Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 161(4): 833-44, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25913193

ABSTRACT

Angiotensin II type 1 receptor (AT(1)R) is a G protein-coupled receptor that serves as a primary regulator for blood pressure maintenance. Although several anti-hypertensive drugs have been developed as AT(1)R blockers (ARBs), the structural basis for AT(1)R ligand-binding and regulation has remained elusive, mostly due to the difficulties of growing high-quality crystals for structure determination using synchrotron radiation. By applying the recently developed method of serial femtosecond crystallography at an X-ray free-electron laser, we successfully determined the room-temperature crystal structure of the human AT(1)R in complex with its selective antagonist ZD7155 at 2.9-Å resolution. The AT(1)R-ZD7155 complex structure revealed key structural features of AT(1)R and critical interactions for ZD7155 binding. Docking simulations of the clinically used ARBs into the AT(1)R structure further elucidated both the common and distinct binding modes for these anti-hypertensive drugs. Our results thereby provide fundamental insights into AT(1)R structure-function relationship and structure-based drug design.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Receptor, Angiotensin, Type 1/chemistry , Amino Acid Sequence , Angiotensin II Type 1 Receptor Blockers/chemistry , Crystallography, X-Ray , Humans , Molecular Sequence Data , Mutagenesis , Naphthyridines/chemistry , Naphthyridines/pharmacology , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Sequence Alignment
2.
J Chem Inf Model ; 59(1): 373-385, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30608150

ABSTRACT

We present a succession of structural changes involved in hormone peptide activation of a prototypical GPCR. Microsecond molecular dynamics simulation generated conformational ensembles reveal propagation of structural changes through key "microswitches" within human AT1R bound to native hormone. The endocrine octa-peptide angiotensin II (AngII) activates AT1R signaling in our bodies which maintains physiological blood pressure, electrolyte balance, and cardiovascular homeostasis. Excessive AT1R activation is associated with pathogenesis of hypertension and cardiovascular diseases which are treated by sartan drugs. The mechanism of AT1R inhibition by sartans has been elucidated by 2.8 Å X-ray structures, mutagenesis, and computational analyses. Yet, the mechanism of AT1R activation by AngII is unclear. The current study delineates an activation scheme initiated by AngII binding. A van der Waals "grasp" interaction between Phe8AngII with Ile2887.39 in AT1R induced mechanical strain pulling Tyr2927.43 and breakage of critical interhelical H-bonds, first between Tyr2927.43 and Val1083.32 and second between Asn1113.35 and Asn2957.46. Subsequently changes are observed in conserved microswitches DRYTM3, Yx7K(R)TM5, CWxPTM6, and NPxxYTM7 in AT1R. Activating the microswitches in the intracellular region of AT1R may trigger formation of the G-protein binding pocket as well as exposure of helix-8 to cytoplasm. Thus, the active-like conformation of AT1R is initiated by the van der Waals interaction of Phe8AngII with Ile2887.39, followed by systematic reorganization of critical interhelical H-bonds and activation of microswitches.


Subject(s)
Angiotensin II/pharmacology , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/metabolism , Entropy , Humans , Models, Molecular , Protein Conformation
3.
J Chem Inf Model ; 58(1): 182-193, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29195045

ABSTRACT

Crystal structures of the human angiotensin II type 1 receptor (AT1R) complex with the antihypertensive agent ZD7155 (PDB id: 4YAY ) and the blood pressure medication Benicar (PDB id: 4ZUD ) showed that binding poses of both antagonists are similar. This finding implies that clinically used angiotensin receptor blocking (ARB) drugs may interact in a similar fashion. However, clinically observed differences in pharmacological and therapeutic efficacies of ARBs lead to the question of whether the dynamic interactions of AT1R with ARBs vary. To address this, we performed induced-fit docking (IFD) of eight clinically used ARBs to AT1R followed by 200 ns molecular dynamic (MD) simulation. The experimental Ki values for ARBs correlated remarkably well with calculated free energy with R2 = 0.95 and 0.70 for AT1R-ARB models generated respectively by IFD and MD simulation. The eight ARB-AT1R complexes share a common set of binding residues. In addition, MD simulation results validated by mutagenesis data discovered distinctive spatiotemporal interactions that display unique bonding between an individual ARB and AT1R. These findings provide a reasonably broader picture reconciling the structure-based observations with clinical studies reporting efficacy variations for ARBs. The unique differences unraveled for ARBs in this study will be useful for structure-based design of the next generation of more potent and selective ARBs.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemistry , Drug Design , Receptor, Angiotensin, Type 1/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Crystallography, X-Ray , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/genetics , Spatio-Temporal Analysis
4.
Pharmacol Rev ; 67(4): 754-819, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26315714

