Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Med Chem ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38960379

ABSTRACT

Amylin, a member of the calcitonin family, acts via amylin receptors in the hindbrain and hypothalamus to suppress appetite. Native ligands of these receptors are peptides with short half-lives. Conjugating fatty acids to these peptides can increase their half-lives. The long-acting human amylin analog, NN1213, was generated from structure-activity efforts optimizing solubility, stability, receptor affinity, and selectivity, as well as in vivo potency and clearance. In both rats and dogs, a single dose of NN1213 reduced appetite in a dose-dependent manner and with a long duration of action. Consistent with the effect on appetite, studies in obese rats demonstrated that daily NN1213 dosing resulted in a dose-dependent reduction in body weight over a 21-day period. Magnetic resonance imaging indicated that this was primarily driven by loss of fat mass. Based on these data, NN1213 could be considered an attractive option for weight management in the clinical setting.

2.
J Med Chem ; 64(15): 11183-11194, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34288673

ABSTRACT

A hallmark of the pancreatic hormone amylin is its high propensity toward the formation of amyloid fibrils, which makes it a challenging drug design effort. The amylin analogue pramlintide is commercially available for diabetes treatment as an adjunct to insulin therapy but requires three daily injections due to its short half-life. We report here the development of the stable, lipidated long-acting amylin analogue cagrilintide (23) and some of the structure-activity efforts that led to the selection of this analogue for clinical development with obesity as an indication. Cagrilintide is currently in clinical trial and has induced significant weight loss when dosed alone or in combination with the GLP-1 analogue semaglutide.


Subject(s)
Drug Development , Hypoglycemic Agents/pharmacology , Islet Amyloid Polypeptide/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Islet Amyloid Polypeptide/chemical synthesis , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/pharmacology , Models, Molecular , Molecular Structure , Structure-Activity Relationship
3.
Mol Pharmacol ; 98(1): 49-60, 2020 07.
Article in English | MEDLINE | ID: mdl-32358164

ABSTRACT

Negative allosteric modulation of the metabotropic glutamate 5 (mGlu5) receptor has emerged as a potential strategy for the treatment of neurologic disorders. Despite the success in preclinical studies, many mGlu5 negative allosteric modulators (NAMs) that have reached clinical trials failed due to lack of efficacy. In this study, we provide a detailed in vitro pharmacological characterization of nine clinically and preclinically tested NAMs. We evaluated inhibition of l-glutamate-induced signaling with Ca2+ mobilization, inositol monophosphate (IP1) accumulation, extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation, and real-time receptor internalization assays on rat mGlu5 expressed in HEK293A cells. Moreover, we determined association rates (kon) and dissociation rates (koff), as well as NAM affinities with [3H]methoxy-PEPy binding experiments. kon and koff values varied greatly between the nine NAMs (34- and 139-fold, respectively) resulting in long receptor residence times (>400 min) for basimglurant and mavoglurant, medium residence times (10-30 min) for AZD2066, remeglurant, and (RS)-remeglurant, and low residence times (<10 mins) for dipraglurant, F169521, F1699611, and STX107. We found that all NAMs inhibited l-glutamate-induced mGlu5 receptor internalization, generally with a similar potency to IP1 accumulation and ERK1/2 phosphorylation, whereas Ca2+ mobilization was less potently inhibited. Operational model of allosterism analyses revealed that dipraglurant and (RS)-remeglurant were biased toward (affinity) receptor internalization and away (cooperativity) from the ERK1/2 phosphorylation pathway, respectively. Our study is the first to measure mGlu5 NAM binding kinetics and negative allosteric modulation of mGlu5 receptor internalization and adds significant new knowledge about the molecular pharmacology of a diverse range of clinically relevant NAMs. SIGNIFICANCE STATEMENT: The metabotropic glutamate 5 (mGlu5) receptor is important in many brain functions and implicated in several neurological pathologies. Negative allosteric modulators (NAMs) have shown promising results in preclinical models but have so far failed in human clinical trials. Here we provide the most comprehensive and comparative molecular pharmacological study to date of nine preclinically/clinically tested NAMs at the mGlu5 receptor, which is also the first study to measure ligand binding kinetics and negative allosteric modulation of mGlu5 receptor internalization.


