Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Sci Signal ; 17(842): eadi0934, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38917219

ABSTRACT

The stabilization of different active conformations of G protein-coupled receptors is thought to underlie the varying efficacies of biased and balanced agonists. Here, profiling the activation of signal transducers by angiotensin II type 1 receptor (AT1R) agonists revealed that the extent and kinetics of ß-arrestin binding exhibited substantial ligand-dependent differences, which were lost when receptor internalization was inhibited. When AT1R endocytosis was prevented, even weak partial agonists of the ß-arrestin pathway acted as full or near-full agonists, suggesting that receptor conformation did not exclusively determine ß-arrestin recruitment. The ligand-dependent variance in ß-arrestin translocation was much larger at endosomes than at the plasma membrane, showing that ligand efficacy in the ß-arrestin pathway was spatiotemporally determined. Experimental investigations and mathematical modeling demonstrated how multiple factors concurrently shaped the effects of agonists on endosomal receptor-ß-arrestin binding and thus determined the extent of functional selectivity. Ligand dissociation rate and G protein activity had particularly strong, internalization-dependent effects on the receptor-ß-arrestin interaction. We also showed that endocytosis regulated the agonist efficacies of two other receptors with sustained ß-arrestin binding: the V2 vasopressin receptor and a mutant ß2-adrenergic receptor. In the absence of endocytosis, the agonist-dependent variance in ß-arrestin2 binding was markedly diminished. Our results suggest that endocytosis determines the spatiotemporal bias in GPCR signaling and can aid in the development of more efficacious, functionally selective compounds.


Subject(s)
Endocytosis , Receptor, Angiotensin, Type 1 , Signal Transduction , beta-Arrestins , Endocytosis/physiology , Humans , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , beta-Arrestins/metabolism , beta-Arrestins/genetics , HEK293 Cells , Receptors, Vasopressin/metabolism , Receptors, Vasopressin/genetics , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-2/genetics , Endosomes/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Animals , Ligands , Protein Binding , Protein Transport
2.
Cell Rep ; 43(5): 114241, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38758647

ABSTRACT

The binding and function of ß-arrestins are regulated by specific phosphorylation motifs present in G protein-coupled receptors (GPCRs). However, the exact arrangement of phosphorylated amino acids responsible for establishing a stable interaction remains unclear. We employ a 1D sequence convolution model trained on GPCRs with established ß-arrestin-binding properties. With this approach, amino acid motifs characteristic of GPCRs that form stable interactions with ß-arrestins can be identified, a pattern that we name "arreSTick." Intriguingly, the arreSTick pattern is also present in numerous non-receptor proteins. Using proximity biotinylation assay and mass spectrometry analysis, we demonstrate that the arreSTick motif controls the interaction between many non-receptor proteins and ß-arrestin2. The HIV-1 Tat-specific factor 1 (HTSF1 or HTATSF1), a nuclear transcription factor, contains the arreSTick pattern, and its subcellular localization is influenced by ß-arrestin2. Our findings unveil a broader role for ß-arrestins in phosphorylation-dependent interactions, extending beyond GPCRs to encompass non-receptor proteins as well.


Subject(s)
Amino Acid Motifs , Protein Binding , beta-Arrestins , Phosphorylation , Humans , beta-Arrestins/metabolism , HEK293 Cells , beta-Arrestin 2/metabolism , Amino Acid Sequence , Protein Stability
3.
Biomedicines ; 11(11)2023 Nov 09.
Article in English | MEDLINE | ID: mdl-38002004

ABSTRACT

Angiotensin II (Ang II) is a hormone with much more complex actions than is typical for other agonists with heterotrimeric G protein-coupled receptors (GPCRs) [...].

4.
Front Endocrinol (Lausanne) ; 14: 1173601, 2023.
Article in English | MEDLINE | ID: mdl-37293495

ABSTRACT

The diluting and concentrating function of the kidney plays a crucial role in regulating the water homeostasis of the body. This function is regulated by the antidiuretic hormone, arginine vasopressin through the type 2 vasopressin receptor (V2R), allowing the body to adapt to periods of water load or water restriction. Loss-of-function mutations of the V2R cause X-linked nephrogenic diabetes insipidus (XNDI), which is characterized by polyuria, polydipsia, and hyposthenuria. Gain-of-function mutations of the V2R lead to nephrogenic syndrome of inappropriate antidiuresis disease (NSIAD), which results in hyponatremia. Various mechanisms may be responsible for the impaired receptor functions, and this review provides an overview of recent findings about the potential therapeutic interventions in the light of the current experimental data.