ABSTRACT

The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors­the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor­and a type II trans-membrane zinc protein­the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.


Subject(s)
GTP-Binding Proteins/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Renin-Angiotensin System/physiology , Animals , Cardiovascular Diseases/physiopathology , Cell Proliferation , Endothelium/physiopathology , Humans , Inflammation/physiopathology , Kidney Diseases/physiopathology , Mice , Nervous System Diseases/physiopathology , Polymorphism, Genetic , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Angiotensin/metabolism , Signal Transduction/physiology , Structure-Activity Relationship
5.
Pharmacol Res ; 123: 40-50, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28648738

ABSTRACT

Although the octapeptide hormone angiotensin II (Ang II) regulates cardiovascular and renal homeostasis through the Ang II type 1 receptor (AT1R), overstimulation of AT1R causes various human diseases, such as hypertension and cardiac hypertrophy. Therefore, AT1R blockers (ARBs) have been widely used as therapeutic drugs for these diseases. Recent basic research and clinical studies have resulted in the discovery of interesting phenomena associated with AT1R function. For example, ligand-independent activation of AT1R by mechanical stress and agonistic autoantibodies, as well as via receptor mutations, has been shown to decrease the inverse agonistic efficacy of ARBs, though the molecular mechanisms of such phenomena had remained elusive until recently. Furthermore, although AT1R is believed to exist as a monomer, recent studies have demonstrated that AT1R can homodimerize and heterodimerize with other G-protein coupled receptors (GPCR), altering the receptor signaling properties. Therefore, formation of both AT1R homodimers and AT1R-GPCR heterodimer may be involved in the pathogenesis of human disease states, such as atherosclerosis and preeclampsia. Finally, biased AT1R ligands that can preferentially activate the ß-arrestin-mediated signaling pathway have been discovered. Such ß-arrestin-biased AT1R ligands may be better therapeutic drugs for cardiovascular diseases. New findings on AT1R described herein could provide a conceptual framework for application of ARBs in the treatment of diseases, as well as for novel drug development. Since AT1R is an extensively studied member of the GPCR superfamily encoded in the human genome, this review is relevant for understanding the functions of other members of this superfamily.


Subject(s)
Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Drug Inverse Agonism , Humans , Protein Conformation , Protein Multimerization , Receptor, Angiotensin, Type 1/chemistry
6.
J Biol Chem ; 290(49): 29127-39, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26420482

ABSTRACT

Angiotensin II type 1 receptor (AT1R) is the primary blood pressure regulator. AT1R blockers (ARBs) have been widely used in clinical settings as anti-hypertensive drugs and share a similar chemical scaffold, although even minor variations can lead to distinct therapeutic efficacies toward cardiovascular etiologies. The structural basis for AT1R modulation by different peptide and non-peptide ligands has remained elusive. Here, we report the crystal structure of the human AT1R in complex with an inverse agonist olmesartan (Benicar(TM)), a highly potent anti-hypertensive drug. Olmesartan is anchored to the receptor primarily by the residues Tyr-35(1.39), Trp-84(2.60), and Arg-167(ECL2), similar to the antagonist ZD7155, corroborating a common binding mode of different ARBs. Using docking simulations and site-directed mutagenesis, we identified specific interactions between AT1R and different ARBs, including olmesartan derivatives with inverse agonist, neutral antagonist, or agonist activities. We further observed that the mutation N111(3.35)A in the putative sodium-binding site affects binding of the endogenous peptide agonist angiotensin II but not the ß-arrestin-biased peptide TRV120027.