Subject(s)
Imidazoles/pharmacology , Indoles/pharmacology , Isoxazoles/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Triazoles/pharmacology , Allosteric Regulation/drug effects , Animals , Calcium/metabolism , HEK293 Cells , Humans , Imidazoles/chemistry , Indoles/chemistry , Inositol Phosphates/metabolism , Isoxazoles/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Structure , Phosphorylation/drug effects , Pyridines/chemistry , Rats , Time Factors , Triazoles/chemistry
4.
Basic Clin Pharmacol Toxicol ; 122(5): 460-469, 2018 May.
Article in English | MEDLINE | ID: mdl-29136335

ABSTRACT

The angiotensin II type I receptor (AT1R) is involved in the regulation of cardiovascular function. Excessive activation of AT1R by angiotensin II (Ang II) leads to cardiovascular disease and may be involved in the development of insulin resistance and diabetes. Functionally selective Ang II analogues, such as the [Sar1, Ile4, Ile8]-angiotensin II (SII Ang II) analogue, that only activate a subset of signalling networks have been demonstrated to have beneficial effects on cardiovascular function in certain settings, including lowering blood pressure and increasing cardiac performance. Here, we studied the effect of SII Ang II on insulin receptor (IR) signalling and glucose metabolism in primary rat hepatocytes. We show that long-term pre-treatment of hepatocytes with SII Ang II increased insulin-stimulated glycogen synthesis, while Ang II and the AT1R antagonist losartan had no effect. Insulin-stimulated suppression of hepatic glucose output was not affected by Ang II or SII Ang II. It is well known that insulin regulates glycogen synthesis and glucose output through Akt-mediated phosphorylation of glycogen synthase kinase α/ß (GSK3α/ß) and forkhead box protein O1 (FOXO1), respectively. In line with this, we show that SII Ang II potentiated insulin-stimulated phosphorylation of Akt and GSK3α/ß, but not FOXO1. Furthermore, we demonstrate that the effect of SII Ang II on insulin-stimulated signalling and glycogen synthesis was dependent on Src and Gαq, as inhibitors of these proteins abolished the potentiating effect of SII Ang II. Thus, our results demonstrate that SII Ang II may have a positive effect on IR signalling and glucose metabolism in hepatocytes.


Subject(s)
Angiotensin II/analogs & derivatives , Energy Metabolism/drug effects , Glucose/metabolism , Glycogen/biosynthesis , Hepatocytes/drug effects , Insulin/pharmacology , Receptor, Angiotensin, Type 1/agonists , Receptor, Insulin/agonists , Angiotensin II/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hepatocytes/metabolism , Male , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Time Factors
5.
J Recept Signal Transduct Res ; 37(6): 590-599, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28854843

ABSTRACT

The angiotensin AT1 receptor is a seven transmembrane (7TM) receptor, which mediates the regulation of blood pressure. Activation of angiotensin AT1 receptor may lead to impaired insulin signaling indicating crosstalk between angiotensin AT1 receptor and insulin receptor signaling pathways. To elucidate the molecular mechanisms behind this crosstalk, we applied the BRET2 technique to monitor the effect of angiotensin II on the interaction between Rluc8 tagged insulin receptor and GFP2 tagged insulin receptor substrates 1, 4, 5 (IRS1, IRS4, IRS5) and Src homology 2 domain-containing protein (Shc). We demonstrate that angiotensin II reduces the interaction between insulin receptor and IRS1 and IRS4, respectively, while the interaction with Shc is unaffected, and this effect is dependent on Gαq activation. Activation of other Gαq-coupled 7TM receptors led to a similar reduction in insulin receptor and IRS4 interactions whereas Gαs- and Gαi-coupled 7TM receptors had no effect. Furthermore, we used a panel of kinase inhibitors to show that angiotensin II engages different pathways when regulating insulin receptor interactions with IRS1 and IRS4. Angiotensin II inhibited the interaction between insulin receptor and IRS1 through activation of ERK1/2, while the interaction between insulin receptor and IRS4 was partially inhibited through protein kinase C dependent mechanisms. We conclude that the crosstalk between angiotensin AT1 receptor and insulin receptor signaling shows a high degree of specificity, and involves Gαq protein, and activation of distinct kinases. Thus, the BRET2 technique can be used as a platform for studying molecular mechanisms of crosstalk between insulin receptor and 7TM receptors.


