155 results on '"Bünemann M"'
Search Results
2. Temporally resolved cAMP monitoring in endothelial cells uncovers a thrombin-induced [cAMP] elevation mediated via the Ca2+-dependent production of prostacyclin
- Author
-
Werthmann, R. C., Lohse, M. J., and Bünemann, M.
- Published
- 2011
- Full Text
- View/download PDF
3. Real-time monitoring of cAMP levels in living endothelial cells: thrombin transiently inhibits adenylyl cyclase 6
- Author
-
Werthmann, R. C., von Hayn, K., Nikolaev, V. O., Lohse, M. J., and Bünemann, M.
- Published
- 2009
- Full Text
- View/download PDF
4. Conformational changes in G-protein-coupled receptors—the quest for functionally selective conformations is open
- Author
-
Hoffmann, C, Zürn, A, Bünemann, M, and Lohse, M J
- Published
- 2008
- Full Text
- View/download PDF
5. Kinetics of G-protein-coupled receptor signals in intact cells
- Author
-
Lohse, M J, Hein, P, Hoffmann, C, Nikolaev, V O, Vilardaga, J P, and Bünemann, M
- Published
- 2008
- Full Text
- View/download PDF
6. Potentiation of the activation of cholinergic receptors by multivalent presentation of ligands supported on gold nanoparticles
- Author
-
Mattern, A., primary, Machka, F., additional, Wickleder, M. S., additional, Ilyaskina, O. S., additional, Bünemann, M., additional, Diener, M., additional, and Pouokam, E., additional
- Published
- 2018
- Full Text
- View/download PDF
7. Kontinenzverhalten nach Anal- und Rektumatresie und Lebensqualität
- Author
-
Schärli, A. F., Holschneider, A. M., von Kap-herr, S. Hofmann, Koltai, J. L., Herold, A., Bruch, H.-P., Höcht, B., Müller, G., Böhmann, J., Kolb, H., Krahe, Th., Landwehr, P., Schaarschmidt, K., Willital, G. H., Bünemann, M., Jung, K., Hamelmann, H., and Ungeheuer, E., editor
- Published
- 1989
- Full Text
- View/download PDF
8. β-Nicotinamide adenine dinucleotide: a novel bronchodilator inducing increase in intracellular cAMP
- Author
-
Jurastow, I, primary, Engel, S, additional, Wiegand, S, additional, Rafiq, A, additional, Zakrzewicz, A, additional, Grau, V, additional, Nassenstein, C, additional, Bünemann, M, additional, and Kummer, W, additional
- Published
- 2015
- Full Text
- View/download PDF
9. Molecular and functional characterization of PACAP/VIP receptors in Lewis lung carcinoma cells
- Author
-
Kalmbach, N, primary, Krasel, C, additional, Schäfer, MKH, additional, Bünemann, M, additional, and Weihe, E, additional
- Published
- 2012
- Full Text
- View/download PDF
10. Role of Rac 1 and cAMP in endothelial barrier stabilization and thrombin-induced barrier breakdown
- Author
-
Baumer, Y., primary, Spindler, V., additional, Werthmann, R.C., additional, Bünemann, M., additional, and Waschke, J., additional
- Published
- 2009
- Full Text
- View/download PDF
11. Kinetics of G-protein-coupled receptor signalling and desensitization
- Author
-
Krasel, C., primary, Vilardaga, J.-P., additional, Bünemann, M., additional, and Lohse, M.J., additional
- Published
- 2004
- Full Text
- View/download PDF
12. DESENSITIZATION OF G-PROTEIN–COUPLED RECEPTORS IN THE CARDIOVASCULAR SYSTEM
- Author
-
Bünemann, M., primary, Lee, K. B., additional, Pals-Rylaarsdam, R., additional, Roseberry, A. G., additional, and Hosey, M. M., additional
- Published
- 1999
- Full Text
- View/download PDF
13. Inhibition of muscarinic K+ current in guinea-pig atrial myocytes by PD 81,723, an allosteric enhancer of adenosine binding to A1 receptors
- Author
-
Brandts, B, primary, Bünemann, M, additional, Hluchy, J, additional, Sabin, G V, additional, and Pott, L, additional
- Published
- 1997
- Full Text
- View/download PDF
14. A novel membrane receptor with high affinity for lysosphingomyelin and sphingosine 1-phosphate in atrial myocytes.
- Author
-
Bünemann, M., primary, Liliom, K., additional, Brandts, B. K., additional, Pott, L., additional, Tseng, J. L., additional, Desiderio, D. M., additional, Sun, G., additional, Miller, D., additional, and Tigyi, G., additional
- Published
- 1996
- Full Text
- View/download PDF
15. Downregulation of muscarinic M2 receptors linked to K+ current in cultured guinea-pig atrial myocytes.
- Author
-
Bünemann, M, primary, Brandts, B, additional, and Pott, L, additional
- Published
- 1996
- Full Text
- View/download PDF
16. Activation of muscarinic K+ current in guinea-pig atrial myocytes by sphingosine-1-phosphate.
- Author
-
Bünemann, M, primary, Brandts, B, additional, zu Heringdorf, D M, additional, van Koppen, C J, additional, Jakobs, K H, additional, and Pott, L, additional
- Published
- 1995
- Full Text
- View/download PDF
17. Down-regulation of A1 adenosine receptors coupled to muscarinic K+ current in cultured guinea-pig atrial myocytes.
- Author
-
Bünemann, M, primary and Pott, L, additional
- Published
- 1995
- Full Text
- View/download PDF
18. Inhibition of muscarinic K+ current in guinea-pig atrial myocytes by PD 81,723, an allosteric enhancer of adenosine binding to A1 receptors.
- Author
-
Brandts, B, Bünemann, M, Hluchy, J, Sabin, G V, and Pott, L
- Published
- 1997
- Full Text
- View/download PDF
19. Novel inhibition of gbetagamma-activated potassium currents induced by M(2) muscarinic receptors via a pertussis toxin-insensitive pathway.
- Author
-
Bünemann, M, Meyer, T, Pott, L, and Hosey, M
- Abstract
G(i) protein-coupled receptors such as the M(2) muscarinic acetylcholine receptor (mAChR) and A(1) adenosine receptor have been shown to activate G protein-activated inwardly rectifying K(+) channels (GIRKs) via pertussis toxin-sensitive G proteins in atrial myocytes and in many neuronal cells. Here we show that muscarinic M(2) receptors not only activate but also reversibly inhibit these K(+) currents when stimulated with agonist for up to 2 min. The M(2) mAChR-mediated inhibition of the channel was also observed when the channels were first activated by inclusion of guanosine 5'-O-(thiotriphosphate) in the pipette. Under these conditions the M(2) mAChR-induced inhibition was quasi-irreversible, suggesting a role for G proteins in the inhibitory process. In contrast, when GIRK currents were maximally activated by co-expressing exogenous Gbetagamma, the extent of acetylcholine (ACh)-induced inhibition was significantly reduced, suggesting competition between the receptor-mediated inhibition and the large pool of available Gbetagamma subunits. The signaling pathway that led to the ACh-induced inhibition of GIRK channels was unaffected by pertussis toxin pretreatment. Furthermore, the internalization and agonist-induced phosphorylation of M(2) mAChR was not required because a phosphorylation- and internalization-deficient mutant of the M(2) mAChR was as potent as the wild-type counterpart. Pharmacological agents modulating various protein kinases or phosphatidylinositol 3-kinase did not affect the inhibition of GIRK currents. Furthermore, the signaling pathway that mediates GIRK current inhibition was found to be membrane-delimited because bath application of ACh did not inhibit GIRK channel activity in cell-attached patches. Other G protein-coupled receptors including M(4) mAChR and alpha(1A) adrenergic receptors also caused the inhibition, whereas other G protein-coupled receptors including A(1) and A(3) adenosine receptors and alpha(2A) and alpha(2C) adrenergic receptors could not induce the inhibition. The presented results suggest the existence of a novel signaling pathway that can be activated selectively by M(2) and M(4) mAChR but not by adenosine receptors and that involves non-pertussis toxin-sensitive G proteins leading to an inhibition of Gbetagamma-activated GIRK currents in a membrane-delimited fashion.