Subject(s)
Antihypertensive Agents/chemistry , Imidazoles/chemistry , Receptor, Angiotensin, Type 1/chemistry , Tetrazoles/chemistry , Allosteric Site , Animals , Binding, Competitive , COS Cells , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Computer Simulation , Crystallography, X-Ray , Humans , Ions , Ligands , Mutagenesis, Site-Directed , Mutation , Oligopeptides/chemistry , Protein Binding , Protein Structure, Tertiary , Sf9 Cells , Sodium/chemistry
7.
Mol Pharmacol ; 88(3): 488-501, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26121982

ABSTRACT

Ligand-independent signaling by the angiotensin II type 1 receptor (AT1R) can be activated in clinical settings by mechanical stretch and autoantibodies as well as receptor mutations. Transition of the AT1R to the activated state is known to lower inverse agonistic efficacy of clinically used AT1R blockers (ARBs). The structure-function basis for reduced efficacy of inverse agonists is a fundamental aspect that has been understudied not only in relation to the AT1R but also regarding other homologous receptors. Here, we demonstrate that the active-state transition in the AT1R indeed attenuates an inverse agonistic effect of four biphenyl-tetrazole ARBs through changes in specific ligand-receptor interactions. In the ground state, tight interactions of four ARBs with a set of residues (Ser109(TM3), Phe182(ECL2), Gln257(TM6), Tyr292(TM7), and Asn295(TM7)) results in potent inverse agonism. In the activated state, the ARB-AT1R interactions shift to a different set of residues (Val108(TM3), Ser109(TM3), Ala163(TM4), Phe182(ECL2), Lys199(TM5), Tyr292(TM7), and Asn295(TM7)), resulting in attenuated inverse agonism. Interestingly, V108I, A163T, N295A, and F182A mutations in the activated state of the AT1R shift the functional response to the ARB binding toward agonism, but in the ground state the same mutations cause inverse agonism. Our data show that the second extracellular loop is an important regulator of the functional states of the AT1R. Our findings suggest that the quest for discovering novel ARBs, and improving current ARBs, fundamentally depends on the knowledge of the unique sets of residues that mediate inverse agonistic potency in the two states of the AT1R.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Drug Inverse Agonism , Receptor, Angiotensin, Type 1/chemistry , Amino Acid Sequence , Angiotensin II Type 1 Receptor Blockers/chemistry , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Molecular Sequence Data , Mutation, Missense , Protein Binding , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism
8.
J Biol Chem ; 289(44): 30763-30771, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25217633

ABSTRACT

STAT3 is a pleiotropic transcription factor that is activated by the phosphorylation of tyrosine 705 in response to many cytokines and growth factors. STAT3 without Tyr-705 phosphorylation (U-STAT3) is also a potent transcription factor, and its concentration in cells increases greatly in response to STAT3 activation because the STAT3 gene can be driven by phosphorylated STAT3 dimers. We have now searched for post-translational modifications of U-STAT3 that might have a critical role in its function. An analysis by mass spectroscopy indicated that U-STAT3 is acetylated on Lys-685, and the integrity of Lys-685 is required for the expression of most U-STAT3-dependent genes. In contrast, we found only a very minor role for Lys-685 in gene expression induced in response to tyrosine-phosphorylated STAT3. U-STAT3 plays an important role in angiotensin II-induced gene expression and in the consequent development of cardiac hypertrophy and dysfunction. Mutation of Lys-685 inhibits this function of STAT3, providing new information on the role of U-STAT3 in augmenting the development of heart failure.