Subject(s)
Receptors, Vasopressin , Vasopressins , Receptors, Vasopressin/genetics , Vasopressins/genetics , Mutation , Water , Molecular Biology
5.
Int J Mol Sci ; 24(4)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36835391

ABSTRACT

Angiotensin II (AngII) is a vasoactive peptide hormone, which, under pathological conditions, contributes to the development of cardiovascular diseases. Oxysterols, including 25-hydroxycholesterol (25-HC), the product of cholesterol-25-hydroxylase (CH25H), also have detrimental effects on vascular health by affecting vascular smooth muscle cells (VSMCs). We investigated AngII-induced gene expression changes in VSMCs to explore whether AngII stimulus and 25-HC production have a connection in the vasculature. RNA-sequencing revealed that Ch25h is significantly upregulated in response to AngII stimulus. The Ch25h mRNA levels were elevated robustly (~50-fold) 1 h after AngII (100 nM) stimulation compared to baseline levels. Using inhibitors, we specified that the AngII-induced Ch25h upregulation is type 1 angiotensin II receptor- and Gq/11 activity-dependent. Furthermore, p38 MAPK has a crucial role in the upregulation of Ch25h. We performed LC-MS/MS to identify 25-HC in the supernatant of AngII-stimulated VSMCs. In the supernatants, 25-HC concentration peaked 4 h after AngII stimulation. Our findings provide insight into the pathways mediating AngII-induced Ch25h upregulation. Our study elucidates a connection between AngII stimulus and 25-HC production in primary rat VSMCs. These results potentially lead to the identification and understanding of new mechanisms in the pathogenesis of vascular impairments.


Subject(s)
Angiotensin II , Muscle, Smooth, Vascular , Steroid Hydroxylases , Animals , Rats , Angiotensin II/metabolism , Cells, Cultured , Chromatography, Liquid , Gene Expression , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/metabolism , Tandem Mass Spectrometry , Steroid Hydroxylases/genetics
6.
Front Pharmacol ; 13: 811836, 2022.
Article in English | MEDLINE | ID: mdl-35153784

ABSTRACT

The urine concentrating function of the kidney is essential to maintain the water homeostasis of the human body. It is mainly regulated by the arginine-vasopressin (AVP), which targets the type 2 vasopressin receptor (V2R) in the kidney. The inability of V2R to respond to AVP stimulation leads to decreased urine concentration and congenital nephrogenic diabetes insipidus (NDI). NDI is characterized by polyuria, polydipsia, and hyposthenuria. In this study, we identified a point mutation (S127F) in the AVPR2 gene of an NDI patient, and we characterized the impaired function of the V2R mutant in HEK293 cells. Based on our data, the S127F-V2R mutant is almost exclusively located intracellularly in the endoplasmic reticulum (ER), and very few receptors were detected at the cell surface, where the receptor can bind to AVP. The overexpressed S127F-V2R mutant receptor has negligible cAMP generation capability compared to the wild-type receptor in response to AVP stimulation. Since certain misfolded mutant proteins, that are retained in the ER, can be rescued by pharmacological chaperones, we examined the potential rescue effects of two pharmacochaperones on the S127F-V2R. We found that pretreatment with both tolvaptan (an established V2R inverse agonist) and MCF14 compound (a cell-permeable high-affinity agonist for the V2R) were capable of partially restoring the cAMP generating function of the receptor in response to vasopressin stimulation. According to our data, both cell permeant agonists and antagonists can function as pharmacochaperones, and serve as the starting compounds to develop medicines for patients carrying the S127F mutation.