Subject(s)
Blood Pressure/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Insulin/metabolism , Adaptor Proteins, Signal Transducing , Angiotensin II/administration & dosage , Angiotensin II/metabolism , Bioluminescence Resonance Energy Transfer Techniques , Cell Line , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/drug effects , Protein Domains , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Insulin/genetics , Src Homology 2 Domain-Containing, Transforming Protein 2/genetics , Src Homology 2 Domain-Containing, Transforming Protein 2/metabolism
6.
J Biol Chem ; 291(26): 13689-98, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27189946

ABSTRACT

Calcitonin is a peptide hormone consisting of 32 amino acid residues and the calcitonin receptor is a Class B G protein-coupled receptor (GPCR). The crystal structure of the human calcitonin receptor ectodomain (CTR ECD) in complex with a truncated analogue of salmon calcitonin ([BrPhe(22)]sCT(8-32)) has been determined to 2.1-Å resolution. Parallel analysis of a series of peptide ligands showed that the rank order of binding of the CTR ECD is identical to the rank order of binding of the full-length CTR, confirming the structural integrity and relevance of the isolated CTR ECD. The structure of the CTR ECD is similar to other Class B GPCRs and the ligand binding site is similar to the binding site of the homologous receptors for the calcitonin gene-related peptide (CGRP) and adrenomedulin (AM) recently published (Booe, J. M., Walker, C. S., Barwell, J., Kuteyi, G., Simms, J., Jamaluddin, M. A., Warner, M. L., Bill, R. M., Harris, P. W., Brimble, M. A., Poyner, D. R., Hay, D. L., and Pioszak, A. A. (2015) Mol. Cell 58, 1040-1052). Interestingly the receptor-bound structure of the ligand [BrPhe(22)]sCT(8-32) differs from the receptor-bound structure of the homologous ligands CGRP and AM. They all adopt an extended conformation followed by a C-terminal ß turn, however, [BrPhe(22)]sCT(8-32) adopts a type II turn (Gly(28)-Thr(31)), whereas CGRP and AM adopt type I turns. Our results suggest that a type II turn is the preferred conformation of calcitonin, whereas a type I turn is the preferred conformation of peptides that require RAMPs; CGRP, AM, and amylin. In addition the structure provides a detailed molecular explanation and hypothesis regarding ligand binding properties of CTR and the amylin receptors.


Subject(s)
Calcitonin/chemistry , Fish Proteins/chemistry , Receptors, Calcitonin/chemistry , Salmon , Animals , Calcitonin/genetics , Calcitonin/metabolism , Crystallography, X-Ray , Fish Proteins/genetics , Fish Proteins/metabolism , Humans , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism
7.
PLoS One ; 8(3): e58890, 2013.
Article in English | MEDLINE | ID: mdl-23516570

ABSTRACT

BACKGROUND: Functional cross-talk between seven transmembrane (7TM) receptors can dramatically alter their pharmacological properties, both in vitro and in vivo. This represents an opportunity for the development of novel therapeutics that potentially target more specific biological effects while causing fewer adverse events. Although several studies convincingly have established the existence of 7TM receptor cross-talk, little is known about the frequencey and biological significance of this phenomenon. METHODOLOGY/PRINCIPAL FINDINGS: To evaluate the extent of synergism in 7TM receptor signaling, we took a comprehensive approach and co-expressed 123 different 7TM receptors together with the angiotensin II type 1 receptor (AT1R) and analyzed how each receptor affected the angiotensin II (AngII) response. To monitor the effect we used integrative receptor activation/signaling assay called Receptor Selection and Amplification Technology (R-SAT). In this screen the thromboxane A2α receptor (TPαR) was the only receptor which significantly enhanced the AngII-mediated response. The TPαR-mediated enhancement of AngII signaling was significantly reduced when a signaling deficient receptor mutant (TPαR R130V) was co-expressed instead of the wild-type TPαR, and was completely blocked both by TPαR antagonists and COX inhibitors inhibiting formation of thromboxane A2 (TXA2). CONCLUSIONS/SIGNIFICANCE: We found a functional enhancement of AT1R only when co-expressed with TPαR, but not with 122 other 7TM receptors. In addition, the TPαR must be functionally active, indicating the AT1R enhancement is mediated by a paracrine mechanism. Since we only found one receptor enhancing AT1R potency, our results suggest that functional augmentation through 7TM receptor cross-talk is a rare event that may require specific conditions to occur.