Subject(s)
Lysine/genetics , Protein Processing, Post-Translational , STAT3 Transcription Factor/physiology , Acetylation , Angiotensin II/physiology , Cardiomegaly/metabolism , Cell Line , Humans , Interleukin-6/physiology , Mutation, Missense , Phosphorylation , Transcriptional Activation
9.
J Mol Cell Cardiol ; 75: 25-39, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24976017

ABSTRACT

Improper regulation of signaling in vascular smooth muscle cells (VSMCs) by angiotensin II (AngII) can lead to hypertension, vascular hypertrophy and atherosclerosis. The extent to which the homeostatic levels of the components of signaling networks are regulated through microRNAs (miRNA) modulated by AngII type 1 receptor (AT1R) in VSMCs is not fully understood. Whether AT1R blockers used to treat vascular disorders modulate expression of miRNAs is also not known. To report differential miRNA expression following AT1R activation by AngII, we performed microarray analysis in 23 biological and technical replicates derived from humans, rats and mice. Profiling data revealed a robust regulation of miRNA expression by AngII through AT1R, but not the AngII type 2 receptor (AT2R). The AT1R-specific blockers, losartan and candesartan antagonized >90% of AT1R-regulated miRNAs and AngII-activated AT2R did not modulate their expression. We discovered VSMC-specific modulation of 22 miRNAs by AngII, and validated AT1R-mediated regulation of 17 of those miRNAs by real-time polymerase chain reaction analysis. We selected miR-483-3p as a novel representative candidate for further study because mRNAs of multiple components of the renin-angiotensin system (RAS) were predicted to contain the target sequence for this miRNA. MiR-483-3p inhibited the expression of luciferase reporters bearing 3'-UTRs of four different RAS genes and the inhibition was reversed by antagomir-483-3p. The AT1R-regulated expression levels of angiotensinogen and angiotensin converting enzyme 1 (ACE-1) proteins in VSMCs are modulated specifically by miR-483-3p. Our study demonstrates that the AT1R-regulated miRNA expression fingerprint is conserved in VSMCs of humans and rodents. Furthermore, we identify the AT1R-regulated miR-483-3p as a potential negative regulator of steady-state levels of RAS components in VSMCs. Thus, miRNA-regulation by AngII to affect cellular signaling is a novel aspect of RAS biology, which may lead to discovery of potential candidate prognostic markers and therapeutic targets.


Subject(s)
Angiotensin II/pharmacology , Gene Expression Regulation/drug effects , MicroRNAs/genetics , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/genetics , Animals , Base Sequence , Gene Expression Profiling , Genome/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Organ Specificity/drug effects , Organ Specificity/genetics , Rats , Receptor, Angiotensin, Type 1/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
10.
J Biol Chem ; 288(1): 540-51, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23139413

ABSTRACT

The topology of the second extracellular loop (ECL2) and its interaction with ligands is unique in each G protein-coupled receptor. When the orthosteric ligand pocket located in the transmembrane (TM) domain is occupied, ligand-specific conformational changes occur in the ECL2. In more than 90% of G protein-coupled receptors, ECL2 is tethered to the third TM helix via a disulfide bond. Therefore, understanding the extent to which the TM domain and ECL2 conformations are coupled is useful. To investigate this, we examined conformational changes in ECL2 of the angiotensin II type 1 receptor (AT1R) by introducing mutations in distant sites that alter the activation state equilibrium of the AT1R. Differential accessibility of reporter cysteines introduced at four conformation-sensitive sites in ECL2 of these mutants was measured. Binding of the agonist angiotensin II (AngII) and inverse agonist losartan in wild-type AT1R changed the accessibility of reporter cysteines, and the pattern was consistent with ligand-specific "lid" conformations of ECL2. Without agonist stimulation, the ECL2 in the gain of function mutant N111G assumed a lid conformation similar to AngII-bound wild-type AT1R. In the presence of inverse agonists, the conformation of ECL2 in the N111G mutant was similar to the inactive state of wild-type AT1R. In contrast, AngII did not induce a lid conformation in ECL2 in the loss of function D281A mutant, which is consistent with the reduced AngII binding affinity in this mutant. However, a lid conformation was induced by [Sar(1),Gln(2),Ile(8)] AngII, a specific analog that binds to the D281A mutant with better affinity than AngII. These results provide evidence for the emerging paradigm of domain coupling facilitated by long range interactions at distant sites on the same receptor.