- Published
- 2000
20. Proteolytic processing of the C terminus of the alpha(1C) subunit of L-type calcium channels and the role of a proline-rich domain in membrane tethering of proteolytic fragments.
- Author
-
Gerhardstein, B L, Gao, T, Bünemann, M, Puri, T S, Adair, A, Ma, H, and Hosey, M M
- Abstract
Although most L-type calcium channel alpha(1C) subunits isolated from heart or brain are approximately 190-kDa proteins that lack approximately 50 kDa of the C terminus, the C-terminal domain is present in intact cells. To test the hypothesis that the C terminus is processed but remains functionally associated with the channels, expressed, full-length alpha(1C) subunits were cleaved in vitro by chymotrypsin to generate a 190-kDa C-terminal truncated protein and C-terminal fragments of 30-56 kDa. These hydrophilic C-terminal fragments remained membrane-associated. A C-terminal proline-rich domain (PRD) was identified as the mediator of membrane association. The alpha(1C) PRD bound to SH3 domains in Src, Lyn, Hck, and the channel beta(2) subunit. Mutant alpha(1C) subunits lacking either approximately 50 kDa of the C terminus or the PRD produced increased barium currents through the channels, demonstrating that these domains participate in the previously described (Wei, X., Neely, a., Lacerda, A. E. Olcese, r., Stefani, E., Perez-Reyes, E., and Birnbaumer, L. (1994) J. Biol. Chem. 269, 1635-1640) inhibition of channel function by the C terminus.
- Published
- 2000
21. Functional regulation of L-type calcium channels via protein kinase A-mediated phosphorylation of the beta(2) subunit.
- Author
-
Bünemann, M, Gerhardstein, B L, Gao, T, and Hosey, M M
- Abstract
Activation of protein kinase A (PKA) through the beta-adrenergic receptor pathway is crucial for the positive regulation of cardiac L-type currents; however it is still unclear which phosphorylation events cause the robust regulation of channel function. In order to study whether or not the recently identified PKA phosphorylation sites on the beta(2) subunit are of functional significance, we coexpressed wild-type (WT) or mutant beta(2) subunits in tsA-201 cells together with an alpha(1C) subunit, alpha(1C)Delta1905, that lacked the C-terminal 265 amino acids, including the only identified PKA site at Ser-1928. This truncated alpha(1C) subunit was similar to the truncated alpha(1C) subunit isolated from cardiac tissue not only in size ( approximately 190 kDa), but also with respect to its failure to serve as a PKA substrate. In cells transfected with the WT beta(2) subunit, voltage-activated Ba(2+) currents were significantly increased when purified PKA was included in the patch pipette. Furthermore, mutations of Ser-478 and Ser-479 to Ala, but not Ser-459 to Ala, on the beta(2) subunit, completely abolished the PKA-induced increase of currents. The data indicate that the PKA-mediated stimulation of cardiac L-type Ca(2+) currents may be at least partially caused by phosphorylation of the beta(2) subunit at Ser-478 and Ser-479.
- Published
- 1999
22. A dominant-negative strategy for studying roles of G proteins in vivo.
- Author
-
Gilchrist, A, Bünemann, M, Li, A, Hosey, M M, and Hamm, H E
- Abstract
G proteins play a critical role in transducing a large variety of signals into intracellular responses. Increasingly, there is evidence that G proteins may play other roles as well. Dominant-negative constructs of the alpha subunit of G proteins would be useful in studying the roles of G proteins in a variety of processes, but the currently available dominant-negative constructs, which target Mg2+-binding sites, are rather leaky. A variety of studies have implicated the carboxyl terminus of G protein alpha subunits in both mediating receptor-G protein interaction and in receptor selectivity. Thus we have made minigene plasmid constructs that encode oligonucleotide sequences corresponding to the carboxyl-terminal undecapeptide of Galphai, Galphaq, or Galphas. To determine whether overexpression of the carboxyl-terminal peptide would block cellular responses, we used as a test system the activation of the M2 muscarinic receptor activated K+ channels in HEK 293 cells. The minigenes were transiently transfected along with G protein-regulated inwardly rectifying K+ channels (GIRK) into HEK 293 cells that stably express the M2 muscarinic receptor. The presence of the Galphai carboxyl-terminal peptide results in specific inhibition of GIRK activity in response to agonist stimulation of the M2 muscarinic receptor. The Galphai minigene construct completely blocks agonist-mediated M2 mAChR K+ channel response whereas the control minigene constructs (empty vector, pcDNA3.1, and the Galpha carboxyl peptide in random order, pcDNA-GalphaiR) had no effect on agonist-mediated M2 muscarinic receptor GIRK response. The inhibitory effects of the Galphai minigene construct were specific because overexpression of peptides corresponding to the carboxyl terminus of Galphaq or Galphas had no effect on M2 muscarinic receptor stimulation of the K+ channel.
- Published
- 1999
23. Inhibition of muscarinic K+current in guinea‐pig atrial myocytes by PD 81,723, an allosteric enhancer of adenosine binding to A1receptors
- Author
-
Brandts, B, Bünemann, M, Hluchy, J, Sabin, G V, and Pott, L
- Abstract
PD 81,723 has been shown to enhance binding of adenosine to A1receptors by stabilizing G protein‐receptor coupling (‘allosteric enhancement’). Evidence has been provided that in the perfused hearts and isolated atria PD 81,723 causes a sensitization to adenosine via this mechanism.We have studied the effect of PD 81,723 in guinea‐pig isolated atrial myocytes by use of whole‐cell measurement of the muscarinic K+current (IK(ACh)) activated by different Gi‐coupled receptors (A1, M2, sphingolipid). PD 81,273 caused inhibition of IK(ACh)(IC50≃5μm) activated by either of the three receptors. Receptor‐independent IK(ACh)in cells loaded with GTP‐γ‐S and background IK(ACh), which contributes to the resting conductance of atrial myocytes, were equally sensitive to PD 81,723. At no combination of concentrations of adenosine and PD 81,723 could an enhancing effect be detected.The compound was active from the outside only. Loading of the cells with PD 81,723 (50μm) via the patch pipette did not affect either IK(ACh)or its sensitivity to adenosine. We suggest that PD 81,723 acts as an inhibitor of inward rectifying K+channels; this is supported by the finding that ventricular IK1, which shares a large degree of homology with the proteins (GIRK1/GIRK4) forming IK(ACh)but is not G protein‐gated, was also blocked by this compound.It is concluded that the functional effects of PD 81,723 described in the literature are not mediated by the A1adenosine receptor‐Gi‐IK(ACh)pathway.