7.
Cells ; 10(12)2021 12 15.
Article in English | MEDLINE | ID: mdl-34944046

ABSTRACT

Activation of the type I angiotensin receptor (AT1-R) in vascular smooth muscle cells (VSMCs) plays a crucial role in the regulation of blood pressure; however, it is also responsible for the development of pathological conditions such as vascular remodeling, hypertension and atherosclerosis. Stimulation of the VSMC by angiotensin II (AngII) promotes a broad variety of biological effects, including gene expression changes. In this paper, we have taken an integrated approach in which an analysis of AngII-induced gene expression changes has been combined with the use of small-molecule inhibitors and lentiviral-based gene silencing, to characterize the mechanism of signal transduction in response to AngII stimulation in primary rat VSMCs. We carried out Affymetrix GeneChip experiments to analyze the effects of AngII stimulation on gene expression; several genes, including DUSP5, DUSP6, and DUSP10, were identified as upregulated genes in response to stimulation. Since various dual-specificity MAPK phosphatase (DUSP) enzymes are important in the regulation of mitogen-activated protein kinase (MAPK) signaling pathways, these genes have been selected for further analysis. We investigated the kinetics of gene-expression changes and the possible signal transduction processes that lead to altered expression changes after AngII stimulation. Our data shows that the upregulated genes can be stimulated through multiple and synergistic signal transduction pathways. We have also found in our gene-silencing experiments that epidermal growth factor receptor (EGFR) transactivation is not critical in the AngII-induced expression changes of the investigated genes. Our data can help us understand the details of AngII-induced long-term effects and the pathophysiology of AT1-R. Moreover, it can help to develop potential interventions for those symptoms that are induced by the over-functioning of this receptor, such as vascular remodeling, cardiac hypertrophy or atherosclerosis.


Subject(s)
Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinase Phosphatases/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/enzymology , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/pharmacology , Animals , Cell Line , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Kinetics , Lentivirus/metabolism , Male , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Rats, Wistar , Signal Transduction/drug effects , Time Factors , Up-Regulation/genetics
8.
Front Endocrinol (Lausanne) ; 12: 714561, 2021.
Article in English | MEDLINE | ID: mdl-34484125

ABSTRACT

ß-arrestins are partners of the G protein-coupled receptors (GPCRs), regulating their intracellular trafficking and signaling. Development of biased GPCR agonists, selectively targeting either G protein or ß-arrestin pathways, are in the focus of interest due to their therapeutic potential in different pathological conditions. The CB2 cannabinoid receptor (CB2R) is a GPCR involved in various functions in the periphery and the central nervous system. Two common occurring variants of CB2R, harboring Q63R or L133I missense mutations, have been implicated in the development of a diverse set of disorders. To evaluate the effect of these mutations, we characterized the binding profile of these mutant CB2 receptors to G proteins and ß-arrestin2. Although their ability to inhibit cAMP signaling was similar, the Q63R mutant had increased, whereas the L133I mutant receptor had decreased ß-arrestin2 binding. In line with these observations, the variants also had altered intracellular trafficking. Our results show that two common variants of the CB2 receptor have biased signaling properties, which may contribute to the pathogenesis of the associated disorders and may offer CB2R as a target for further development of biased receptor activation strategies.


Subject(s)
Mutation, Missense , Receptor, Cannabinoid, CB2/metabolism , beta-Arrestins/metabolism , HEK293 Cells , Humans , Protein Binding , Protein Transport , Receptor, Cannabinoid, CB2/chemistry , Receptor, Cannabinoid, CB2/genetics , beta-Arrestins/genetics
9.
J Biol Chem ; 296: 100366, 2021.
Article in English | MEDLINE | ID: mdl-33545176

ABSTRACT

Reliable measurement of ligand binding to cell surface receptors is of outstanding biological and pharmacological importance. Resonance energy transfer-based assays are powerful approaches to achieve this goal, but the currently available methods are hindered by the necessity of receptor tagging, which can potentially alter ligand binding properties. Therefore, we developed a tag-free system to measure ligand‒receptor interactions in live cells using the Gaussia luciferase (GLuc) as a bioluminescence resonance energy transfer donor. GLuc is as small as the commonly applied Nanoluciferase but has enhanced brightness, and its proper substrate is the frequently used coelenterazine. In our assay, bystander bioluminescence resonance energy transfer is detected between a GLuc-based extracellular surface biosensor and fluorescent ligands bound to their unmodified receptors. The broad spectrum of applications includes equilibrium and kinetic ligand binding measurements for both labeled and competitive unlabeled ligands, and the assay can be utilized for different classes of plasma membrane receptors. Furthermore, the assay is suitable for high-throughput screening, as evidenced by the identification of novel α1 adrenergic receptor ligands. Our data demonstrate that GLuc-based biosensors provide a simple, sensitive, and cost-efficient platform for drug characterization and development.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Luciferases/chemistry , Luciferases/metabolism , Biological Assay , Cell Membrane/metabolism , Energy Transfer , HEK293 Cells , Humans , Kinetics , Ligands , Protein Binding , Protein Transport , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
10.
Exp Suppl ; 111: 317-339, 2019.
Article in English | MEDLINE | ID: mdl-31588538