Subject(s)
Mutation , Receptor, Angiotensin, Type 1/genetics , Animals , Biotin/chemistry , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Cysteine/genetics , Ligands , Molecular Conformation , Molecular Dynamics Simulation , Mutagenesis , Protein Conformation , Protein Structure, Tertiary , Rats , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/chemistry , Signal Transduction
11.
J Biol Chem ; 285(21): 16341-50, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20299456

ABSTRACT

The orientation of the second extracellular loop (ECL2) is divergent in G-protein coupled receptor (GPCR) structures determined. This discovery provoked the question, is the ECL2 conformation differentially regulated in the GPCRs that respond to diffusible ligands? We have determined the conformation of the ECL2 of the angiotensin II type 1 receptor by reporter-cysteine accessibility mapping in different receptor states (i.e. empty, agonist-bound and antagonist-bound). We introduced cysteines at each position of ECL2 of an N-terminal epitope-tagged receptor surrogate lacking all non-essential cysteines and then measured reaction of these with a cysteine-reactive biotin probe. The ability of biotinylated mutant receptors to react with a steptavidin-HRP-conjugated antibody was used as the basis for examining differences in accessibility. Two segments of ECL2 were accessible in the empty receptor, indicating an open conformation of ECL2. These segments were inaccessible in the ligand-bound states of the receptor. Using the accessibility constraint, we performed molecular dynamics simulation to predict ECL2 conformation in different states of the receptor. Analysis suggested that a lid conformation similar to that of ECL2 in rhodopsin was induced upon binding both agonist and antagonist, but exposing different accessible segments delimited by the highly conserved disulfide bond. Our study reveals the ability of ECL2 to interact with diffusing ligands and to adopt a ligand-specific lid conformation, thus, slowing down dissociation of ligands when bound. Distinct conformations induced by the bound agonist and the antagonist around the conserved disulfide bond suggest an important role for this disulfide bond in producing different functional states of the receptor.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemistry , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/chemistry , Angiotensin II Type 1 Receptor Blockers/metabolism , Animals , Biotinylation , COS Cells , Chlorocebus aethiops , Disulfides , Ligands , Mutation, Missense , Peptide Mapping , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Rabbits , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Rhodopsin/chemistry , Rhodopsin/genetics , Rhodopsin/metabolism , Streptavidin/chemistry , Structural Homology, Protein
12.
Methods Cell Biol ; 149: 19-30, 2019.
Article in English | MEDLINE | ID: mdl-30616819

ABSTRACT

G protein-coupled receptors (GPCRs) play an active role in numerous cellular processes, from cell proliferation to differentiation, by modulating gene transcription through various signal transduction pathways. Transcriptional regulation coupled to reporter gene expression may be used to study both G protein-dependent and G protein-independent responses activated by GPCR ligands. Reporter genes are typically used to monitor changes in receptor-mediated cellular responses at the transcription/translation level. Genetic reporter assays are based on reporter gene expression in response to activation of specific signaling cascade, followed by monitoring the presence of the reporter protein by directly measuring its enzymatic activity. These optimized genes are expressed under the control of a response element to assess its transcriptional activity that can be readily detected by a luminescent signal. Firefly luciferase gene has been widely used as a genetic reporter that responds rapidly to modulation of a GPCR by agonists or antagonists. Luciferase assays have been successfully developed for deorphanization of GPCRs, high-throughput screening (HTS) applications for drug discovery and deciphering both canonical and non-canonical signaling of numerous GPCRs. The protocol outlined for STAT3-driven luciferase assay could be adapted with appropriate changes to any aspect of GPCR signaling.


Subject(s)
Biological Assay/methods , Genes, Reporter , Luciferases/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , CHO Cells , Cricetinae , Cricetulus , Genetic Vectors/metabolism , HEK293 Cells , Humans , Ligands , Rats
13.
Methods Cell Biol ; 149: 215-238, 2019.
Article in English | MEDLINE | ID: mdl-30616822

ABSTRACT

Maintenance of normal blood pressure under conditions of drug treatment is a measure of system-wide neuro-hormonal controls and electrolyte/fluid volume homeostasis in the body. With increased interest in designing and evaluating novel drugs that may functionally select or allosterically modulate specific GPCR signaling pathways, techniques that allow us to measure acute and long-term effects on blood pressure are very important. Therefore, this chapter describes techniques to measure acute and long-term impact of novel GPCR ligands on blood pressure regulation. We will use the angiotensin type 1 receptor, a powerful blood pressure regulating GPCR, in detailing the methodology. Normal blood pressure maintenance depends upon dynamic modulation of angiotensin type 1 receptor activity by the hormone peptide angiotensin II. Chronic activation of angiotensin type 1 receptor creates hypertension and related cardiovascular disease states which are treated with angiotensin type 1 receptor blockers (ARBs). Thus, a prototype for evaluation of blood pressure control under experimental evaluation of novel drugs.