- Published
- 1997
- Full Text
- View/download PDF
24. Activation of a high affinity Gi protein-coupled plasma membrane receptor by sphingosine-1-phosphate.
- Author
-
van Koppen, C, Meyer zu Heringdorf, M, Laser, K T, Zhang, C, Jakobs, K H, Bünemann, M, and Pott, L
- Abstract
Sphingosine-1-phosphate (SPP) has attracted much attention as a possible second messenger controlling cell proliferation and motility and as an intracellular Ca(2+)-releasing agent. Here, we present evidence that SPP activates a G protein-coupled receptor in the plasma membrane of various cells, leading to increase in cytoplasmic Ca2+ concentration ([Ca2+]i), inhibition of adenylyl cyclase, and opening of G protein-regulated potassium channels. In human enbryonic kidney (HEK) cells, SPP potently (EC50, 2 nM) and rapidly increased [Ca2+]i in a pertussis toxin-sensitive manner. Pertussis toxin-sensitive increase in [Ca2+]i was also observed with sphingosylphosphorylcholine (EC50, 460 nM), whereas other sphingolipids, including ceramide-1-phosphate, N-palmitoyl-sphingosine, psychosine, and D-erythro-sphingosine at micromolar concentrations did not or only marginally increased [Ca2+]i. Furthermore, SPP inhibited forskolin-stimulated cAMP accumulation in HEK cells and increased binding of guanosine 5'3-O-(thio) triphosphate to HEK cell membranes. Rapid [Ca2+]i responses were also observed in human transitional bladder carcinoma (J82) cells, monkey COS-1 cells, mouse NIH 3T3 cells, Chinese hamster ovary (CHO-K1) cells, and rat C6 glioma cells, whereas human HL-60 leukemia cells and human erythroleukemia cells failed to respond to SPP. In guinea pig atrial myocytes, SPP activated Gi protein-regulated inwardly rectifying potassium channels. Activation of these channels occurred strictly when SPP was applied at the extracellular face of atrial myocyte plasma membrane as measured in cell-attached and inside-out patch clamp current recordings. We conclude that SPP, in addition to its proposed direct action on intracellular Ca2+ stores, interacts with a high affinity Gi protein-coupled receptor in the plasma membrane of apparently many different cell types.
- Published
- 1996
25. Regulators of G protein signaling (RGS) proteins constitutively activate Gbeta gamma-gated potassium channels.
- Author
-
Bünemann, M and Hosey, M M
- Abstract
Here we report novel effects of regulators of G protein signaling (RGS) on G protein-regulated ion channels. RGS3 and RGS4 induced a substantial increase in currents through the Gbeta gamma-regulated inwardly rectifying K+ channels, IK(ACh), in the absence of receptor activation. Concomitantly, the amount of current that could be activated by agonist was reduced. Pretreatment with pertussis toxin or a muscarinic receptor antagonist abolished agonist-induced currents but did not modify RGS effects. Cotransfection of cells with a Gbetagamma-binding protein significantly reduced the RGS4-induced basal IK(ACh) currents. The RGS proteins also modified the properties of another Gbeta gamma effector, the N-type Ca2+ channels. These observations strongly suggest that RGS proteins increase the availability of Gbeta gamma in addition to their previously described GTPase-activating function.
- Published
- 1998
26. Receptor/G-protein interaction in individual living cells,Rezeptor/G-Protein - Interaktion in einzelnen lebenden Zellen
- Author
-
Peter Hein, Frank, M., Lohse, M. J., and Bünemann, M.
27. Mechanisms of Ewing sarcoma metastasis : biochemistry and biophysics
- Author
-
Beletkaia, E., Schmidt, T., Snaar-Jagalska, B.E., Fenz, S.F., Bünemann, M., Dogterom, M., Eliel, E., Hogendorn, P.C.W., and Leiden University
- Subjects
Mechanisensing ,CXCR4 ,GPCR ,Single-molecule ,Ewing sarcoma ,Metastasis - Abstract
Ewing sarcoma (ES) is a special type of bone cancer, first described by Dr. James Ewing in his paper __Diffusive endothelioma of bone__. Today Ewing sarcoma represents the second most common bone cancer among adolescents and young adults. Contrary to the positive achievement in treatment of localized tumors, the long-term (5-years) survival for Ewing sarcoma patients with metastasis, however, remain below the 30% mark. In this thesis a report on experimental work aiming for a better understanding of the mechanisms underlying Ewing sarcoma metastasis is presented. Two distinct mechanisms are investigated: (1) a biochemical approach in which the initial steps in the CXCR4 signaling cascade are followed, and (2) a biophysical approach in which the guidance of Ewing sarcoma metastasis by the stiffness of their microenvironment is demonstrated. The results presented in this thesis provide deeper insights into the mechanisms controlling signaling of the chemokine receptor CXCR4 and into the role of the micro-environment in Ewing sarcoma cells behavior.Through various experimental approaches it was shown that both biochemical and biophysical guidance control how Ewing sarcoma develops into its distinct metastatic phenotype.
- Published
- 2015
28. The voltage sensitivity of G-protein coupled receptors: Unraveling molecular mechanisms and physiological implications.
- Author
-
Boutonnet M, Bünemann M, and Perroy J
- Abstract
In the landscape of proteins controlled by membrane voltage (V
m ), like voltage-gated ionotropic channels, the emergence of the voltage sensitivity within the vast family of G-protein coupled receptors (GPCRs) marked a significant milestone at the onset of the 21st century. Since its discovery, extensive research has been devoted to understanding the intricate relationship between Vm and GPCRs. Approximately 30 GPCRs out of a family comprising more than 800 receptors have been implicated in Vm -dependent positive and negative regulation. GPCRs stand out as the quintessential regulators of synaptic transmission in neurons, where they encounter substantial variations in Vm . However, the molecular mechanism underlying the Vm sensor of GPCRs remains enigmatic, hindered by the scarcity of mutant GPCRs insensitive to Vm yet functionally intact, impeding a comprehensive understanding of this unique property in physiology. Nevertheless, two decades of dedicated research have furnished numerous insights into the molecular aspects of GPCR Vm -sensing, accompanied by recently proposed physiological roles as well as pharmacological potential, which we encapsulate in this review. The Vm sensitivity of GPCRs emerges as a pivotal attribute, shedding light on previously unforeseen roles in synaptic transmission and extending beyond, underscoring its significance in cellular signaling and physiological processes., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
29. Investigation of muscarinic acetylcholine receptor M3 activation in atomistic detail: a chemist's viewpoint.
- Author
-
Drabek M, Emmerich R, Djulic N, Bünemann M, Schlitzer M, and Kolb P
- Abstract
We analyzed the precise ligand:receptor interactions required for activation of the muscarinic acetylcholine receptor M3, a prototypical G protein-coupled receptor and potential diabetes target. Starting from literature-known compounds and docking solutions, ligands were tailored for the modulation of this receptor's activation. Several aspects of the structure-activity relationship of agonists were investigated in atomistic detail, in order to delineate how the receptor can be activated via the orthosteric site. Such exquisitely precise knowledge is instrumental for designing potent and effiacious ligands. We put this strategy into practice and acquired or synthesized and measured a diverse set of 55 ligands ranging from small fragment-like amines coordinating D3.32 to bigger molecules extending towards helices 5 and 6 with diphenyl moieties. In the course of these investigations, we showed that the polarizability of the amine nitrogen and the rigidity and size of the moieties in the space delimited by helices 5 and 6 are the two key elements distinguishing potent and efficacious ligands from those that are not. The resulting data set will be highly useful in drug design and molecular machine learning alike., (© 2024 Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
