Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Mol Cell Proteomics ; 9(7): 1540-53, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20363803

ABSTRACT

Seven-transmembrane receptors (7TMRs) signal through the well described heterotrimeric G proteins but can also activate G protein-independent signaling pathways of which the impact and complexity are less understood. The angiotensin II type 1 receptor (AT(1)R) is a prototypical 7TMR and an important drug target in cardiovascular diseases. "Biased agonists" with intrinsic "functional selectivity" that simultaneously blocks Galpha(q) protein activity and activates G protein-independent pathways of the AT(1)R confer important perspectives in treatment of cardiovascular diseases. In this study, we performed a global quantitative phosphoproteomics analysis of the AT(1)R signaling network. We analyzed ligand-stimulated SILAC (stable isotope labeling by amino acids in cell culture) cells by high resolution (LTQ-Orbitrap) MS and compared the phosphoproteomes of the AT(1)R agonist angiotensin II and the biased agonist [Sar(1),Ile(4),Ile(8)]angiotensin II (SII angiotensin II), which only activates the Galpha(q) protein-independent signaling. We quantified more than 10,000 phosphorylation sites of which 1183 were regulated by angiotensin II or its analogue SII angiotensin II. 36% of the AT(1)R-regulated phosphorylations were regulated by SII angiotensin II. Analysis of phosphorylation site patterns showed a striking distinction between protein kinases activated by Galpha(q) protein-dependent and -independent mechanisms, and we now place protein kinase D as a key protein involved in both Galpha(q)-dependent and -independent AT(1)R signaling. This study provides substantial novel insight into angiotensin II signal transduction and is the first study dissecting the differences between a full agonist and a biased agonist from a 7TMR on a systems-wide scale. Importantly, it reveals a previously unappreciated diversity and quantity of Galpha(q) protein-independent signaling and uncovers novel signaling pathways. We foresee that the amount and diversity of G protein-independent signaling may be more pronounced than previously recognized for other 7TMRs as well. Quantitative mass spectrometry is a promising tool for evaluation of the signaling properties of biased agonists to other receptors in the future.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , Phosphoproteins/analysis , Proteome/analysis , Receptor, Angiotensin, Type 1 , Amino Acid Sequence , Cell Line , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Mass Spectrometry/methods , Molecular Sequence Data , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology
7.
Protein Sci ; 19(3): 579-92, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20082307

ABSTRACT

Recombinant expression of native or modified eukaryotic proteins is pivotal for structural and functional studies and for industrial and pharmaceutical production of proteins. However, it is often impeded by the lack of proper folding. Here, we present a stringent and broadly applicable eukaryotic in vivo selection system for folded proteins. It is based on genetic complementation of the Schizosaccharomyces pombe growth marker gene invertase fused C-terminally to a protein library. The fusion proteins are directed to the secretion system, utilizing the ability of the eukaryotic protein quality-control systems to retain misfolded proteins in the ER and redirect them for cytosolic degradation, thereby only allowing folded proteins to reach the cell surface. Accordingly, the folding potential of the tested protein determines the ability of autotrophic colony growth. This system was successfully demonstrated using a complex insertion mutant library of TNF-alpha, from which different folding competent mutant proteins were uncovered.


Subject(s)
Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Selection, Genetic , Sequence Analysis, Protein/methods , Genetic Complementation Test , Humans , Peptide Library , Recombinant Proteins/genetics , Schizosaccharomyces/genetics , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics
8.
FEBS J ; 276(10): 2786-800, 2009 May.
Article in English | MEDLINE | ID: mdl-19459935

ABSTRACT

Residues that mediate helix-helix interactions within the seven transmembranes (TM) of G protein-coupled receptors are important for receptor biogenesis and the receptor switch mechanism. By contrast, the residues directly contacting the lipid bilayer have only recently garnered attention as potential receptor dimerization interfaces. In the present study, we aimed to determine the contributions of these lipid-facing residues to receptor function and oligomerization by systemically generating chimeric complement factor 5a receptors in which the entire lipid-exposed surface of a single TM helix was exchanged with the cognate residues from the angiotensin type 1 receptor. Disulfide-trapping and bioluminescence resonance energy transfer (BRET) studies demonstrated robust homodimerization of both complement factor 5a receptor and angiotensin type 1 receptor, but no evidence for heterodimerization. Despite relatively conservative substitutions, the lipid-facing chimeras (TM1, TM2, TM4, TM5, TM6 or TM7) were retained in the endoplasmic reticulum/cis-Golgi network. With the exception of the TM7 chimera that did not bind ligand, the lipid-facing chimeras bound ligand with low affinity, but similar to wild-type complement factor 5a receptors trapped in the endoplasmic reticulum with brefeldin A. These results suggest that the chimeric receptors were properly folded; moreover, native complement factor 5a receptors are not fully competent to bind ligand when present in the endoplasmic reticulum. BRET oligomerization studies demonstrated energy transfer between the wild-type complement factor 5a receptor and the lipid-facing chimeras, suggesting that the lipid-facing residues within a single TM segment are not essential for oligomerization. These studies highlight the importance of the lipid-facing residues in the complement factor 5a receptor for transport competence.


Subject(s)
Lipid Bilayers , Receptor, Anaphylatoxin C5a/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Animals , Biological Transport , Biopolymers , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Energy Transfer , Humans , Luminescence , Models, Molecular , Molecular Sequence Data , Receptor, Anaphylatoxin C5a/chemistry , Sequence Homology, Amino Acid
9.
J Biol Chem ; 284(3): 1831-9, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19017652

ABSTRACT

It has been suggested previously ( AbdAlla, S., Lother, H., and Quitterer, U. (2000) Nature 407, 94-98 ) that the angiotensin II type 1 receptor (AT1R) and the bradykinin B2 receptor (B2R) form constitutive heterodimers. Furthermore they demonstrate that AT1R signaling significantly increases in the presence of the B2R. These findings suggest that heterodimerization and potentiation of AT1R signaling is a universal phenomenon that occurs as a natural consequence of simultaneous expression of the two receptors. Hence this potential interaction is of great pharmacological and biological interest that adds an additional layer of complexity to the understanding of the cross-talk between the renin-angiotensin and kallikrein-kinin systems. Given the remarkable significance of this finding, scientists from four independent research groups have set out to reproduce and further examine the potential AT1R/B2R interaction. We have investigated functional potentiation by the B2R of AT1R signaling in three different cell lines using multiple assays including phosphoinositide hydrolysis, ERK activation, beta-arrestin recruitment, and receptor selection and amplification technology, and we have examined dimerization using bioluminescence resonance energy transfer and regulated secretion/aggregation technology. However, although both the AT1Rs and B2Rs were functional in our systems and the systems were fine tuned to detect small changes in receptor function, we failed to detect any functional modulation by or physical interaction between the two receptor proteins. In contrast to the previous observations, our data collectively suggest that AT1R/B2R heterodimerization does not occur as a natural consequence of their simultaneous expression in the same cell nor does the B2R influence the AT1R signaling.


Subject(s)
Receptor, Angiotensin, Type 1/metabolism , Receptor, Bradykinin B2/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dimerization , Gene Expression Regulation/physiology , Humans , Kallikrein-Kinin System/physiology , Mice , NIH 3T3 Cells , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Bradykinin B2/genetics , Renin-Angiotensin System/physiology , Signal Transduction/physiology
10.
Mol Cell Endocrinol ; 302(2): 203-12, 2009 Apr 29.
Article in English | MEDLINE | ID: mdl-18930783

ABSTRACT

The Renin-Angiotensin System (RAS) is important for the regulation of cardiovascular physiology, where it controls blood pressure, and salt- and water homeostasis. Dysregulation of RAS can lead to severe diseases including hypertension, diabetic nephropathy, and cardiac arrhythmia, and -failure. The importance of the RAS is clearly emphasised by the widespread use of drugs targeting this system in clinical practice. These include, renin inhibitors, angiotensin II receptor type I blockers, and inhibitors of the angiotensin converting enzyme. Some of the important effectors within the system are 7 transmembrane (7TM) receptors (or G-protein-coupled receptors) such as the angiotensin II Receptors type I and II (AT1R and AT2R) and the MAS-oncogene receptor. Several findings indicate that the 7TM receptors can form both homo- and heterodimers, or higher orders of oligomers. Furthermore, dimerization may be important for receptor function, and in the development of cardiovascular diseases. This is very significant, since "dimers" may provide pharmacologists with novel targets for improved drug therapy. However, we know that 7TM receptors can mediate signals as monomeric units, and so far it has been very difficult to establish if our observations reflect actual well-defined dimerization or merely reflect close proximity between the receptors and/or various types of functional interaction. In this review, we will present and critically discuss the current data on 7TM receptor dimerization with a clear focus on the RAS, and delineate future challenges within the field.


Subject(s)
Receptor Cross-Talk , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System , Humans , Protein Multimerization , Receptors, Angiotensin/metabolism , Receptors, Bradykinin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...