Subject(s)
Blood Pressure/physiology , Blood Vessels/physiology , Homeostasis , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Ligands , Mice , Receptor, Angiotensin, Type 1/metabolism
14.
Br J Pharmacol ; 175(12): 2454-2469, 2018 06.
Article in English | MEDLINE | ID: mdl-29570771

ABSTRACT

BACKGROUND AND PURPOSE: Conditions such as hypertension and renal allograft rejection are accompanied by chronic, agonist-independent, signalling by angiotensin II AT1 receptors. The current treatment paradigm for these diseases entails the preferred use of inverse agonist AT1 receptor blockers (ARBs). However, variability in the inverse agonist activities of common biphenyl-tetrazole ARBs for the active state of AT1 receptors often leads to treatment failure. Therefore, characterization of robust inverse agonist ARBs for the active state of AT1 receptors is necessary. EXPERIMENTAL APPROACH: To identify the robust inverse agonist for active state of AT1 receptors and its molecular mechanism, we performed site-directed mutagenesis, competition binding assay, inositol phosphate production assay and molecular modelling for both ground-state wild-type AT1 receptors and active-state N111G mutant AT1 receptors. KEY RESULTS: Although candesartan and telmisartan exhibited weaker inverse agonist activity for N111G- compared with WT-AT1 receptors, only eprosartan exhibited robust inverse agonist activity for both N111G- and WT- AT1 receptors. Specific ligand-receptor contacts for candesartan and telmisartan are altered in the active-state N111G- AT1 receptors compared with the ground-state WT-AT1 receptors, suggesting an explanation of their attenuated inverse agonist activity for the active state of AT1 receptors. In contrast, interactions between eprosartan and N111G-AT1 receptors were not significantly altered, and the inverse agonist activity of eprosartan was robust. CONCLUSIONS AND IMPLICATIONS: Eprosartan may be a better therapeutic option than other ARBs. Comparative studies investigating eprosartan and other ARBs for the treatment of diseases caused by chronic, agonist-independent, AT1 receptor activation are warranted.


Subject(s)
Acrylates/pharmacology , Imidazoles/pharmacology , Receptor, Angiotensin, Type 1/agonists , Thiophenes/pharmacology , Acrylates/chemistry , Animals , Catalytic Domain/drug effects , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Ligands , Models, Molecular , Molecular Structure , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Structure-Activity Relationship , Thiophenes/chemistry
16.
Br J Pharmacol ; 174(9): 737-753, 2017 05.
Article in English | MEDLINE | ID: mdl-28194766

ABSTRACT

Angiotensins are a group of hormonal peptides and include angiotensin II and angiotensin 1-7 produced by the renin angiotensin system. The biology, pharmacology and biochemistry of the receptors for angiotensins were extensively reviewed recently. In the review, the receptor nomenclature committee was not emphatic on designating MAS1 as the angiotensin 1-7 receptor on the basis of lack of classical G protein signalling and desensitization in response to angiotensin 1-7, as well as a lack of consensus on confirmatory ligand pharmacological analyses. A review of recent publications (2013-2016) on the rapidly progressing research on angiotensin 1-7 revealed that MAS1 and two additional receptors can function as 'angiotensin 1-7 receptors', and this deserves further consideration. In this review we have summarized the information on angiotensin 1-7 receptors and their crosstalk with classical angiotensin II receptors in the context of the functions of the renin angiotensin system. It was concluded that the receptors for angiotensin II and angiotensin 1-7 make up a sophisticated cross-regulated signalling network that modulates the endogenous protective and pathogenic facets of the renin angiotensin system.