30. Arachidonic Acid Directly Activates the Human DP2 Receptor.
- Author
-
Kurz M, Ulrich M, Kirchhofer SB, Bittner A, Daude M, Diederich WE, Pauck K, Garn H, and Bünemann M
- Subjects
- Humans, HEK293 Cells, Prostaglandin D2 metabolism, Prostaglandin D2 pharmacology, Fluorescence Resonance Energy Transfer, Arachidonic Acid metabolism, Arachidonic Acid pharmacology, Receptors, Prostaglandin metabolism, Receptors, Immunologic metabolism
- Abstract
Aberrant type 2 inflammatory responses are the underlying cause of the pathophysiology of allergic asthma, allergic rhinitis, and other atopic diseases, with an alarming prevalence in relevant parts of the Western world. A bulk of evidence points out the important role of the DP2 receptor in these inflammation processes. A screening of different polyunsaturated fatty acids at a fluorescence resonance energy transfer-based DP2 receptor conformation sensor expressed in human embryonic kidney (HEK) cells revealed an agonistic effect of the prostaglandin (PG)-D
2 precursor arachidonic acid on DP2 receptor activity of about 80% of the effect induced by PGD2 In a combination of experiments at the conformation sensor and using a bioluminescence resonance energy transfer-based G protein activation sensor expressed together with DP2 receptor wild type in HEK cells, we found that arachidonic acid acts as a direct activator of the DP2 receptor, but not the DP1 receptor, in a concentration range considered physiologically relevant. Pharmacological inhibition of cyclooxygenases and lipoxygenases as well as cytochrome P450 did not lead to a diminished arachidonic acid response on the DP2 receptor, confirming a direct action of arachidonic acid on the receptor. SIGNIFICANCE STATEMENT: This study identified the prostaglandin precursor arachidonic acid to directly activate the DP2 receptor, a G protein-coupled receptor that is known to play an important role in type 2 inflammation., (Copyright © 2024 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2024
- Full Text
- View/download PDF
31. Opioid receptors: single molecule studies shed light on mechanisms of efficacy.
- Author
-
Krasel C and Bünemann M
- Published
- 2024
- Full Text
- View/download PDF
32. Orphan GPCR GPRC5C Facilitates Angiotensin II-Induced Smooth Muscle Contraction.
- Author
-
Wang T, Shao J, Kumar S, Alnouri MW, Carvalho J, Günther S, Krasel C, Murphy KT, Bünemann M, Offermanns S, and Wettschureck N
- Subjects
- Animals, Humans, Male, Mice, Cells, Cultured, Hypertension metabolism, Hypertension physiopathology, Hypertension chemically induced, Hypertension genetics, Mesenteric Arteries metabolism, Mice, Inbred C57BL, Mice, Knockout, Muscle Contraction, Myocytes, Smooth Muscle metabolism, Vasoconstriction, Angiotensin II pharmacology, Muscle, Smooth, Vascular metabolism, Receptors, G-Protein-Coupled metabolism, Receptors, G-Protein-Coupled genetics
- Abstract
Background: GPCRs (G-protein-coupled receptors) play a central role in the regulation of smooth muscle cell (SMC) contractility, but the function of SMC-expressed orphan GPCR class C group 5 member C (GPRC5C) is unclear. The aim of this project is to define the role of GPRC5C in SMC in vitro and in vivo., Methods: We studied the role of GPRC5C in the regulation of SMC contractility and differentiation in human and murine SMC in vitro, as well as in tamoxifen-inducible, SMC-specific GPRC5C knockout mice under basal conditions and in vascular disease in vivo., Results: Mesenteric arteries from tamoxifen-inducible, SMC-specific GPRC5C knockout mice showed ex vivo significantly reduced angiotensin II (Ang II)-dependent calcium mobilization and contraction, whereas responses to other relaxant or contractile factors were normal. In vitro, the knockdown of GPRC5C in human aortic SMC resulted in diminished Ang II-dependent inositol phosphate production and lower myosin light chain phosphorylation. In line with this, tamoxifen-inducible, SMC-specific GPRC5C knockout mice showed reduced Ang II-induced arterial hypertension, and acute inactivation of GPRC5C was able to ameliorate established arterial hypertension. Mechanistically, we show that GPRC5C and the Ang II receptor AT1 dimerize, and knockdown of GPRC5C resulted in reduced binding of Ang II to AT1 receptors in HEK293 cells, human and murine SMC, and arteries from tamoxifen-inducible, SMC-specific GPRC5C knockout mice., Conclusions: Our data show that GPRC5C regulates Ang II-dependent vascular contraction by facilitating AT1 receptor-ligand binding and signaling., Competing Interests: Disclosures None.
- Published
- 2024
- Full Text
- View/download PDF
33. DP2 receptor activity sensor suited for antagonist screening and measurement of receptor dynamics in real-time.
- Author
-
Kurz M, Ulrich M, Bittner A, and Bünemann M
- Subjects
- Humans, Receptors, Prostaglandin, Prostaglandin D2 pharmacology, Inflammation
- Abstract
The DP2 receptor is a G-protein coupled receptor involved in allergic inflammation and is the target of recently developed antagonists already being tested in clinics. To get insights into DP2 receptor dynamics and to study its pharmacology on the level of the receptor, we constructed a fluorescence resonance energy transfer-based conformation sensor. The sensor reflects the selectivity profile of the DP2 receptor-wt and is suited for screening of agonists and antagonists due to its robust response. Furthermore, the sensor enables the direct measurement of DP2 receptor dynamics in real-time and revealed markedly distinct on- and off-rates of prostaglandin D
2 between DP2 and DP1 receptors, suggesting a different mechanism of ligand receptor interaction., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
34. Differential interaction patterns of opioid analgesics with µ opioid receptors correlate with ligand-specific voltage sensitivity.
- Author
-
Kirchhofer SB, Lim VJY, Ernst S, Karsai N, Ruland JG, Canals M, Kolb P, and Bünemann M
- Subjects
- Humans, Ligands, Receptors, Opioid, mu metabolism, Pain, Analgesics, Opioid pharmacology, Receptors, Opioid
- Abstract
The µ opioid receptor (MOR) is the key target for analgesia, but the application of opioids is accompanied by several issues. There is a wide range of opioid analgesics, differing in their chemical structure and their properties of receptor activation and subsequent effects. A better understanding of ligand-receptor interactions and the resulting effects is important. Here, we calculated the respective binding poses for several opioids and analyzed interaction fingerprints between ligand and receptor. We further corroborated the interactions experimentally by cellular assays. As MOR was observed to display ligand-induced modulation of activity due to changes in membrane potential, we further analyzed the effects of voltage sensitivity on this receptor. Combining in silico and in vitro approaches, we defined discriminating interaction patterns responsible for ligand-specific voltage sensitivity and present new insights into their specific effects on activation of the MOR., Competing Interests: SK, VL, SE, NK, JR, MC, PK, MB No competing interests declared, (© 2023, Kirchhofer et al.)
- Published
- 2023
- Full Text
- View/download PDF
35. EP4 Receptor Conformation Sensor Suited for Ligand Screening and Imaging of Extracellular Prostaglandins.
- Author
-
Kurz M, Ulrich M, Bittner A, Scharf MM, Shao J, Wallenstein I, Lemoine H, Wettschureck N, Kolb P, and Bünemann M
- Subjects
- Mice, Animals, Dogs, Humans, Ligands, Receptors, Prostaglandin E, EP4 Subtype metabolism, Receptors, Prostaglandin, Receptors, Prostaglandin E, EP2 Subtype metabolism, Prostaglandins, Dinoprostone metabolism
- Abstract
Prostaglandins are important lipid mediators with a wide range of functions in the human body. They act mainly via plasma membrane localized prostaglandin receptors, which belong to the G-protein coupled receptor class. Due to their localized formation and short lifetime, it is important to be able to measure the distribution and abundance of prostaglandins in time and/or space. In this study, we present a Foerster resonance energy transfer (FRET)-based conformation sensor of the human prostaglandin E receptor subtype 4 (EP4 receptor), which was capable of detecting prostaglandin E
2 (PGE2 )-induced receptor activation in the low nanomolar range with a good signal-to-noise ratio. The sensor retained the typical selectivity for PGE2 among arachidonic acid products. Human embryonic kidney cells stably expressing the sensor did not produce detectable amounts of prostaglandins making them suitable for a coculture approach allowing us, over time, to detect prostaglandin formation in Madin-Darby canine kidney cells and primary mouse macrophages. Furthermore, the EP4 receptor sensor proved to be suited to detect experimentally generated PGE2 gradients by means of FRET-microscopy, indicating the potential to measure gradients of PGE2 within tissues. In addition to FRET-based imaging of prostanoid release, the sensor allowed not only for determination of PGE2 concentrations, but also proved to be capable of measuring ligand binding kinetics. The good signal-to-noise ratio at a commercial plate reader and the ability to directly determine ligand efficacy shows the obvious potential of this sensor interest for screening and characterization of novel ligands of the pharmacologically important human EP4 receptor. SIGNIFICANCE STATEMENT: The authors present a biosensor based on the prostaglandin E receptor subtype 4, which is well suited to measure extracellular prostaglandin E2 (PGE2 ) concentration with high temporal and spatial resolution. It can be used for the imaging of PGE2 levels and gradients by means of Foerster resonance energy transfer microscopy, and for determining PGE2 release of primary cells as well as for screening purposes in a plate reader setting., (Copyright © 2023 by The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
36. Interaction kinetics between p115-RhoGEF and Gα 13 are determined by unique molecular interactions affecting agonist sensitivity.
- Author
-
Redlin F, Krett AL, and Bünemann M
- Subjects
- Rho Guanine Nucleotide Exchange Factors genetics, Kinetics, Amino Acids, Receptors, G-Protein-Coupled, Signal Transduction
- Abstract
The three RH-RhoGEFs (Guanine nucleotide exchange factors) p115-RhoGEF, LARG (leukemia-associated RhoGEF) and PDZ-RhoGEF link G-protein coupled receptors (GPCRs) with RhoA signaling through activation of Gα
12/13 . In order to find functional differences in signaling between the different RH-RhoGEFs we examined their interaction with Gα13 in high spatial and temporal resolution, utilizing a FRET-based single cell assay. We found that p115-RhoGEF interacts significantly shorter with Gα13 than LARG and PDZ-RhoGEF, while narrowing the structural basis for these differences down to a single amino acid in the rgRGS domain of p115-RhoGEF. The mutation of this amino acid led to an increased interaction time with Gα13 and an enhanced agonist sensitivity, comparable to LARG, while mutating the corresponding amino acid in Gα13 the same effect could be achieved. While the rgRGS domains of RH-RhoGEFs showed GAP (GTPase-activating protein) activity towards Gα13 in vitro, our approach suggests higher GAP activity of p115-RhoGEF in intact cells., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
37. Differential effects of glucose-dependent insulinotropic polypeptide receptor/glucagon-like peptide-1 receptor heteromerization on cell signaling when expressed in HEK-293 cells.
- Author
-
Al-Zaid B, Chacko S, Ezeamuzie CI, Bünemann M, Krasel C, Karimian T, Lanzerstorfer P, and Al-Sabah S
- Subjects
- Arrestins metabolism, Gastric Inhibitory Polypeptide metabolism, Glucose pharmacology, HEK293 Cells, Humans, Incretins, Ligands, Peptides, Receptors, G-Protein-Coupled metabolism, Secretin metabolism, Signal Transduction, Glucagon-Like Peptide-1 Receptor metabolism, Receptors, Gastrointestinal Hormone metabolism
- Abstract
The incretin hormones: glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are important regulators of many aspects of metabolism including insulin secretion. Their receptors (GIPR and GLP-1R) are closely related members of the secretin class of G-protein-coupled receptors. As both receptors are expressed on pancreatic β-cells there is at least the hypothetical possibility that they may form heteromers. In the present study, we investigated GIPR/GLP-1R heteromerization and the impact of GIPR on GLP-1R-mediated signaling and vice versa in HEK-293 cells. Real-time fluorescence resonance energy transfer (FRET) and bioluminescence resonance energy transfer (BRET) saturation experiments confirm that GLP-1R and GIPR form heteromers. Stimulation with 1 μM GLP-1 caused an increase in both FRET and BRET ratio, whereas stimulation with 1 μM GIP caused a decrease. The only other ligand tested to cause a significant change in BRET signal was the GLP-1 metabolite, GLP-1 (9-36). GIPR expression had no significant effect on mini-G
s recruitment to GLP-1R but significantly inhibited GLP-1 stimulated mini-Gq and arrestin recruitment. In contrast, the presence of GLP-1R improved GIP stimulated mini-Gs and mini-Gq recruitment to GIPR. These data support the hypothesis that GIPR and GLP-1R form heteromers with differential consequences on cell signaling., (© 2022 The Authors. Pharmacology Research & Perspectives published by British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics and John Wiley & Sons Ltd.)- Published
- 2022
- Full Text
- View/download PDF
38. Residency time of agonists does not affect the stability of GPCR-arrestin complexes.
- Author
-
Mösslein N, Pohle LG, Fuss A, Bünemann M, and Krasel C
- Subjects
- Arrestins, Receptors, G-Protein-Coupled metabolism, beta-Arrestin 2, beta-Arrestins, Arrestin, Internship and Residency
- Abstract
Background and Purpose: The interaction of arrestins with G-protein coupled receptors (GPCRs) desensitizes agonist-dependent receptor responses and often leads to receptor internalization. GPCRs that internalize without arrestin have been classified as "class A" GPCRs whereas "class B" GPCRs co-internalize with arrestin into endosomes. The interaction of arrestins with GPCRs requires both agonist activation and receptor phosphorylation. Here, we ask the question whether agonists with very slow off-rates can cause the formation of particularly stable receptor-arrestin complexes., Experimental Approach: The stability of GPCR-arrestin-3 complexes at two class A GPCRs, the β
2 -adrenoceptor and the μ opioid receptor, was assessed using two different techniques, fluorescence resonance energy transfer (FRET) and fluorescence recovery after photobleaching (FRAP) employing several ligands with very different off-rates. Arrestin trafficking was determined by confocal microscopy., Key Results: Upon agonist washout, GPCR-arrestin-3 complexes showed markedly different dissociation rates in single-cell FRET experiments. In FRAP experiments, however, all full agonists led to the formation of receptor-arrestin complexes of identical stability whereas the complex between the μ receptor and arrestin-3 induced by the partial agonist morphine was less stable. Agonists with very slow off-rates could not mediate the co-internalization of arrestin-3 with class A GPCRs into endosomes., Conclusions and Implications: Agonist off-rates do not affect the stability of GPCR-arrestin complexes but phosphorylation patterns do. Our results imply that orthosteric agonists are not able to pharmacologically convert class A into class B GPCRs., (© 2022 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)- Published
- 2022
- Full Text
- View/download PDF
39. The pocketome of G-protein-coupled receptors reveals previously untargeted allosteric sites.
- Author
-
Hedderich JB, Persechino M, Becker K, Heydenreich FM, Gutermuth T, Bouvier M, Bünemann M, and Kolb P
- Subjects
- Allosteric Regulation physiology, Allosteric Site physiology, Binding Sites, Ligands, Receptors, G-Protein-Coupled chemistry, Receptors, Muscarinic metabolism
- Abstract
G-protein-coupled receptors do not only feature the orthosteric pockets, where most endogenous agonists bind, but also a multitude of other allosteric pockets that have come into the focus as potential binding sites for synthetic modulators. Here, to better characterise such pockets, we investigate 557 GPCR structures by exhaustively docking small molecular probes in silico and converting the ensemble of binding locations to pocket-defining volumes. Our analysis confirms all previously identified pockets and reveals nine previously untargeted sites. In order to test for the feasibility of functional modulation of receptors through binding of a ligand to such sites, we mutate residues in two sites, in two model receptors, the muscarinic acetylcholine receptor M
3 and β2 -adrenergic receptor. Moreover, we analyse the correlation of inter-residue contacts with the activation states of receptors and show that contact patterns closely correlating with activation indeed coincide with these sites., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
40. Disentangling bias between G q , GRK2, and arrestin3 recruitment to the M 3 muscarinic acetylcholine receptor.
- Author
-
Flöser A, Becker K, Kostenis E, König G, Krasel C, Kolb P, and Bünemann M
- Subjects
- Animals, CHO Cells, Cricetulus, HEK293 Cells, Humans, G-Protein-Coupled Receptor Kinase 2 metabolism, Receptors, G-Protein-Coupled metabolism, Receptors, Muscarinic physiology, beta-Arrestin 2 metabolism
- Abstract
G protein-coupled receptors (GPCRs) transmit extracellular signals to the inside by activation of intracellular effector proteins. Different agonists can promote differential receptor-induced signaling responses - termed bias - potentially by eliciting different levels of recruitment of effector proteins. As activation and recruitment of effector proteins might influence each other, thorough analysis of bias is difficult. Here, we compared the efficacy of seven agonists to induce G protein, G protein-coupled receptor kinase 2 (GRK2), as well as arrestin3 binding to the muscarinic acetylcholine receptor M
3 by utilizing FRET-based assays. In order to avoid interference between these interactions, we studied GRK2 binding in the presence of inhibitors of Gi and Gq proteins and analyzed arrestin3 binding to prestimulated M3 receptors to avoid differences in receptor phosphorylation influencing arrestin recruitment. We measured substantial differences in the agonist efficacies to induce M3 R-arrestin3 versus M3 R-GRK2 interaction. However, the rank order of the agonists for G protein- and GRK2-M3 R interaction was the same, suggesting that G protein and GRK2 binding to M3 R requires similar receptor conformations, whereas requirements for arrestin3 binding to M3 R are distinct., Competing Interests: AF, KB, EK, GK, CK, PK, MB No competing interests declared, (© 2021, Floeser et al.)- Published
- 2021
- Full Text
- View/download PDF
41. Structures in G proteins important for subtype selective receptor binding and subsequent activation.
- Author
-
Jelinek V, Mösslein N, and Bünemann M
- Subjects
- Animals, GTP-Binding Protein alpha Subunits physiology, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 physiology, GTP-Binding Protein alpha Subunits, Gq-G11 ultrastructure, GTP-Binding Proteins metabolism, GTP-Binding Proteins physiology, GTP-Binding Proteins ultrastructure, Humans, Mice, Protein Binding, Rats, Receptors, G-Protein-Coupled physiology, Receptors, G-Protein-Coupled ultrastructure, Signal Transduction, GTP-Binding Protein alpha Subunits metabolism, GTP-Binding Protein alpha Subunits ultrastructure, Receptors, G-Protein-Coupled metabolism
- Abstract
G protein-coupled receptors (GPCRs) selectively couple to specific heterotrimeric G proteins comprised of four subfamilies in order to induce appropriate physiological responses. However, structural determinants in Gα subunits responsible for selective recognition by approximately 800 human GPCRs have remained elusive. Here, we directly compare the influence of subtype-specific Gα structures on the stability of GPCR-G protein complexes and the activation by two Gq-coupled receptors. We used FRET-assays designed to distinguish multiple Go and Gq-based Gα chimeras in their ability to be selectively bound and activated by muscarinic M
3 and histaminic H1 receptors. We identify the N-terminus including the αN/β1-hinge, the β2/β3-loop and the α5 helix of Gα to be key selectivity determinants which differ in their impact on selective binding to GPCRs and subsequent activation depending on the specific receptor. Altogether, these findings provide new insights into the molecular basis of G protein-coupling selectivity even beyond the Gα C-terminus.- Published
- 2021
- Full Text
- View/download PDF
42. Pharmacological Characterization of the Stick Insect Carausius morosus Allatostatin-C Receptor with Its Endogenous Agonist.
- Author
-
Işbilir A, Duan Sahbaz B, Tuncgenc G, Bünemann M, Lohse MJ, and Birgül-Iyison N
- Abstract
G protein-coupled receptors (GPCRs) play a pivotal role in regulating key physiological events in all animal species. Recent advances in collective analysis of genes and proteins revealed numerous potential neuropeptides and GPCRs from insect species, allowing for the characterization of peptide-receptor pairs. In this work, we used fluorescence resonance energy transfer (FRET)-based genetically encoded biosensors in intact mammalian cells to study the pharmacological features of the cognate GPCR of the type-C allatostatin (AST-C) peptide from the stick insect, Carausius morosus . Analysis of multiple downstream pathways revealed that AST-C can activate the human Gi
2 protein, and not Gs or Gq, through AST-C receptor (AlstRC). Activated AlstRC recruits β-arrestin2 independent of the Gi protein but stimulates ERK phosphorylation in a Gi protein-dependent manner. Identification of Gαi-, arrestin-, and GRK-like transcripts from C. morosus revealed high evolutionary conservation at the G protein level, while β-arrestins and GRKs displayed less conservation. In conclusion, our study provides experimental and homology-based evidence on the functionality of vertebrate G proteins and downstream signaling biosensors to characterize early signaling steps of an insect GPCR. These results may serve as a scaffold for developing assays to characterize pharmacological and structural aspects of other insect GPCRs and can be used in deorphanization and pesticide studies., Competing Interests: The authors declare no competing financial interest., (© 2020 American Chemical Society.)- Published
- 2020
- Full Text
- View/download PDF
43. The Effect of Cell Surface Expression and Linker Sequence on the Recruitment of Arrestin to the GIP Receptor.
- Author
-
Al-Sabah S, Adi L, Bünemann M, and Krasel C
- Abstract
The glucose-dependent insulinotropic polypeptide (GIP) and the glucagon-like peptide-1 (GLP-1) receptor are important targets in the treatment of both type 2 diabetes mellitus (T2DM) and obesity. Originally identified for their role in desensitization, internalization and recycling of G protein-coupled receptors (GPCRs), arrestins have since been shown to act as scaffolding proteins that allow GPCRs to signal in a G protein-independent manner. While GLP-1R has been reported to interact with arrestins, this aspect of cell signaling remains controversial for GIPR. Using a (FRET)-based assay we have previously shown that yellow fluorescent protein (YFP)-labeled GIPR does not recruit arrestin. This GIPR-YFP construct contained a 10 amino acid linker between the receptor and a XbaI restriction site upstream of the YFP. This linker was not present in the modified GIPR-SYFP2 used in subsequent FRET and bioluminescence resonance energy transfer (BRET) assays. However, its removal results in the introduction of a serine residue adjacent to the end of GIPR's C-terminal tail which could potentially be a phosphorylation site. The resulting receptor was indeed able to recruit arrestin. To find out whether the serine/arginine (SR) coded by the XbaI site was indeed the source of the problem, it was substituted with glycine/glycine (GG) by site-directed mutagenesis. This substitution abolished arrestin recruitment in the BRET assay but only significantly reduced it in the FRET assay. In addition, we show that the presence of a N-terminal FLAG epitope and influenza hemagglutinin signal peptide were also required to detect arrestin recruitment to the GIPR, most likely by increasing receptor cell surface expression. These results demonstrate how arrestin recruitment assay configuration can dramatically alter the result. This becomes relevant when drug discovery programs aim to identify ligands with "biased agonist" properties., (Copyright © 2020 Al-Sabah, Adi, Bünemann and Krasel.)
- Published
- 2020
- Full Text
- View/download PDF
44. Voltage modulates the effect of μ-receptor activation in a ligand-dependent manner.
- Author
-
Ruland JG, Kirchhofer SB, Klindert S, Bailey CP, and Bünemann M
- Subjects
- Animals, Enkephalin, Ala(2)-MePhe(4)-Gly(5)-, HEK293 Cells, Humans, Ligands, Morphine pharmacology, Rats, Locus Coeruleus metabolism, Receptors, Opioid, mu metabolism
- Abstract
Background and Purpose: Various GPCRs have been described as being modulated in a voltage-dependent manner. Opioid analgesics act via activation of μ receptors in various neurons. As neurons are exposed to large changes in membrane potential, we were interested in studying the effects of depolarization on μ receptor signalling., Experimental Approach: We investigated potential voltage sensitivity of μ receptors in heterologous expression systems (HEK293T cells) using electrophysiology in combination with Förster resonance energy transfer-based assays. Depolarization-induced changes in signalling were also tested in physiological rat tissue containing locus coeruleus neurons. We applied depolarization steps across the physiological range of membrane potentials., Key Results: Studying μ receptor function and signalling in cells, we discovered that morphine-induced signalling was strongly dependent on the membrane potential (V
M ). This became apparent at the level of G-protein activation, G-protein coupled inwardly rectifying potassium channel (Kir 3.X) currents and binding of GPCR kinases and arrestin3 to μ receptors by a robust increase in signalling upon membrane depolarization. The pronounced voltage sensitivity of morphine-induced μ receptor activation was also observed at the level of Kir 3.X currents in rat locus coeruleus neurons. The efficacy of peptide ligands to activate μ receptors was not (Met-enkephalin) or only moderately ([D-Ala2 , N-Me-Phe4 , Gly5 -ol]-enkephalin) enhanced upon depolarization. In contrast, depolarization reduced the ability of the analgesic fentanyl to activate μ receptors., Conclusion and Implications: Our results indicate a strong ligand-dependent modulation of μ receptor activity by the membrane potential, suggesting preferential activity of morphine in neurons with high neuronal activity., (© 2020 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)- Published
- 2020
- Full Text
- View/download PDF
45. Beliefs about immunosuppressant medication and correlates in a German kidney transplant population.
- Author
-
Bünemann M, Bauer-Hohmann M, Klewitz F, Kyaw Tha Tun EM, Tegtbur U, Pape L, Schiffer L, Schiffer M, de Zwaan M, and Nöhre M
- Subjects
- Cross-Sectional Studies, Female, Germany, Humans, Male, Middle Aged, Surveys and Questionnaires, Immunosuppressive Agents therapeutic use, Kidney Transplantation psychology, Medication Adherence psychology
- Abstract
Objective: A common reason for organ rejection after transplantation is the lack of adherence regarding immunosuppressive medication (ISM). A variety of different aspects can promote non-adherent behavior, including the relationship between perceived benefits and concerns regarding ISM ("necessity-concerns-framework"). Little is known about the variables associated with this framework., Methods: As part of this cross-sectional study, 570 patients after kidney transplantation who participated in a structured multimodal follow-up program (KTx360°) were examined in two transplant centers in Lower Saxony. We used the Beliefs about Medicines Questionnaire (BMQ) to evaluate the patients' believes and concerns regarding their ISM., Results: The mean age of the participants was 51.9 (SD 14.17) years, 58.4% were men, and 25.8% had ≥12 years of school attendance. The mean time since transplantation was 65.9 months. In patients undergoing kidney transplantation, the perceived benefit of ISM mostly exceeded the concerns. We found an association between lower perceived benefits and greater concerns and lower adherence. Also, a higher perceived necessity was significantly associated with higher age and lower levels of depression and anxiety. Greater concerns were significantly associated with more symptoms of depression and anxiety, lower perceived social support, and lower kidney functioning (eGFR)., Conclusion: Even though patients after kidney transplantation usually acknowledge the importance of their ISM, they still have considerable concerns that are associated with less adherence and various psychosocial risk factors. Further longitudinal studies are needed to assess the extent to which beliefs about medication are variable and can be individually addressed to improve adherence., Competing Interests: Declaration of Competing Interest None., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
46. Orphan G Protein-Coupled Receptor GPRC5B Controls Smooth Muscle Contractility and Differentiation by Inhibiting Prostacyclin Receptor Signaling.
- Author
-
Carvalho J, Chennupati R, Li R, Günther S, Kaur H, Zhao W, Tonack S, Kurz M, Mößlein N, Bünemann M, Offermanns S, and Wettschureck N
- Subjects
- Animals, Cell Differentiation, Humans, Mice, Signal Transduction, Epoprostenol metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Background: G protein-coupled receptors are important regulators of contractility and differentiation in vascular smooth muscle cells (SMCs), but the specific function of SMC-expressed orphan G protein-coupled receptor class C group 5 member B (GPRC5B) is unclear., Methods: We studied the role of GPRC5B in the regulation of contractility and dedifferentiation in human and murine SMCs in vitro and in iSM- Gprc5b -KO (tamoxifen-inducible, SMC-specific knockout) mice under conditions of arterial hypertension and atherosclerosis in vivo., Results: Mesenteric arteries from SMC-specific Gprc5b -KOs showed ex vivo significantly enhanced prostacyclin receptor (IP)-dependent relaxation, whereas responses to other relaxant or contractile factors were normal. In vitro, knockdown of GPRC5B in human aortic SMCs resulted in increased IP-dependent cAMP production and consecutive facilitation of SMC relaxation. In line with this facilitation of IP-mediated relaxation, iSM- Gprc5b -KO mice were protected from arterial hypertension, and this protective effect was abrogated by IP antagonists. Mechanistically, we show that knockdown of GPRC5B increased the membrane localization of IP both in vitro and in vivo and that GPRC5B, but not other G protein-coupled receptors, physically interacts with IP. Last, we show that enhanced IP signaling in GPRC5B-deficient SMCs not only facilitates relaxation but also prevents dedifferentiation during atherosclerosis development, resulting in reduced plaque load and increased differentiation of SMCs in the fibrous cap., Conclusions: Taken together, our data show that GPRC5B regulates vascular SMC tone and differentiation by negatively regulating IP signaling.
- Published
- 2020
- Full Text
- View/download PDF
47. Voltage Dependence of Prostanoid Receptors.
- Author
-
Kurz M, Krett AL, and Bünemann M
- Subjects
- Arginine genetics, Binding Sites genetics, Cell Membrane drug effects, Cell Membrane metabolism, Fluorescence Resonance Energy Transfer, HEK293 Cells, Humans, Iloprost pharmacology, Ligands, Mutagenesis, Site-Directed, Patch-Clamp Techniques, Receptors, Epoprostenol metabolism, Receptors, Prostaglandin genetics, Recombinant Proteins genetics, Recombinant Proteins metabolism, Transfection, 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Receptors, Prostaglandin metabolism, Signal Transduction drug effects
- Abstract
G protein-coupled receptors (GPCRs) are the largest class of transmembrane receptors and serve as signal mediators to transduce information from extracellular signals such as neurotransmitters, hormones, or drugs to cellular responses. They are exposed to the strong electrical field of the plasma membrane. In the last decade voltage modulation of ligand-induced GPCR activity has been reported for several GPCRs. Using Foerster resonance energy transfer-based biosensors in patch clamp experiments, we discovered a robust voltage dependence of the thromboxane receptor (TP receptor) on the receptor level as well as on downstream signaling. TP receptor activity doubled upon depolarization from -90 to +60 mV in the presence of U46619, a stable analog of prostaglandin H
2 Half-maximal effective potential (V0.5 ) determined for TP receptor was -46 mV, which is within the physiologic range. We identified that depolarization affected the agonist affinity for the TP receptor. Depolarization enhanced responses of several structural analogs of U46619 with modifications to a similar extent all around the molecule, indicating that voltage modulates the general conformation of TP receptor. By means of site direct mutagenesis, we identified TP receptor R2957.40 , which showed alteration of voltage sensitivity of TP receptor upon mutation. Voltage sensitivity was not limited to TP receptor because prostaglandin F receptor activated with U46619 and prostaglandin E2 receptor subtype 3 activated with iloprost showed a similar reaction to depolarization as TP receptor. However, prostacyclin receptor activated with iloprost showed no detectable voltage dependence. SIGNIFICANCE STATEMENT: Prostanoids mediate many of their physiological effects via transmembrane receptors expressed in the plasma membrane of excitable cells. We found that agonist-mediated activation of prostaglandin F receptors and prostaglandin E2 receptors as well as thromboxane receptors are activated upon depolarization, whereas prostacyclin receptors are not. The voltage-induced modulation of thromboxane receptor activity was observed on the level of receptor conformation and downstream signaling. The range of voltage dependence was restricted by R2957.40 in the agonist-binding pocket., (Copyright © 2020 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2020
- Full Text
- View/download PDF
48. The N-termini of GRK2 and GRK3 simulate the stimulating effects of RKIP on β-adrenoceptors.
- Author
-
Maimari T, Krasel C, Bünemann M, and Lorenz K
- Subjects
- Animals, Binding Sites, Cells, Cultured, G-Protein-Coupled Receptor Kinase 2 genetics, G-Protein-Coupled Receptor Kinase 3 genetics, G-Protein-Coupled Receptor Kinase 5 metabolism, HEK293 Cells, Humans, Mice, Inbred Strains, Myocytes, Cardiac, Peptide Fragments genetics, Peptide Fragments metabolism, Phosphatidylethanolamine Binding Protein metabolism, Phosphorylation, Receptors, Adrenergic, beta-2 genetics, G-Protein-Coupled Receptor Kinase 2 metabolism, G-Protein-Coupled Receptor Kinase 3 metabolism, Receptors, Adrenergic, beta-2 metabolism
- Abstract
The Raf kinase inhibitor protein (RKIP) activates β-adrenoceptors (β-AR) and thereby induces a well-tolerated cardiac contractility and prevents heart failure in mice. Different to RKIP-mediated β-AR activation, chronic activation of β-AR by catecholamines was shown to be detrimental for the heart. RKIP is an endogenous inhibitor of G protein coupled receptor kinase 2 (GRK2); it binds GRK2 and thereby inhibits GRK2 mediated β-AR phosphorylation and desensitization. Here, we evaluate RKIP-mediated effects on β-AR to explore new strategies for β-AR modulation. Co-immunoprecipitation assays and pull-down assays revealed subtype specificity of RKIP for the cardiac GRK isoforms GRK2 and GRK3 - not GRK5 - as well as several RKIP binding sites within their N-termini (GRK2
1-185 and GRK31-185 ). Overexpression of these N-termini prevented β2 -AR phosphorylation and internalization, subsequently increased receptor signaling in HEK293 cells and cardiomyocyte contractility. Co-immunoprecipitation assays of β2 -AR with these N-terminal GRK fragments revealed a direct interaction suggesting a steric interference of the fragments with the functional GRK-receptor interaction. Altogether, N-termini of GRK2 and GRK3 efficiently simulate RKIP effects on β-AR signaling in HEK293 cells and in cardiomyocytes by their binding to β2 -AR and, thus, provide important insights for the development of new strategies to modulate β2 -AR signaling., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
49. Comparative Docking to Distinct G Protein-Coupled Receptor Conformations Exclusively Yields Ligands with Agonist Efficacy.
- Author
-
Scharf MM, Bünemann M, Baker JG, and Kolb P
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Dose-Response Relationship, Drug, HEK293 Cells, Humans, Ligands, Protein Conformation, Protein Structure, Secondary, Receptors, G-Protein-Coupled agonists, Adrenergic beta-2 Receptor Agonists metabolism, Adrenergic beta-2 Receptor Agonists pharmacology, Molecular Docking Simulation methods, Receptors, Adrenergic, beta-2 chemistry, Receptors, Adrenergic, beta-2 metabolism
- Abstract
G protein-coupled receptors exist in a whole spectrum of conformations that are stabilized by the binding of ligands with different efficacy or intracellular effector proteins. Here, we investigate whether three-dimensional structures of receptor conformations in different states of activation can be used to enrich ligands with agonist behavior in prospective docking calculations. We focused on the β
2 -adrenergic receptor, as it is currently the receptor with the highest number of active-state crystal structures. Comparative docking calculations to distinct conformations of the receptor were used for the in silico prediction of ligands with agonist efficacy. The pharmacology of molecules selected based on these predictions was characterized experimentally, resulting in a hit rate of 37% ligands, all of which were agonists. The ligands furthermore contain a pyrazole moiety that has previously not been described for β2 -adrenergic receptor ligands, and one of them shows an intrinsic efficacy comparable to salbutamol. SIGNIFICANCE STATEMENT: Structure-based ligand design for G protein-coupled receptors crucially depends on receptor conformation and, hence, their activation state. We explored the influence of using multiple active-conformation X-ray structures on the hit rate of docking calculations to find novel agonists, and how to predict the most fruitful strategy to apply. The results suggest that aggregating the ranks of molecules across docking calculations to more than one active-state structure exclusively yields agonists., (Copyright © 2019 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2019
- Full Text
- View/download PDF
50. Direct targeting of Gα q and Gα 11 oncoproteins in cancer cells.
- Author
-
Annala S, Feng X, Shridhar N, Eryilmaz F, Patt J, Yang J, Pfeil EM, Cervantes-Villagrana RD, Inoue A, Häberlein F, Slodczyk T, Reher R, Kehraus S, Monteleone S, Schrage R, Heycke N, Rick U, Engel S, Pfeifer A, Kolb P, König G, Bünemann M, Tüting T, Vázquez-Prado J, Gutkind JS, Gaffal E, and Kostenis E
- Subjects
- Animals, Cell Line, Tumor, Depsipeptides chemistry, HEK293 Cells, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Xenograft Model Antitumor Assays, Depsipeptides pharmacology, Drug Delivery Systems, GTP-Binding Protein alpha Subunits antagonists & inhibitors, GTP-Binding Protein alpha Subunits genetics, GTP-Binding Protein alpha Subunits metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 antagonists & inhibitors, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Gain of Function Mutation, Melanoma drug therapy, Melanoma enzymology, Melanoma genetics, Melanoma pathology, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Uveal Neoplasms drug therapy, Uveal Neoplasms enzymology, Uveal Neoplasms genetics, Uveal Neoplasms pathology
- Abstract
Somatic gain-of-function mutations of GNAQ and GNA11 , which encode α subunits of heterotrimeric Gα
q/11 proteins, occur in about 85% of cases of uveal melanoma (UM), the most common cancer of the adult eye. Molecular therapies to directly target these oncoproteins are lacking, and current treatment options rely on radiation, surgery, or inhibition of effector molecules downstream of these G proteins. A hallmark feature of oncogenic Gαq/11 proteins is their reduced intrinsic rate of hydrolysis of guanosine triphosphate (GTP), which results in their accumulation in the GTP-bound, active state. Here, we report that the cyclic depsipeptide FR900359 (FR) directly interacted with GTPase-deficient Gαq/11 proteins and preferentially inhibited mitogenic ERK signaling rather than canonical phospholipase Cβ (PLCβ) signaling driven by these oncogenes. Thereby, FR suppressed the proliferation of melanoma cells in culture and inhibited the growth of Gαq -driven UM mouse xenografts in vivo. In contrast, FR did not affect tumor growth when xenografts carried mutated B-RafV600E as the oncogenic driver. Because FR enabled suppression of malignant traits in cancer cells that are driven by activating mutations at codon 209 in Gαq/11 proteins, we envision that similar approaches could be taken to blunt the signaling of non-Gαq/11 G proteins., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.