ABSTRACT

Body fluid homeostasis is essential for normal life. In the maintenance of water balance, the most important factor and regulated process is the excretory function of the kidneys. The kidneys are capable to compensate not only the daily fluctuations of water intake but also the consequences of fluid loss (respiration, perspiration, sweating, hemorrhage). The final volume and osmolality of the excreted urine is set in the collecting duct via hormonal regulation. The hormone of water conservation is the vasopressin (AVP), and a large volume of urine is produced and excreted in the absence of AVP secretion or if AVP is ineffective in the kidneys. The aquaporin-2 water channel (AQP2) is expressed in the principal cells, and it plays an essential role in the reabsorption of water in the collecting ducts via type 2 vasopressin receptor (V2R)-mediated mechanism. If neural or hormonal regulation fails to operate the normal function of AVP-V2R-AQP2 system, it can result in various diseases such as diabetes insipidus (DI) or nephrogenic syndrome of inappropriate diuresis (NSIAD). The DI is characterized by excessive production of hyposmotic urine ("insipidus" means tasteless) due to the inability of the kidneys to concentrate urine. In this chapter, we focus and discuss the pathophysiology of nephrogenic DI (NDI) and the potential therapeutic interventions in the light of the current experimental data.


Subject(s)
Diabetes Insipidus, Nephrogenic/physiopathology , Aquaporin 2/metabolism , Diabetes Insipidus, Nephrogenic/therapy , Humans , Receptors, Vasopressin/metabolism , Water-Electrolyte Balance
11.
Article in English | MEDLINE | ID: mdl-31447777

ABSTRACT

AT1 angiotensin receptor plays important physiological and pathophysiological roles in the cardiovascular system. Renin-angiotensin system represents a target system for drugs acting at different levels. The main effects of ATR1 stimulation involve activation of Gq proteins and subsequent IP3, DAG, and calcium signaling. It has become evident in recent years that besides the well-known G protein pathways, AT1R also activates a parallel signaling pathway through ß-arrestins. ß-arrestins were originally described as proteins that desensitize G protein-coupled receptors, but they can also mediate receptor internalization and G protein-independent signaling. AT1R is one of the most studied receptors, which was used to unravel the newly recognized ß-arrestin-mediated pathways. ß-arrestin-mediated signaling has become one of the most studied topics in recent years in molecular pharmacology and the modulation of these pathways of the AT1R might offer new therapeutic opportunities in the near future. In this paper, we review the recent advances in the field of ß-arrestin signaling of the AT1R, emphasizing its role in cardiovascular regulation and heart failure.

12.
Article in English | MEDLINE | ID: mdl-29678287

ABSTRACT

AT1 angiotensin receptor (AT1R), a prototypical G protein-coupled receptor (GPCR), is the main receptor, which mediates the effects of the renin-angiotensin system (RAS). AT1R plays a crucial role in the regulation of blood pressure and salt-water homeostasis, and in the development of pathological conditions, such as hypertension, heart failure, cardiovascular remodeling, renal fibrosis, inflammation, and metabolic disorders. Stimulation of AT1R leads to pleiotropic signal transduction pathways generating arrays of complex cellular responses. Growing amount of evidence shows that AT1R is a versatile GPCR, which has multiple unique faces with distinct conformations and signaling properties providing new opportunities for functionally selective pharmacological targeting of the receptor. Biased ligands of AT1R have been developed to selectively activate the ß-arrestin pathway, which may have therapeutic benefits compared to the conventional angiotensin converting enzyme inhibitors and angiotensin receptor blockers. In this review, we provide a summary about the most recent findings and novel aspects of the AT1R function, signaling, regulation, dimerization or oligomerization and its cross-talk with other receptors, including epidermal growth factor (EGF) receptor, adrenergic receptors and CB1 cannabinoid receptor. Better understanding of the mechanisms and structural aspects of AT1R activation and cross-talk can lead to the development of novel type of drugs for the treatment of cardiovascular and other diseases.


Subject(s)
Receptor Cross-Talk , Receptor, Angiotensin, Type 1/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Animals , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins/physiology , Receptor Cross-Talk/physiology
13.
J Biol Chem ; 293(3): 876-892, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29146594

ABSTRACT

ß-Arrestins are key regulators and signal transducers of G protein-coupled receptors (GPCRs). The interaction between receptors and ß-arrestins is generally believed to require both receptor activity and phosphorylation by GPCR kinases. In this study, we investigated whether ß-arrestins are able to bind second messenger kinase-phosphorylated, but inactive receptors as well. Because heterologous phosphorylation is a common phenomenon among GPCRs, this mode of ß-arrestin activation may represent a novel mechanism of signal transduction and receptor cross-talk. Here we demonstrate that activation of protein kinase C (PKC) by phorbol myristate acetate, Gq/11-coupled GPCR, or epidermal growth factor receptor stimulation promotes ß-arrestin2 recruitment to unliganded AT1 angiotensin receptor (AT1R). We found that this interaction depends on the stability lock, a structure responsible for the sustained binding between GPCRs and ß-arrestins, formed by phosphorylated serine-threonine clusters in the receptor's C terminus and two conserved phosphate-binding lysines in the ß-arrestin2 N-domain. Using improved FlAsH-based serine-threonine clusters ß-arrestin2 conformational biosensors, we also show that the stability lock not only stabilizes the receptor-ß-arrestin interaction, but also governs the structural rearrangements within ß-arrestins. Furthermore, we found that ß-arrestin2 binds to PKC-phosphorylated AT1R in a distinct active conformation, which triggers MAPK recruitment and receptor internalization. Our results provide new insights into the activation of ß-arrestins and reveal their novel role in receptor cross-talk.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , beta-Arrestins/metabolism , Angiotensin II/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Immunoblotting , Microscopy, Confocal , Phosphorylation , Receptors, G-Protein-Coupled/metabolism
14.
J Biol Chem ; 292(46): 18862-18877, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28939768

ABSTRACT

Plasma membrane (PM) localization of Ras proteins is crucial for transmitting signals upon mitogen stimulation. Post-translational lipid modification of Ras proteins plays an important role in their recruitment to the PM. Electrostatic interactions between negatively charged PM phospholipids and basic amino acids found in K-Ras4B (K-Ras) but not in H-Ras are important for permanent K-Ras localization to the PM. Here, we investigated how acute depletion of negatively charged PM polyphosphoinositides (PPIns) from the PM alters the intracellular distribution and activity of K- and H-Ras proteins. PPIns depletion from the PM was achieved either by agonist-induced activation of phospholipase C ß or with a rapamycin-inducible system in which various phosphatidylinositol phosphatases were recruited to the PM. Redistribution of the two Ras proteins was monitored with confocal microscopy or with a recently developed bioluminescence resonance energy transfer-based approach involving fusion of the Ras C-terminal targeting sequences or the entire Ras proteins to Venus fluorescent protein. We found that PM PPIns depletion caused rapid translocation of K-Ras but not H-Ras from the PM to the Golgi. PM depletion of either phosphatidylinositol 4-phosphate (PtdIns4P) or PtdIns(4,5)P2 but not PtdIns(3,4,5)P3 was sufficient to evoke K-Ras translocation. This effect was diminished by deltarasin, an inhibitor of the Ras-phosphodiesterase interaction, or by simultaneous depletion of the Golgi PtdIns4P. The PPIns depletion decreased incorporation of [3H]leucine in K-Ras-expressing cells, suggesting that Golgi-localized K-Ras is not as signaling-competent as its PM-bound form. We conclude that PPIns in the PM are important regulators of K-Ras-mediated signals.


Subject(s)
Cell Membrane/metabolism , Golgi Apparatus/metabolism , Phosphatidylinositol Phosphates/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , COS Cells , Chlorocebus aethiops , Diphosphates/metabolism , HEK293 Cells , Humans , Protein Transport
15.
Biochim Biophys Acta ; 1861(3): 177-87, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26692031

ABSTRACT

Deciphering many roles played by inositol lipids in signal transduction and membrane function demands experimental approaches that can detect their dynamic accumulation with subcellular accuracy and exquisite sensitivity. The former criterion is met by imaging of fluorescence biosensors in living cells, whereas the latter is facilitated by biochemical measurements from populations. Here, we introduce BRET-based biosensors able to detect rapid changes in inositol lipids in cell populations with both high sensitivity and subcellular resolution in a single, convenient assay. We demonstrate robust and sensitive measurements of PtdIns4P, PtdIns(4,5)P2 and PtdIns(3,4,5)P3 dynamics, as well as changes in cytoplasmic Ins(1,4,5)P3 levels. Measurements were made during either experimental activation of lipid degradation, or PI 3-kinase and phospholipase C mediated signal transduction. Our results reveal a previously unappreciated synthesis of PtdIns4P that accompanies moderate activation of phospholipase C signaling downstream of both EGF and muscarinic M3 receptor activation. This signaling-induced PtdIns4P synthesis relies on protein kinase C, and implicates a feedback mechanism in the control of inositol lipid metabolism during signal transduction.


Subject(s)
Biosensing Techniques , Carbachol/pharmacology , Epidermal Growth Factor/pharmacology , ErbB Receptors/agonists , Fluorescence Resonance Energy Transfer , Muscarinic Agonists/pharmacology , Phosphatidylinositol Phosphates/metabolism , Protein Kinase C/metabolism , Receptors, Muscarinic/drug effects , Signal Transduction/drug effects , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Feedback, Physiological , HEK293 Cells , Humans , Hydrolysis , Inositol 1,4,5-Trisphosphate/metabolism , Kinetics , Lipolysis , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Receptor, Muscarinic M3 , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , Type C Phospholipases/metabolism , Up-Regulation
16.
Kidney Int ; 88(5): 1070-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26131744

ABSTRACT

Nephrogenic syndrome of inappropriate antidiuresis (NSIAD) is a recently discovered rare disease caused by gain-of-function mutations of the V2 vasopressin receptor gene, AVPR2. To date, mutations of Phe229 and Arg137 have been identified as gain-of-function in the V2 vasopressin receptor (V2R). These receptor mutations lead to hyponatremia, which may lead to clinical symptoms in infants. Here we present a newly identified I130N substitution in exon 2 of the V2R gene in a family, causing NSIAD. This I130N mutation resulted in constitutive activity of the V2R with constitutive cyclic adenosine monophosphate (cAMP) generation in HEK293 cells. This basal activity could be blocked by the inverse agonist tolvaptan and arginine-vasopressin stimulation enhanced the cAMP production of I130N-V2R. The mutation causes a biased receptor conformation as the basal cAMP generation activity of I130N does not lead to interaction with ß-arrestin. The constitutive activity of the mutant receptor caused constitutive dynamin-dependent and ß-arrestin-independent internalization. The inhibition of basal internalization using dominant-negative dynamin resulted in an increased cell surface expression. In contrast to the constitutive internalization, agonist-induced endocytosis was ß-arrestin dependent. Thus, tolvaptan could be used for treatment of hyponatremia in patients with NSIAD who carry the I130N-V2R mutation.


Subject(s)
Cyclic AMP/biosynthesis , Genetic Diseases, X-Linked/genetics , Hyponatremia/genetics , Inappropriate ADH Syndrome/genetics , Receptors, Vasopressin/genetics , Adult , Antidiuretic Hormone Receptor Antagonists/pharmacology , Arrestins/metabolism , Benzazepines/pharmacology , Cell Membrane/chemistry , DNA Mutational Analysis , Dynamins/metabolism , Endocytosis/drug effects , Exons , Female , HEK293 Cells , Humans , Hyponatremia/drug therapy , Male , Mutation , Pedigree , Receptors, Vasopressin/analysis , Receptors, Vasopressin/metabolism , Tolvaptan , beta-Arrestins
17.
Mol Pharmacol ; 87(6): 972-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25804845