Subject(s)
Angiotensin I/metabolism , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System/physiology , Animals , Humans , Protein Binding/physiology , Proto-Oncogene Mas , Signal Transduction/physiology
18.
Adv Pharmacol ; 70: 155-74, 2014.
Article in English | MEDLINE | ID: mdl-24931196

ABSTRACT

The pathophysiological actions of the renin-angiotensin system hormone, angiotensin II (AngII), are mainly mediated by the AngII type 1 (AT1) receptor, a GPCR. The intrinsic spontaneous activity of the AT1 receptor in native tissues is difficult to detect due to its low expression levels. However, factors such as the membrane environment, interaction with autoantibodies, and mechanical stretch are known to increase G protein signaling in the absence of AngII. Naturally occurring and disease-causing activating mutations have not been identified in AT1 receptor. Constitutively active mutants (CAMs) of AT1 receptor have been engineered using molecular modeling and site-directed mutagenesis approaches among which substitution of Asn(111) in the transmembrane helix III with glycine or serine results in the highest basal activity of the receptor. Transgenic animal models expressing the CAM AT1 receptors that mimic various in vivo disease conditions have been useful research tools for discovering the pathophysiological role of AT1 receptor and evaluating the therapeutic potential of inverse agonists. This chapter summarizes the studies on the constitutive activity of AT1 receptor in recombinant as well as physiological systems. The impact of the availability of CAM AT1 receptors on our understanding of the molecular mechanisms underlying receptor activation and inverse agonism is described.


Subject(s)
Drug Discovery , Mutation/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Animals , Biomedical Research , Drug Inverse Agonism , Humans , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/chemistry
19.
PLoS One ; 8(1): e52689, 2013.
Article in English | MEDLINE | ID: mdl-23326349

ABSTRACT

Heterotrimeric G-protein signal transduction initiated by G-protein-coupled receptors (GPCRs) in the plasma membrane is thought to propagate through protein-protein interactions of subunits, Gα and Gßγ in the cytosol. In this study, we show novel nuclear functions of Gßγ through demonstrating interaction of Gß(2) with integral components of chromatin and effects of Gß(2) depletion on global gene expression. Agonist activation of several GPCRs including the angiotensin II type 1 receptor specifically augmented Gß(2) levels in the nucleus and Gß(2) interacted with specific nucleosome core histones and transcriptional modulators. Depletion of Gß(2) repressed the basal and angiotensin II-dependent transcriptional activities of myocyte enhancer factor 2. Gß(2) interacted with a sequence motif that was present in several transcription factors, whose genome-wide binding accounted for the Gß(2)-dependent regulation of approximately 2% genes. These findings suggest a wide-ranging mechanism by which direct interaction of Gßγ with specific chromatin bound transcription factors regulates functional gene networks in response to GPCR activation in cells.


Subject(s)
Chromatin/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Angiotensin II/pharmacology , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chromatin/genetics , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Immunoblotting , MEF2 Transcription Factors , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Protein Binding , RNA Interference , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/genetics , Sequence Homology, Amino Acid
20.
Trends Pharmacol Sci ; 33(2): 79-88, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22037017

ABSTRACT

Recent solved structures of G protein-coupled receptors (GPCRs) provide insights into variation of the structure and molecular mechanisms of GPCR activation. In this review, we provide evidence for the emerging paradigm of domain coupling facilitated by intrinsic disorder of the ligand-free state in GPCRs. The structure-function and dynamic studies suggest that ligand-bound GPCRs exhibit multiple active conformations in initiating cellular signals. Long-range intramolecular and intermolecular interactions at distant sites on the same receptor are crucial factors that modulate signaling function of GPCRs. Positive or negative coupling between the extracellular, the transmembrane and the intracellular domains facilitates cooperativity of activating 'switches' as requirements for the functional plasticity of GPCRs. Awareness that allosteric ligands robustly affect domain coupling provides a novel mechanistic basis for rational drug development, small molecule antagonism and GPCR regulation by classical as well as nonclassical modes.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Ligands , Protein Binding , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL