scholarly journals Insights into the Interaction of LVV-Hemorphin-7 with Angiotensin II Type 1 Receptor

2020 ◽  
Vol 22 (1) ◽  
pp. 209
Author(s):  
Amanat Ali ◽  
Elizabeth K. M. Johnstone ◽  
Bincy Baby ◽  
Heng B. See ◽  
Angela Song ◽  
...  

Hemorphins are known for their role in the control of blood pressure. Recently, we revealed the positive modulation of the angiotensin II (AngII) type 1 receptor (AT1R) by LVV-hemorphin-7 (LVV-H7) in human embryonic kidney (HEK293) cells. Here, we examined the molecular binding behavior of LVV-H7 on AT1R and its effect on AngII binding using a nanoluciferase-based bioluminescence resonance energy transfer (NanoBRET) assay in HEK293FT cells, as well as molecular docking and molecular dynamics (MD) studies. Saturation and real-time kinetics supported the positive effect of LVV-H7 on the binding of AngII. While the competitive antagonist olmesartan competed with AngII binding, LVV-H7 slightly, but significantly, decreased AngII’s kD by 2.6 fold with no effect on its Bmax. Molecular docking and MD simulations indicated that the binding of LVV-H7 in the intracellular region of AT1R allosterically potentiates AngII binding. LVV-H7 targets residues on intracellular loops 2 and 3 of AT1R, which are known binding sites of allosteric modulators in other GPCRs. Our data demonstrate the allosteric effect of LVV-H7 on AngII binding, which is consistent with the positive modulation of AT1R activity and signaling previously reported. This further supports the pharmacological targeting of AT1R by hemorphins, with implications in vascular and renal physiology.

2021 ◽  
Vol 22 (3) ◽  
pp. 1082
Author(s):  
Elizabeth K. M. Johnstone ◽  
Heng B. See ◽  
Rekhati S. Abhayawardana ◽  
Angela Song ◽  
K. Johan Rosengren ◽  
...  

Receptor heteromerization is the formation of a complex involving at least two different receptors with pharmacology that is distinct from that exhibited by its constituent receptor units. Detection of these complexes and monitoring their pharmacology is crucial for understanding how receptors function. The Receptor-Heteromer Investigation Technology (Receptor-HIT) utilizes ligand-dependent modulation of interactions between receptors and specific biomolecules for the detection and profiling of heteromer complexes. Previously, the interacting biomolecules used in Receptor-HIT assays have been intracellular proteins, however in this study we have for the first time used bioluminescence resonance energy transfer (BRET) with fluorescently-labeled ligands to investigate heteromerization of receptors on the cell surface. Using the Receptor-HIT ligand binding assay with NanoBRET, we have successfully investigated heteromers between the angiotensin II type 1 (AT1) receptor and the β2 adrenergic receptor (AT1-β2AR heteromer), as well as between the AT1 and angiotensin II type 2 receptor (AT1-AT2 heteromer).


2017 ◽  
Vol 292 (8) ◽  
pp. 3341-3350 ◽  
Author(s):  
Brent M. Young ◽  
Elaine Nguyen ◽  
Matthew A. J. Chedrawe ◽  
Jan K. Rainey ◽  
Denis J. Dupré

G protein-coupled receptors (GPCRs) play an important role in drug therapy and represent one of the largest families of drug targets. The angiotensin II type 1 receptor (AT1R) is notable as it has a central role in the treatment of cardiovascular disease. Blockade of AT1R signaling has been shown to alleviate hypertension and improve outcomes in patients with heart failure. Despite this, it has become apparent that our initial understanding of AT1R signaling is oversimplified. There is considerable evidence to suggest that AT1R signaling is highly modified in the presence of receptor-receptor interactions, but there is very little structural data available to explain this phenomenon even with the recent elucidation of the AT1R crystal structure. The current study investigates the involvement of transmembrane domains in AT1R homomer assembly with the goal of identifying hydrophobic interfaces that contribute to receptor-receptor affinity. A recently published crystal structure of the AT1R was used to guide site-directed mutagenesis of outward-facing hydrophobic residues within the transmembrane region of the AT1R. Bioluminescence resonance energy transfer was employed to analyze how receptor mutation affects the assembly of AT1R homomers with a specific focus on hydrophobic residues. Mutations within transmembrane domains IV, V, VI, and VII had no effect on angiotensin-mediated β-arrestin1 recruitment; however, they exhibited differential effects on the assembly of AT1R into oligomeric complexes. Our results demonstrate the importance of hydrophobic amino acids at the AT1R transmembrane interface and provide the first glimpse of the requirements for AT1R complex assembly.