ABSTRACT

Biased agonism on the type I angiotensin receptor (AT1-R) can achieve different outcomes via activation of G protein-dependent and -independent cellular responses. In this study, we investigated whether the biased activation of AT1-R can lead to different regulation and intracellular processing of the receptor. We analyzed ß-arrestin binding, endocytosis, and subsequent trafficking steps, such as early and late phases of recycling of AT1-R in human embryonic kidney 293 cells expressing wild-type or biased mutant receptors in response to different ligands. We used Renilla luciferase-tagged receptors and yellow fluorescent protein-tagged ß-arrestin2, Rab5, Rab7, and Rab11 proteins in bioluminescence resonance energy transfer measurements to follow the fate of the receptor after stimulation. We found that not only is the signaling of the receptor different upon using selective ligands, but the fate within the cells is also determined by the type of the stimulation. ß-arrestin binding and the internalization kinetics of the angiotensin II-stimulated AT1-R differed from those stimulated by the biased agonists. Similarly, angiotensin II-stimulated wild-type AT1-R showed differences compared with a biased mutant AT1-R (DRY/AAY AT1-R) with regards to ß-arrestin binding and endocytosis. We found that the differences in the internalization kinetics of the receptor in response to biased agonist stimulation are due to the differences in plasma membrane phosphatidylinositol 4,5-bisphosphate depletion. Moreover, the stability of the ß-arrestin binding is a major determinant of the later fate of the internalized AT1-R receptor.


Subject(s)
Receptor, Angiotensin, Type 1/metabolism , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Angiotensin II/pharmacology , Arrestins/genetics , Arrestins/metabolism , Bioluminescence Resonance Energy Transfer Techniques , Cell Membrane/metabolism , Endocytosis/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Hydrolysis , Ligands , Luciferases, Renilla/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoinositide Phospholipase C/genetics , Phosphoinositide Phospholipase C/metabolism , Phosphoinositide-3 Kinase Inhibitors , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/genetics , beta-Arrestins
18.
Mol Endocrinol ; 28(5): 634-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24628417

ABSTRACT

Loss-of-function mutations of the type 2 vasopressin receptor (V2R) in kidney can lead to nephrogenic diabetes insipidus (NDI). We studied a previously described, but uncharacterized, mutation of the V2R (N321K missense mutation) of a patient with NDI. The properties of the mutant receptor were evaluated. We constructed a highly sensitive Epac-based bioluminescence resonance energy transfer biosensor to perform real-time cAMP measurements after agonist stimulation of transiently transfected HEK293 cells with V2Rs. ß-Arrestin binding of the activated receptors was examined with luciferase-tagged ß-arrestin and mVenus-tagged V2Rs using the bioluminescence resonance energy transfer technique. Cell surface expression levels of hemagglutinin-tagged receptors were determined with flow cytometry using anti-hemagglutinin-Alexa 488 antibodies. Cellular localization examinations were implemented with fluorescent tagged receptors visualized with confocal laser scanning microscopy. The effect of various vasopressin analogs on the type 1 vasopressin receptor (V1R) was tested on mouse arteries by wire myography. The N321K mutant V2R showed normal cell surface expression, but the potency of arginine vasopressin for cAMP generation was low, whereas the clinically used desmopressin was not efficient. The ß-arrestin binding and internalization properties of the mutant receptor were also different than those for the wild type. The function of the mutant receptor can be rescued with administration of the V2R agonist Val(4)-desmopressin, which had no detectable side effects on V1R in the effective cAMP generating concentrations. Based on these findings we propose a therapeutic strategy for patients with NDI carrying the N321K mutation, as our in vivo experiments suggest that Val(4)-desmopressin could rescue the function of the N321K-V2R without significant side effects on the V1R.


Subject(s)
Diabetes Insipidus, Nephrogenic/genetics , Receptors, Vasopressin/genetics , Adult , Animals , Antidiuretic Agents/pharmacology , Cyclic AMP/metabolism , DNA Mutational Analysis , Deamino Arginine Vasopressin/pharmacology , Diabetes Insipidus, Nephrogenic/drug therapy , Endoplasmic Reticulum , HEK293 Cells , Humans , In Vitro Techniques , Male , Mice , Mutation, Missense , Neurophysins/pharmacology , Neurophysins/physiology , Protein Precursors/pharmacology , Protein Precursors/physiology , Rats , Receptors, Vasopressin/agonists , Second Messenger Systems , Vasopressins/pharmacology , Vasopressins/physiology
19.
J Biol Chem ; 287(37): 31540-50, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22787147

ABSTRACT

In the vascular system angiotensin II (Ang II) causes vasoconstriction via the activation of type 1 angiotensin receptors. Earlier reports have shown that in cellular expression systems diacylglycerol produced during type 1 angiotensin receptor signaling can be converted to 2-arachidonoylglycerol, an important endocannabinoid. Because activation of CB(1) cannabinoid receptors (CB(1)R) induces vasodilation and reduces blood pressure, we have tested the hypothesis that Ang II-induced 2-arachidonoylglycerol release can modulate its vasoconstrictor action in vascular tissue. Rat and mouse skeletal muscle arterioles and mouse saphenous arteries were isolated, pressurized, and subjected to microangiometry. Vascular expression of CB(1)R was demonstrated using Western blot and RT-PCR. In accordance with the functional relevance of these receptors WIN55212, a CB(1)R agonist, caused vasodilation, which was absent in CB(1)R knock-out mice. Inhibition of CB(1)Rs using O2050, a neutral antagonist, enhanced the vasoconstrictor effect of Ang II in wild type but not in CB(1)R knock-out mice. Inverse agonists of CB(1)R (SR141716 and AM251) and inhibition of diacylglycerol lipase using tetrahydrolipstatin also augmented the Ang II-induced vasoconstriction, suggesting that endocannabinoid release modulates this process via CB(1)R activation. This effect was independent of nitric-oxide synthase activity and endothelial function. These data demonstrate that Ang II stimulates vascular endocannabinoid formation, which attenuates its vasoconstrictor effect, suggesting that endocannabinoid release from the vascular wall and CB(1)R activation reduces the vasoconstrictor and hypertensive effects of Ang II.


Subject(s)
Angiotensin II/metabolism , Arteries/metabolism , Endocannabinoids/metabolism , Endothelium, Vascular/metabolism , Muscle, Skeletal/metabolism , Receptor, Cannabinoid, CB1/metabolism , Vasoconstriction/physiology , Analgesics/pharmacology , Angiotensin II/genetics , Animals , Benzoxazines/pharmacology , Endocannabinoids/antagonists & inhibitors , Endocannabinoids/genetics , Hypertension/genetics , Hypertension/metabolism , Male , Mice , Mice, Knockout , Morpholines/pharmacology , Muscle, Skeletal/blood supply , Naphthalenes/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/genetics , Rimonabant , Vasoconstriction/drug effects
20.
J Biol Chem ; 287(12): 9090-9, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22291018

ABSTRACT

Initiation and termination of signaling of the type I angiotensin receptor (AT(1)-R) can lead to dynamic changes in its localization in plasma membrane microdomains. Several markers were recently developed to investigate membrane microdomains. Here, we used several YFP-labeled fusion constructs (i.e. raft or non-raft plasma membrane markers) to analyze the agonist-induced changes in compartmentalization of AT(1)-R, including internalization or lateral movement between plasma membrane compartments in response to stimulation using bioluminescence resonance energy transfer measurements. Our data demonstrate that angiotensin II (AngII) stimulus changes the microdomain localization of wild type or mutated (DRY → AAY or TSTS → AAAA) AT(1)-Rs co-expressed with the fluorescent probes in HEK293 cells. The comparison of the trafficking of AT(1)-R upon AngII stimulus with those of [Sar(1),Ile(8)]AngII or [Sar(1),Ile(4),Ile(8)]AngII stimulus revealed different types of changes, depending on the nature of the ligand. The observed changes in receptor compartmentalization of the AT(1)-R are strikingly different from those of 5HT-2C and EGF receptors, which demonstrate the usefulness of the bioluminescence resonance energy transfer-based measurements in the investigation of receptor trafficking in the plasma membrane in living cell experiments.


Subject(s)
Biosensing Techniques/methods , Luminescent Measurements/methods , Membrane Microdomains/metabolism , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Energy Transfer , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/genetics , Microscopy, Confocal , Protein Binding , Protein Transport , Receptor, Angiotensin, Type 1/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL