-Protein-Coupled Receptor Kinase 2/deficiency , Glomerular Filtration Rate , Kidney/drug effects , Kidney/pathology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Losartan/pharmacology , Mice, Inbred C57BL , Phenotype , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism , Renin/blood , Serum/metabolism
9.
Biochem Pharmacol ; 154: 104-117, 2018 08.
Article En | MEDLINE | ID: mdl-29684376

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.


Angiotensin II Type 1 Receptor Blockers/metabolism , Angiotensin II/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Angiotensin II/chemistry , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Receptor, Angiotensin, Type 1/chemistry , Signal Transduction/physiology
10.
Proc Natl Acad Sci U S A ; 114(51): 13477-13482, 2017 12 19.
Article En | MEDLINE | ID: mdl-29192023

The Gαs subunit is classically involved in the signal transduction of G protein-coupled receptors (GPCRs) at the plasma membrane. Recent evidence has revealed noncanonical roles for Gαs in endosomal sorting of receptors to lysosomes. However, the mechanism of action of Gαs in this sorting step is still poorly characterized. Here, we report that Gαs interacts with ubiquitin to regulate the endosomal sorting of receptors for lysosomal degradation. We reveal that the N-terminal extremity of Gαs contains a ubiquitin-interacting motif (UIM), a sorting element usually found in the endosomal sorting complex required for transport (ESCRT) machinery responsible for sorting ubiquitinated receptors into intraluminal vesicles (ILVs) of multivesicular bodies (MVBs). Mutation of the UIM in Gαs confirmed the importance of ubiquitin interaction for the sorting of epidermal growth factor receptor (EGFR) into ILVs for lysosomal degradation. These findings demonstrate a role for Gαs as an integral component of the ubiquitin-dependent endosomal sorting machinery and highlight the dual role of Gαs in receptor trafficking and signaling for the fine-tuning of the cellular response.


Endosomes/metabolism , ErbB Receptors/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Ubiquitin/metabolism , Binding Sites , Endosomal Sorting Complexes Required for Transport/metabolism , GTP-Binding Protein alpha Subunits, Gs/chemistry , HEK293 Cells , HeLa Cells , Humans , Protein Binding , Protein Sorting Signals , Protein Transport
11.
Org Lett ; 19(8): 2018-2021, 2017 04 21.
Article En | MEDLINE | ID: mdl-28368122

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.

12.
J Biol Chem ; 290(25): 15835-15854, 2015 Jun 19.
Article En | MEDLINE | ID: mdl-25934394

Biased signaling represents the ability of G protein-coupled receptors to engage distinct pathways with various efficacies depending on the ligand used or on mutations in the receptor. The angiotensin-II type 1 (AT1) receptor, a prototypical class A G protein-coupled receptor, can activate various effectors upon stimulation with the endogenous ligand angiotensin-II (AngII), including the Gq/11 protein and ß-arrestins. It is believed that the activation of those two pathways can be associated with distinct conformations of the AT1 receptor. To verify this hypothesis, microseconds of molecular dynamics simulations were computed to explore the conformational landscape sampled by the WT-AT1 receptor, the N111G-AT1 receptor (constitutively active and biased for the Gq/11 pathway), and the D74N-AT1 receptor (biased for the ß-arrestin1 and -2 pathways) in their apo-forms and in complex with AngII. The molecular dynamics simulations of the AngII-WT-AT1, N111G-AT1, and AngII-N111G-AT1 receptors revealed specific structural rearrangements compared with the initial and ground state of the receptor. Simulations of the D74N-AT1 receptor revealed that the mutation stabilizes the receptor in the initial ground state. The presence of AngII further stabilized the ground state of the D74N-AT1 receptor. The biased agonist [Sar(1),Ile(8)]AngII also showed a preference for the ground state of the WT-AT1 receptor compared with AngII. These results suggest that activation of the Gq/11 pathway is associated with a specific conformational transition stabilized by the agonist, whereas the activation of the ß-arrestin pathway is linked to the stabilization of the ground state of the receptor.


Arrestins , GTP-Binding Protein alpha Subunits, Gq-G11 , Molecular Dynamics Simulation , Receptor, Angiotensin, Type 1 , Signal Transduction/physiology , Amino Acid Substitution , Arrestins/chemistry , Arrestins/genetics , Arrestins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Mutation, Missense , Protein Binding , Protein Structure, Quaternary , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , beta-Arrestins
13.
Mol Pharmacol ; 87(6): 982-95, 2015 Jun.
Article En | MEDLINE | ID: mdl-25808928

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.


Angiotensin II/metabolism , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/chemistry , Arrestins/metabolism , Enzyme Activation , ErbB Receptors/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Isoenzymes/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/metabolism , Protein Kinase C/metabolism , Receptor, Angiotensin, Type 1/chemistry , Signal Transduction , beta-Arrestins
14.
Biochem Pharmacol ; 92(2): 280-8, 2014 Nov 15.
Article En | MEDLINE | ID: mdl-25175740

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.


Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Binding Sites/physiology , COS Cells , Chlorocebus aethiops , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary/physiology , Rats
15.
Biochem Pharmacol ; 86(11): 1584-93, 2013 Dec 01.
Article En | MEDLINE | ID: mdl-24084430

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.


Receptors, G-Protein-Coupled/metabolism , Urotensins/metabolism , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Cell Culture Techniques , Chlorocebus aethiops , Cysteine/genetics , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Ligands , Models, Molecular , Mutation , Rats , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Transfection
16.
J Biol Chem ; 288(4): 2593-604, 2013 Jan 25.
Article En | MEDLINE | ID: mdl-23223579

G protein-coupled receptors contain selectively important residues that play central roles in the conformational changes that occur during receptor activation. Asparagine 111 (N111(3.35)) is such a residue within the angiotensin II type 1 (AT(1)) receptor. Substitution of N111(3.35) for glycine leads to a constitutively active receptor, whereas substitution for tryptophan leads to an inactivable receptor. Here, we analyzed the AT(1) receptor and two mutants (N111G and N111W) by molecular dynamics simulations, which revealed a novel molecular switch involving the strictly conserved residue D74(2.50). Indeed, D74(2.50) forms a stable hydrogen bond (H-bond) with the residue in position 111(3.35) in the wild-type and the inactivable receptor. However, in the constitutively active mutant N111G-AT(1) receptor, residue D74 is reoriented to form a new H-bond with another strictly conserved residue, N46(1.50). When expressed in HEK293 cells, the mutant N46G-AT(1) receptor was poorly activable, although it retained a high binding affinity. Interestingly, the mutant N46G/N111G-AT(1) receptor was also inactivable. Molecular dynamics simulations also revealed the presence of a cluster of hydrophobic residues from transmembrane domains 2, 3, and 7 that appears to stabilize the inactive form of the receptor. Whereas this hydrophobic cluster and the H-bond between D74(2.50) and W111(3.35) are more stable in the inactivable N111W-AT(1) receptor, the mutant N111W/F77A-AT(1) receptor, designed to weaken the hydrophobic core, showed significant agonist-induced signaling. These results support the potential for the formation of an H-bond between residues D74(2.50) and N46(1.50) in the activation of the AT(1) receptor.


Mutation , Receptor, Angiotensin, Type 1/chemistry , Computer Simulation , Conserved Sequence , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydrogen Bonding , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Angiotensin, Type 1/metabolism , Receptors, CXCR4/metabolism , Receptors, G-Protein-Coupled , Receptors, Opioid, kappa/metabolism , Structure-Activity Relationship
17.
J Biol Chem ; 285(4): 2284-93, 2010 Jan 22.
Article En | MEDLINE | ID: mdl-19940150

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.


Angiotensin II/metabolism , Membrane Proteins , Receptor, Angiotensin, Type 1 , Animals , Arginine/genetics , Arginine/metabolism , Binding Sites , COS Cells , Chlorocebus aethiops , Cysteine/genetics , Cysteine/metabolism , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Humans , Indicators and Reagents/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutagenesis, Site-Directed/methods , Protein Structure, Tertiary , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism
18.
J Biol Chem ; 284(46): 31953-61, 2009 Nov 13.
Article En | MEDLINE | ID: mdl-19773549

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.


Angiotensin II/pharmacology , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/metabolism , Vasoconstrictor Agents/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Humans , Indicators and Reagents/pharmacology , Kinetics , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Protein Binding , Protein Conformation , Receptor, Angiotensin, Type 1/genetics , Transfection , Type C Phospholipases/metabolism
19.
J Biol Chem ; 284(39): 26603-12, 2009 Sep 25.
Article En | MEDLINE | ID: mdl-19635801

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.


Angiotensin II/metabolism , Mutation , Receptor, Angiotensin, Type 1/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Cyanogen Bromide/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Inositol Phosphates/metabolism , Ligands , Methionine/genetics , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Photoaffinity Labels , Protein Binding , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/genetics , Transfection
20.
Biochem Pharmacol ; 77(8): 1374-82, 2009 Apr 15.
Article En | MEDLINE | ID: mdl-19426676

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.


Cysteine/metabolism , Mutation , Receptors, G-Protein-Coupled/metabolism , Alkylating Agents/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Cysteine/genetics , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Ligands , Mesylates/pharmacology , Models, Molecular , Polymerase Chain Reaction , Protein Binding , Protein Structure, Tertiary , Radioligand Assay , Rats , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Transfection , Urotensins/pharmacology
...