2018 ◽  
Vol 11 (559) ◽  
pp. eaat1631 ◽  
Author(s):  
Yoon Namkung ◽  
Christian LeGouill ◽  
Sahil Kumar ◽  
Yubo Cao ◽  
Larissa B. Teixeira ◽  
...  

G protein–coupled receptors (GPCRs) are important therapeutic targets that exhibit functional selectivity (biased signaling), in which different ligands or receptor variants elicit distinct downstream signaling. Understanding all the signaling events and biases that contribute to both the beneficial and adverse effects of GPCR stimulation by given ligands is important for drug discovery. Here, we report the design, validation, and use of pathway-selective bioluminescence resonance energy transfer (BRET) biosensors that monitor the engagement and activation of signaling effectors downstream of G proteins, including protein kinase C (PKC), phospholipase C (PLC), p63RhoGEF, and Rho. Combined with G protein and β-arrestin BRET biosensors, our sensors enabled real-time monitoring of GPCR signaling at different levels in downstream pathways in both native and engineered cells. Profiling of the responses to 14 angiotensin II (AngII) type 1 receptor (AT1R) ligands enabled the clustering of compounds into different subfamilies of biased ligands and showed that, in addition to the previously reported functional selectivity between Gαq and β-arrestin, there are also biases among G protein subtypes. We also demonstrated that biases observed at the receptor and G protein levels propagated to downstream signaling pathways and that these biases could occur through the engagement of different G proteins to activate a common effector. We also used these tools to determine how naturally occurring AT1R variants affected signaling bias. This suite of BRET biosensors provides a useful resource for fingerprinting biased ligands and mutant receptors and for dissecting functional selectivity at various levels of GPCR signaling.


2020 ◽  
Vol 295 (38) ◽  
pp. 13169-13180
Author(s):  
Yubo Cao ◽  
Sahil Kumar ◽  
Yoon Namkung ◽  
Laurence Gagnon ◽  
Aaron Cho ◽  
...  

The angiotensin II (AngII) type 1 receptor (AT1R), a member of the G protein–coupled receptor (GPCR) family, signals through G proteins and β-arrestins, which act as adaptors to regulate AT1R internalization and mitogen-activated protein kinase (MAPK) ERK1/2 activation. β-arrestin–dependent ERK1/2 regulation is the subject of important studies because its spatiotemporal control remains poorly understood for many GPCRs, including AT1R. To study the link between β-arrestin–dependent trafficking and ERK1/2 signaling, we investigated three naturally occurring AT1R variants that show distinct receptor–β-arrestin interactions: A163T, T282M, and C289W. Using bioluminescence resonance energy transfer (BRET)–based and conformational fluorescein arsenical hairpin–BRET sensors coupled with high-resolution fluorescence microscopy, we show that all AT1R variants form complexes with β-arrestin2 at the plasma membrane and efficiently internalize into endosomes upon AngII stimulation. However, mutant receptors imposed distinct conformations in β-arrestin2 and differentially impacted endosomal trafficking and MAPK signaling. Notably, T282M accumulated in endosomes, but its ability to form stable complexes following internalization was reduced, markedly impairing its ability to co-traffic with β-arrestin2. We also found that despite β-arrestin2 overexpression, T282M's and C289W's residency with β-arrestin2 in endosomes was greatly reduced, leading to decreased β-arrestin–dependent ERK1/2 activation, faster recycling of receptors to the plasma membrane, and impaired AngII-mediated proliferation. Our findings reveal that naturally occurring AT1R variants alter the patterns of receptor/β-arrestin2 trafficking and suggest conformationally dependent β-arrestin–mediated MAPK activation as well as endosomal receptor–β-arrestin complex stabilization in the mitogenic response of AT1R.


2018 ◽  
Vol 115 (47) ◽  
pp. 12051-12056 ◽  
Author(s):  
Akil A. Puckerin ◽  
Donald D. Chang ◽  
Zunaira Shuja ◽  
Papiya Choudhury ◽  
Joachim Scholz ◽  
...  

Genetically encoded inhibitors for voltage-dependent Ca2+ (CaV) channels (GECCIs) are useful research tools and potential therapeutics. Rad/Rem/Rem2/Gem (RGK) proteins are Ras-like G proteins that potently inhibit high voltage-activated (HVA) Ca2+ (CaV1/CaV2 family) channels, but their nonselectivity limits their potential applications. We hypothesized that nonselectivity of RGK inhibition derives from their binding to auxiliary CaVβ-subunits. To investigate latent CaVβ-independent components of inhibition, we coexpressed each RGK individually with CaV1 (CaV1.2/CaV1.3) or CaV2 (CaV2.1/CaV2.2) channels reconstituted in HEK293 cells with either wild-type (WT) β2a or a mutant version (β2a,TM) that does not bind RGKs. All four RGKs strongly inhibited CaV1/CaV2 channels reconstituted with WT β2a. By contrast, when channels were reconstituted with β2a,TM, Rem inhibited only CaV1.2, Rad selectively inhibited CaV1.2 and CaV2.2, while Gem and Rem2 were ineffective. We generated mutant RGKs (Rem[R200A/L227A] and Rad[R208A/L235A]) unable to bind WT CaVβ, as confirmed by fluorescence resonance energy transfer. Rem[R200A/L227A] selectively blocked reconstituted CaV1.2 while Rad[R208A/L235A] inhibited CaV1.2/CaV2.2 but not CaV1.3/CaV2.1. Rem[R200A/L227A] and Rad[R208A/L235A] both suppressed endogenous CaV1.2 channels in ventricular cardiomyocytes and selectively blocked 25 and 62%, respectively, of HVA currents in somatosensory neurons of the dorsal root ganglion, corresponding to their distinctive selectivity for CaV1.2 and CaV1.2/CaV2.2 channels. Thus, we have exploited latent β-binding–independent Rem and Rad inhibition of specific CaV1/CaV2 channels to develop selective GECCIs with properties unmatched by current small-molecule CaV channel blockers.


2013 ◽  
Vol 51 (1) ◽  
pp. 191-202 ◽  
Author(s):  
Patricia M Lenhart ◽  
Stefan Broselid ◽  
Cordelia J Barrick ◽  
L M Fredrik Leeb-Lundberg ◽  
Kathleen M Caron

Receptor activity-modifying protein 3 (RAMP3) is a single-pass transmembrane protein known to interact with and affect the trafficking of several G-protein-coupled receptors (GPCRs). We sought to determine whether RAMP3 interacts with GPR30, also known as G-protein-coupled estrogen receptor 1. GPR30 is a GPCR that binds estradiol and has important roles in cardiovascular and endocrine physiology. Using bioluminescence resonance energy transfer titration studies, co-immunoprecipitation, and confocal microscopy, we show that GPR30 and RAMP3 interact. Furthermore, the presence of GPR30 leads to increased expression of RAMP3 at the plasma membrane in HEK293 cells. In vivo, there are marked sex differences in the subcellular localization of GPR30 in cardiac cells, and the hearts of Ramp3−/− mice also show signs of GPR30 mislocalization. To determine whether this interaction might play a role in cardiovascular disease, we treated Ramp3+/+ and Ramp3−/− mice on a heart disease-prone genetic background with G-1, a specific agonist for GPR30. Importantly, this in vivo activation of GPR30 resulted in a significant reduction in cardiac hypertrophy and perivascular fibrosis that is both RAMP3 and sex dependent. Our results demonstrate that GPR30–RAMP3 interaction has functional consequences on the localization of these proteins both in vitro and in vivo and that RAMP3 is required for GPR30-mediated cardioprotection.


2008 ◽  
Vol 86 (8) ◽  
pp. 526-535 ◽  
Author(s):  
Nathan J. Evans ◽  
Jeffery W. Walker

G protein-coupled receptors (GPCRs), including endothelin receptor A (ETA) and B (ETB), may form dimers or higher-order oligomers that profoundly influence signaling. Here we examined a PDZ finger motif within the C-terminus of ETA and its role in heterodimerization with ETB, and in homodimerization with itself, when expressed in HEK293 cells. Receptor dimerization was monitored by (i) fluorescence resonance energy transfer (FRET) between cyan fluorescent protein (CFP) (FRET donor) and tetracysteine/FlAsH (FRET acceptor) fused to the C-termini of ET receptors, and (ii) coimmunoprecipitation of ET receptors after mild detergent solubilization. Mutations in a PDZ finger motif at threonine403/serine404 eliminated FRET and reduced coimmunoprecipitation of heterodimers and homodimers. Functional consequences were evaluated by measuring mobilization of intracellular Ca2+ and internalization of receptors in response to a 10 nmol/L ET-1 challenge. PDZ mutations converted a sustained Ca2+ signal mediated by ETA:ETB heterodimers into a transient response, similar to that observed for homodimers or monomers. Heterodimers containing PDZ mutations were seen to internalize in a similar time domain (approximately 5 min) to the transient Ca2+ elevation and with similar kinetics to internalization of ETA homodimers or monomers. Without the PDZ mutations, heterodimers did not internalize over 15 min, suggesting the intriguing possibility that sustained Ca2+ signaling was a consequence (at least in part) of delayed internalization. The results are consistent with structural models of ETA-receptor dimerization that place threonine403/serine404 of the PDZ finger motif at the interaction interface between heterodimers and homodimers. Sustained Ca2+ signaling and delayed endocytosis of ETA:ETB heterodimers argues strongly for a unique dimer interface that impacts transmembrane signaling and internalization.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2028-2028
Author(s):  
Abdelouahid El Khattouti ◽  
Volker R. Stoldt ◽  
Rüdiger E. Scharf