Subject(s)
Receptor, Angiotensin, Type 1/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Angiotensin II/pharmacology , Animals , Cattle , Cell Line , Gene Expression , Humans , Paracrine Communication/drug effects , Receptor Cross-Talk/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Renal Artery/drug effects , Renal Artery/physiology , Signal Transduction/drug effects , Vasoconstriction/drug effects
8.
Cell Physiol Biochem ; 30(3): 642-52, 2012.
Article in English | MEDLINE | ID: mdl-22854413

ABSTRACT

Angiotensin II (AngII) is a key peptide in cardiovascular homeostasis and is a ligand for the Angiotensin II type 1 and 2 seven transmembrane receptors (AT(1)R and AT(2)R). The AT(1) receptor is a seven-transmembrane (7TM) G protein-coupled receptor (GPCR) mediating the majority of the physiological functions of AngII. The AT(1)R mediates its effects through both G protein-dependent and independent signaling, which can be separated by functionally selective agonists. In the present study we investigate the effect of AngII and the ß-arrestin biased agonist [SII]AngII on ischemia-reperfusion injury in rat hearts. Isolated hearts mounted in a Langendorff perfused rat heart preparations showed that preconditioning with [SII]AngII reduced the infarct size induced by global ischemia from 46±8.4% to 22±3.4%. In contrast, neither preconditioning with AngII nor postconditioning with AngII or [SII]AngII had a protective effect. Together these results demonstrate a cardioprotective effect of simultaneous blockade of G protein signaling and activation of G protein independent signaling through AT(1) receptors.


Subject(s)
Receptor, Angiotensin, Type 1/metabolism , Reperfusion Injury/metabolism , Angiotensin II/pharmacology , Animals , Arrestins/pharmacology , Cardiotonic Agents/pharmacology , GTP-Binding Proteins/metabolism , Heart Rate/drug effects , Heart Ventricles/drug effects , Hemodynamics/drug effects , In Vitro Techniques , Male , Pressure , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , beta-Arrestins
9.
J Recept Signal Transduct Res ; 32(2): 57-64, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22272819

ABSTRACT

The insulin receptor (IR) belongs to the receptor tyrosine kinase super family and plays an important role in glucose homeostasis. The receptor interacts with several large docking proteins that mediate signaling from the receptor, including the insulin receptor substrate (IRS) family and Src homology-2-containing proteins (Src). Here, we applied the bioluminescence resonance energy transfer 2 (BRET2) technique to study the IR signaling pathways. The interaction between the IR and the substrates IRS1, IRS4 and Shc was examined in response to ligands with different signaling properties. The association between IR and the interacting partners could successfully be monitored when co-expressing green fluorescent protein 2 (GFP2) tagged substrates with Renilla reniformis luciferase 8 (Rluc8) tagged IR. Through additional optimization steps, we developed a stable and flexible BRET2 assay for monitoring the interactions between the IR and its substrates. Furthermore, the insulin analogue X10 was characterized in the BRET2 assay and was found to be 10 times more potent with respect to IRS1, IRS4 and Shc recruitment compared to human insulin. This study demonstrates that the BRET2 technique can be applied to study IR signaling pathways, and that this assay can be used as a platform for screening and characterization of IR ligands.


Subject(s)
Green Fluorescent Proteins/analysis , Insulin Receptor Substrate Proteins/metabolism , Insulin/pharmacology , Luminescent Measurements , Receptor, Insulin/metabolism , Shc Signaling Adaptor Proteins/metabolism , Cells, Cultured , Humans , Insulin/analogs & derivatives , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Protein Binding , Protein Interaction Domains and Motifs/drug effects , Recombinant Fusion Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1
10.
J Mol Histol ; 42(4): 289-99, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21643937

ABSTRACT

MicroRNAs (miRNAs) regulate gene expression by mediating translational repression or mRNA degradation of their targets, and several miRNAs control developmental decisions through embryogenesis. In the developing heart, miRNA targets comprise key players mediating cardiac lineage determination. However, although several miRNAs have been identified as differentially regulated during cardiac development and disease, their distinct cell-specific localization remains largely undetermined, likely owing to a lack of adequate methods. We therefore report the development of a markedly improved approach combining fluorescence-based miRNA-in situ hybridization (miRNA-ISH) with immunohistochemistry (IHC). We have applied this protocol to differentiating embryoid bodies (EBs) as well as embryonic and adult mouse hearts, to detect miRNAs that were upregulated during EB cardiomyogenesis, as determined by array-based miRNA expression profiling. In this manner, we found specific co-localization of miR-1 to myosin positive cells (cardiomyocytes) of EBs, developing and mature hearts. In contrast, miR-125b and -199a did not localize to cardiomyocytes, as previously suggested for miR-199a, but were rather expressed in connective tissue cells of the heart. More specifically, by co-staining with α-smooth muscle actin (α-SMA) and collagen-I, we found that miR-125b and -199a localize to perivascular α-SMA(-) stromal cells. Our approach thus proved valid for determining cell-specific localization of miRNAs, and the findings we present highlight the importance of determining exact cell-specific localization of miRNAs by sequential miRNA-ISH and IHC in studies aiming at understanding the role of miRNAs and their targets. This approach will hopefully aid in identifying relevant miRNA targets of both the heart and other organs.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , Myocardium/metabolism , Organogenesis/genetics , Animals , Cells, Cultured , Cluster Analysis , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Immunohistochemistry , In Situ Hybridization , Mice , MicroRNAs/metabolism , Myocardium/cytology , Organ Specificity/genetics
11.
Br J Pharmacol ; 164(2): 394-404, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21449976

ABSTRACT

BACKGROUND AND PURPOSE: The angiotensin II type 1 receptor (AT(1)R) is a key regulator of blood pressure and cardiac contractility and is profoundly involved in development of cardiac disease. Since several microRNAs (miRNAs) have been implicated in cardiac disease, we determined whether miRNAs might be regulated by AT(1)R signals in a Gαq/11-dependent or -independent manner. EXPERIMENTAL APPROACH: We performed a global miRNA array analysis of angiotensin II (Ang II)-mediated miRNA regulation in HEK293N cells overexpressing the AT(1)R and focused on separating the role of Gαq/11-dependent and -independent pathways. MiRNA regulation was verified with quantitative PCR in both HEK293N cells and primary cardiac myocytes and fibroblasts. KEY RESULTS: Our studies revealed five miRNAs (miR-29b, -129-3p, -132, -132* and -212) that were up-regulated by Ang II in HEK293N cells. In contrast, the biased Ang II analogue, [Sar1, Ile4, Ile8] Ang II (SII Ang II), which selectively activates Gαq/11-independent signalling, failed to regulate miRNAs in HEK293N cells. Furthermore, Ang II-induced miRNA regulation was blocked following Gαq/11 and Mek1 inhibition. The observed Ang II regulation of miRNA was confirmed in primary cultures of adult cardiac fibroblasts. Interestingly, Ang II did not regulate miRNA expression in cardiac myocytes, but SII Ang II significantly down-regulated miR-129-3p. CONCLUSIONS AND IMPLICATIONS: Five miRNAs were regulated by Ang II through mechanisms depending on Gαq/11 and Erk1/2 activation. These miRNAs may be involved in Ang II-mediated cardiac biology and disease, as several of these miRNAs have previously been associated with cardiovascular disease and were found to be regulated in cardiac cells.


Subject(s)
Fibroblasts/physiology , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Myocytes, Cardiac/physiology , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/pharmacology , Anthracenes/pharmacology , Butadienes/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Imidazoles/pharmacology , MicroRNAs/genetics , Nitriles/pharmacology , Pyridines/pharmacology , Receptor, Angiotensin, Type 1/genetics , Signal Transduction
12.
Regul Pept ; 167(1): 21-5, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21115072

ABSTRACT

The angiotensin II type 1 receptor (AT1R) blocker (ARB) Losartan has cardioprotective effects during ischemia-reperfusion injury and inhibits reperfusion arrhythmias -effects that go beyond the benefits of lowering blood pressure. The renin-angiotensin and kallikrein-kinin systems are intricately connected and some of the cardioprotective effects of Losartan are abolished by blocking the bradykinin B2 receptor (B2R) signaling. In this study, we investigated the ability of six clinically available ARBs to specifically bind and activate the B2R. First, we investigated their ability to activate phosphoinositide (PI) hydrolysis in COS-7 cells transiently expressing the B2R. We found that only Losartan activated the B2R, working as a partial agonist compared to the endogenous ligand bradykinin. This effect was blocked by the B2R antagonist HOE 140. A competitive binding analysis revealed that Losartan does not significantly compete with bradykinin and does not change the binding affinity of bradykinin on the B2R. Furthermore, Losartan but not Candesartan mimicked the ability of bradykinin to increase the recovery of contractile force after metabolic stress in rat atrial tissue strips. In conclusion, Losartan is a partial agonist of the B2R through direct binding and activation, suggesting that B2R agonism could partly explain the beneficial effects of Losartan.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Bradykinin/pharmacology , Losartan/pharmacology , Receptor, Bradykinin B2 , Angiotensin II Type 1 Receptor Blockers/metabolism , Angiotensins/metabolism , Animals , Benzimidazoles/pharmacology , Binding, Competitive , Biphenyl Compounds , Bradykinin/analogs & derivatives , Bradykinin/metabolism , Bradykinin B2 Receptor Antagonists , COS Cells , Chlorocebus aethiops , Hydrolysis , Kallikrein-Kinin System/physiology , Losartan/metabolism , Myocardial Contraction/drug effects , Phosphatidylinositols/metabolism , Rats , Receptor, Angiotensin, Type 1/metabolism , Receptor, Bradykinin B2/agonists , Receptor, Bradykinin B2/metabolism , Renin-Angiotensin System/physiology , Signal Transduction/drug effects , Tetrazoles/pharmacology
13.
PLoS One ; 5(11): e14135, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21152433

ABSTRACT

BACKGROUND: Seven transmembrane receptors (7TMRs) can adopt different active conformations facilitating a selective activation of either G protein or ß-arrestin-dependent signaling pathways. This represents an opportunity for development of novel therapeutics targeting selective biological effects of a given receptor. Several studies on pathway separation have been performed, many of these on the Angiotensin II type 1 receptor (AT1R). It has been shown that certain ligands or mutations facilitate internalization and/or recruitment of ß-arrestins without activation of G proteins. However, the underlying molecular mechanisms remain largely unresolved. For instance, it is unclear whether such selective G protein-uncoupling is caused by a lack of ability to interact with G proteins or rather by an increased ability of the receptor to recruit ß-arrestins. Since uncoupling of G proteins by increased ability to recruit ß-arrestins could lead to different cellular or in vivo outcomes than lack of ability to interact with G proteins, it is essential to distinguish between these two mechanisms. METHODOLOGY/PRINCIPAL FINDINGS: We studied five AT1R mutants previously published to display pathway separation: D74N, DRY/AAY, Y292F, N298A, and Y302F (Ballesteros-Weinstein numbering: 2.50, 3.49-3.51, 7.43, 7.49, and 7.53). We find that D74N, DRY/AAY, and N298A mutants are more prone to ß-arrestin recruitment than WT. In contrast, receptor mutants Y292F and Y302F showed impaired ability to recruit ß-arrestin in response to Sar1-Ile4-Ile8 (SII) Ang II, a ligand solely activating the ß-arrestin pathway. CONCLUSIONS/SIGNIFICANCE: Our analysis reveals that the underlying conformations induced by these AT1R mutants most likely represent principally different mechanisms of uncoupling the G protein, which for some mutants may be due to their increased ability to recruit ß-arrestin2. Hereby, these findings have important implications for drug discovery and 7TMR biology and illustrate the necessity of uncovering the exact molecular determinants for G protein-coupling and ß-arrestin recruitment, respectively.


Subject(s)
Arrestins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Angiotensin II/metabolism , Animals , Arrestins/genetics , Binding, Competitive , COS Cells , Chlorocebus aethiops , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Mutation , Protein Binding , Receptor, Angiotensin, Type 1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , beta-Arrestins
14.
Biochem Pharmacol ; 80(1): 86-94, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20227396

ABSTRACT

Seven transmembrane (7TM) or G protein-coupled receptors constitute a large superfamily of cell surface receptors sharing a structural motif of seven transmembrane spanning alpha helices. Their activation mechanism most likely involves concerted movements of the transmembrane helices, but remains to be completely resolved. Evolutionary Trace (ET) analysis is a computational method, which identifies clusters of functionally important residues by integrating information on evolutionary important residue variations with receptor structure. Combined with known mutational data, ET predicted a patch of residues in the cytoplasmic parts of TM2, TM3, and TM6 to form an activation switch that is common to all family A 7TM receptors. We tested this hypothesis in the rat Angiotensin II (Ang II) type 1a (AT1a) receptor. The receptor has important roles in the cardiovascular system, but has also frequently been applied as a model for 7TM receptor activation and signaling. Six mutations: F66A, L67R, L70R, L119R, D125A, and I245F were targeted to the putative switch and assayed for changes in activation state by their ligand binding, signaling, and trafficking properties. All but one receptor mutant (that was not expressed well) displayed phenotypes associated with changed activation state, such as increased agonist affinity or basal activity, promiscuous activation, or constitutive internalization highlighting the importance of testing different signaling pathways. We conclude that this evolutionary important patch mediates interactions important for maintaining the inactive state. More broadly, these observations in the AT1 receptor are consistent with computational predictions of a generic role for this patch in 7TM receptor activation.


Subject(s)
Biological Evolution , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin II/metabolism , Animals , Cytoplasm/metabolism , Protein Structure, Secondary/genetics , Protein Structure, Tertiary/genetics , Rats , Receptor, Angiotensin, Type 1/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics
15.
Trends Cardiovasc Med ; 20(7): 221-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-22293022

ABSTRACT

Adrenergic and angiotensin receptors are prominent targets in pharmacological alleviation of cardiac remodeling and heart failure, but their use is associated with cardiodepressant side effects. Recent advances in our understanding of seven transmembrane receptor signaling show that it is possible to design ligands with "functional selectivity," acting as agonists on certain signaling pathways while antagonizing others. This represents a major pharmaceutical opportunity to separate desired from adverse effects governed by the same receptor. Accordingly, functionally selective ligands are currently pursued as next-generation drugs for superior treatment of heart failure.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Heart Failure/drug therapy , Receptor, Angiotensin, Type 1/agonists , Animals , Humans , Ligands , Mice , Mice, Transgenic , Receptor, Angiotensin, Type 1/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
16.
J Neurochem ; 112(3): 797-806, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943848

ABSTRACT

The photopigment melanopsin is expressed in a subtype of mammalian ganglion cells in the retina that project to the circadian clock in the hypothalamic suprachiasmatic nucleus to mediate non-visual light information. Melanopsin renders these retinal ganglion cells intrinsically photosensitive and the cells respond to light by a membrane depolarization and induction of the immediate early response gene Fos. Previous studies showed that the light activated melanopsin-induced signaling, the phototransduction, leading to depolarization of the membrane resembles the invertebrate opsins, which involves a Galpha(q/11) coupled phospholipase C activation. However, the signaling proteins mediating melanopsin-induced Fos expression are unresolved. In this study, we examined the phototransduction leading to Fos expression in melanopsin-transfected PC12 cells. A pivotal role of the extracellular signal-regulated protein kinase 1/2 (ERK1/2) was found as pharmacological blockage of this kinase suppressed the light-induced Fos expression. Illumination increased the inositol phosphate turnover and induced phosphorylation of ERK1/2 and p38 but not the c-Jun N-terminal kinase. The Galpha(q/11) protein inhibitor YM254890 attenuated these intracellular light responses. Our data strongly indicate that Galpha(q/11)-mediated ERK1/2 activation is essential for expression of Fos upon illumination of melanopsin-expressing PC12 cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Light , Oncogene Proteins v-fos/metabolism , Rod Opsins/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation/physiology , Inositol Phosphates/metabolism , Oncogene Proteins v-fos/genetics , PC12 Cells/drug effects , PC12 Cells/physiology , PC12 Cells/radiation effects , Phosphorylation/drug effects , Phosphorylation/radiation effects , Rats , Rod Opsins/genetics , Statistics, Nonparametric , Transfection/methods
17.
Neurochem Res ; 34(10): 1729-37, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19495967

ABSTRACT

The PDZ domain mediated interaction between the NMDA receptor and its intracellular scaffolding protein, PSD-95, is a potential target for treatment of ischemic brain diseases. We have recently developed a number of peptide analogues with improved affinity for the PDZ domains of PSD-95 compared to the endogenous C-terminal peptide of the NMDA receptor, as evaluated by a cell-free protein-protein interaction assay. However, it is important to address both membrane permeability and effect in living cells. Therefore a bioluminescence resonance energy transfer (BRET) assay was established, where the C-terminal of the NMDA receptor and PDZ2 of PSD-95 were fused to green fluorescent protein (GFP) and Renilla luciferase (Rluc) and expressed in COS7 cells. A robust and specific BRET signal was obtained by expression of the appropriate partner proteins and subsequently, the assay was used to evaluate a Tat-conjugated peptide for its ability to disrupt the PSD-95/NMDA receptor interaction in living cells.


Subject(s)
Energy Transfer/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Measurements/methods , Membrane Proteins/metabolism , Protein Interaction Domains and Motifs/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Disks Large Homolog 4 Protein , Gene Products, tat/chemistry , Gene Products, tat/physiology , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Luciferases, Renilla/chemistry , Luciferases, Renilla/metabolism , Membrane Proteins/chemistry , Molecular Sequence Data , Protein Binding/physiology , Receptors, N-Methyl-D-Aspartate/chemistry
18.
J Mol Cell Cardiol ; 46(1): 15-24, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18848837

ABSTRACT

The angiotensin AT(1) receptor is an important pharmacological target in the treatment of cardiovascular disorders, such as hypertension, diabetic nephropathy, cardiac hypertrophy, arrhythmia and failure. Simultaneously, the AT(1) receptor has emerged to be a prominent model for the emerging concept that receptors may attain multiple active states with differentiated functional outcomes. Two major signalling pathways are employed by the AT(1) receptor, namely 1) the canonical G(q) protein-dependent activation of inositol phosphate turnover and intracellular calcium release, and 2) G protein-independent recruitment of beta-arrestin-scaffolded signalling complexes that activate protein kinase pathways. Different states of receptor activation with preference for individual downstream pathways (functional selectivity) have been demonstrated in mutational studies of the AT(1) receptor and by pharmacological probing with analogues of angiotensin II. These studies also provide clues about the conformational changes that underlie different functional outcomes. In this review, we evaluate current knowledge of the molecular determinants of AT(1) receptor activation, which may distinguish G protein-dependent and -independent behaviour. While G protein activation is known to be detrimental, G protein-independent signalling by the AT(1) receptor has been associated with phenotypes such as cell survival and renewal, regulation of cardiac contraction and cell migration. It is therefore currently hypothesized that selective blockade of G protein actions and simultaneous activation of G protein-independent signalling will prove to be a feasible strategy for improved cardiovascular therapy. The pharmacological perspectives of functional selectivity by receptors, such as the AT(1) receptor, urge the elucidation of molecular mechanisms that govern disparate signalling events.


Subject(s)
Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/physiology , Amino Acid Sequence , Angiotensin II/metabolism , Animals , Arrestins/metabolism , Calcium/metabolism , Cytoplasm/metabolism , Epitopes/chemistry , Humans , Ligands , Models, Biological , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , beta-Arrestins
19.
Eur J Pharmacol ; 590(1-3): 255-63, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18565507

ABSTRACT

The angiotensin AT(1) receptor is a key regulator of blood pressure and body fluid homeostasis, and it plays a key role in the pathophysiology of several cardiovascular diseases such as hypertension, cardiac hypertrophy, congestive heart failure, and arrhythmia. The importance of human angiotensin AT(1) receptor signalling is illustrated by the common use of angiotensin AT(1) receptor-inverse agonists in clinical practice. It is well established that rodent orthologues of the angiotensin AT(1) receptor can selectively signal through G protein-dependent and -independent mechanisms in recombinant expression systems, primary cells and in vivo. The in vivo work clearly demonstrates profoundly different cellular consequences of angiotensin AT(1) receptor signalling in the cardiovascular system, suggesting pharmacological potential for drugs which specifically affect a subset of angiotensin AT(1) receptor actions. However, it is currently unknown whether the human angiotensin AT(1) receptor can signal through G protein-independent mechanisms - and if so, what the physiological impact of such signalling is. We have performed a detailed pharmacological analysis of the human angiotensin AT(1) receptor using a battery of angiotensin analogues and registered drugs targeting this receptor. We show that the human angiotensin AT(1) receptor signals directly through G protein-independent pathways and supports NIH3T3 cellular proliferation. The realization of G protein-independent signalling by the human angiotensin AT(1) receptor has clear pharmacological implications for development of drugs with pathway-specific actions and defined biological outcomes.


Subject(s)
GTP-Binding Proteins/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, Angiotensin, Type 1/physiology , Animals , COS Cells , Cell Proliferation , Chlorocebus aethiops , Drug Inverse Agonism , Enzyme Activation , Humans , Mice , NIH 3T3 Cells , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/drug effects , Signal Transduction
20.
Trends Cardiovasc Med ; 18(8): 305-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19345318

ABSTRACT

The angiotensin II type 1 (AT(1)) receptor plays a key role in cardiovascular pathophysiology, and it is a major pharmacologic target in the treatment of many cardiovascular disorders. However, AT(1) receptor activation is also involved in adaptive responses to altered hemodynamic demands and to sudden injury occurring in the circulatory system. Hence, current drugs that block all AT(1) receptor actions most likely leave room for improvement. Recent developments show that two major signaling pathways used by the AT(1) receptor may be dissected by pharmacologic means. Key pathologic responses such as aldosterone secretion, vasoconstriction, and detrimental cardiac hypertrophy are known to result from G protein-dependent or -independent signal transduction, whereas mechanisms have been connected with more adaptive cardiac cell survival, migration, and regeneration phenotypes. Selective blockade of G protein actions and simultaneous activation of G protein-dependent or -independent signaling could therefore be desirable in certain situations. The previously unappreciated concept of "functional selectivity" makes this exact strategy feasible and may yield improved drugs for cardiovascular therapy.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Receptor, Angiotensin, Type 1/metabolism , Aldosterone/metabolism , Angiotensin II/metabolism , Cardiomegaly/metabolism , Cardiovascular Diseases/genetics , Feasibility Studies , GTP-Binding Proteins/metabolism , Humans , Receptor, Angiotensin, Type 1/genetics , Renin-Angiotensin System/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...