Abstract Abstract 2028 Background and Objectives: The HPA-1 polymorphism of αIIbβ3 arises from a Leu→Pro exchange at residue 33 of the β3 subunit resulting in HPA-1a (Leu33) or HPA-1b (Pro33). We have documented that patients with coronary artery disease who are carriers of HPA-1b (Pro33) experience their myocardial infarction 5.2 years earlier than HPA-1a/1a (Leu33) patients (J Thromb Haemost 2005; 3: 1522). Based on these observations, it has been postulated that HPA-1b (Pro33) is a prothrombotic variant of αIIbβ3. We have now generated a model overexpressing fluorescent proteins fused with αIIbβ3 in transfected HEK293 cells. Methods: :A yellow protein (YFP) and a cyan fluorescent protein (CFP) were cloned to the C-termini of the β3 and αIIb subunits prior to transfection of HEK293 cells, subsequently expressing the fusion proteins of both HPA-1 isoforms. Using flow cytometry, Western blotting and specific antibodies directed against αIIb or β3, we identified 12 HPA-1a and 11 HPA-1b positive clones. For further experiments only those cell lines expressing equal amounts of fluorescent fusion proteins, i.e. a 140 kD αIIb-CFP and a 113 kD β3-YFP, were used. Results: Functional integrity of both integrin variants and proper membrane insertion were documented by intact activation of transfected HEK293 cells through G protein-coupled receptors with organic acid (1-stearoyl-2-arachidonoyl-sn-glycerol) or direct phorbol 12-myristate 13-acetate-induced stimulation of protein kinase C and by specific binding of Alexa488 fibrinogen to αIIbβ3 in response to inside-out signaling. In the presencence of pertussis toxin or abciximab, activation or ligand binding of αIIbβ3 were completely (>98%) inhibited in both isoforms. Activation of αIIbβ3 stimulates the tyrosine kinase Src, constitutively associated with the the β-subunit of the integrin. To determine whether αIIbβ3-dependent outside-in signaling is responsible for a polymorphism-related modulation, we performed adhesion experiments under static conditions with fibrinogen (50 μg/ml) in the absence or presence of Mn2+ (0.5 mM). Specific activation of the phosphotyrosine motif (Src-pY418), as determined by Western blotting and quantified by densitometry (ratio of Src-pY418/total Src), was 15 + 1.5% higher in HPA-1b than HPA-1a cells in the presence of Mn2+ (n=6 independent experiments, p<0.01). To explore the molecular nature of this difference in terms of putative changes in the allostery of integrin αIIbβ3 with regard to the HPA-1 polymorphism, dynamic measurements were performed using fluorescence resonance energy transfer (FRET). The relative decrease in FRET signal, indicating spatial separation of the cytoplasmic tails of the α- and β-subunit as a consequence of integrin activation, was recorded every minute over 0.5 hrs in transfected HEK293 cells adherent onto fibrinogen. At every time point, the kinetic measurements revealed a significantly faster and more distict (> 5%) decrease in HPA-1b than in HPA-1a cells under static adhesion (p<0.009). Upon exposure of adherent HEK293 cells to increasing shear rates (stepwise elevation from 50 to 1600 sec-1 by doubling the initial shear rate every minute), the spatial separation of the integrin subunits occurred significantly faster and more distinct (> 10%) in HPA-1b (Pro33) than HPA-1a (Leu33) cells in response to shear (p<0.0014). Under the same conditions, the rate of HPA-1b cells still adherent onto immobilized fibrinogen was 80%, while the relative number of residual HPA-1a cells decreased to 20% upon exposure to 1600 sec-1 (p<0.0001). These displacement experiments suggest that the HPA-1b (Pro33) variant is more resistant to biomechanical stress than the HPA-1a (Leu33) isoform. Conclusions: Our findings suggest that the HPA-1 polymorphism can have a significant impact on the activation of αIIbβ3. This is evident from a higher outside-in signaling and a higher resistance to biomechanical stress upon exposure to increasing shear of HPA-1b (Pro33) in comparison with HPA-1a (Leu33) transfectants. The difference in spatial separation of the cytoplasmic tails of the integrin in response to activation, as demonstrated by FRET analyses under static and flow dynamic conditions, reflects allosteric changes that may contribute to the prothrombotic phenotype of the HPA-1b (Pro33) variant. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document