Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ramona Schrage is active.

Publication


Featured researches published by Ramona Schrage.


Nature Communications | 2015

The experimental power of FR900359 to study Gq-regulated biological processes

Ramona Schrage; Anna-Lena Schmitz; Evelyn Gaffal; Suvi Annala; Stefan Kehraus; Daniela Wenzel; Katrin M. Büllesbach; Tobias Bald; Asuka Inoue; Yuji Shinjo; Ségolène Galandrin; Naveen Shridhar; Michael Hesse; Manuel Grundmann; Nicole Merten; Thomas H. Charpentier; Matthew Martz; Adrian J. Butcher; Tanja Slodczyk; Sylvain Armando; Maike Effern; Yoon Namkung; Laura Jenkins; Velten Horn; Anne Stößel; Harald Dargatz; Daniel Tietze; Diana Imhof; Céline Galés; Christel Drewke

Despite the discovery of heterotrimeric αβγ G proteins ∼25 years ago, their selective perturbation by cell-permeable inhibitors remains a fundamental challenge. Here we report that the plant-derived depsipeptide FR900359 (FR) is ideally suited to this task. Using a multifaceted approach we systematically characterize FR as a selective inhibitor of Gq/11/14 over all other mammalian Gα isoforms and elaborate its molecular mechanism of action. We also use FR to investigate whether inhibition of Gq proteins is an effective post-receptor strategy to target oncogenic signalling, using melanoma as a model system. FR suppresses many of the hallmark features that are central to the malignancy of melanoma cells, thereby providing new opportunities for therapeutic intervention. Just as pertussis toxin is used extensively to probe and inhibit the signalling of Gi/o proteins, we anticipate that FR will at least be its equivalent for investigating the biological relevance of Gq.


Science Signaling | 2013

Decoding Signaling and Function of the Orphan G Protein–Coupled Receptor GPR17 with a Small-Molecule Agonist

Stephanie Hennen; Haibo Wang; Lucas Peters; Nicole Merten; Katharina Simon; Andreas Spinrath; Stefanie Blättermann; Rhalid Akkari; Ramona Schrage; Ralf Schröder; Daniel Schulz; Céline Vermeiren; Katrin Zimmermann; Stefan Kehraus; Christel Drewke; Alexander Pfeifer; Gabriele M. König; Klaus Mohr; Michel Gillard; Christa E. Müller; Q. Richard Lu; Jesus Gomeza; Evi Kostenis

Activation of GPR17 prevents oligodendrocyte maturation and reveals that inhibiting GPR17 may be a therapeutic strategy to treat multiple sclerosis. Overcoming a Myelination Maturity Block Demyelinating diseases, such as multiple sclerosis (MS), are characterized by the failure of oligodendrocytes to mature and produce myelin, the protective sheaths surrounding axons. The role of the orphan G protein (heterotrimeric guanine nucleotide–binding protein)–coupled receptor GPR17 in this process is debated. Hennen et al. identified a GPR17-selective small-molecule agonist and showed that application of this agonist induced G protein–mediated signaling that prevented maturation of cultured oligodendrocytes. The findings establish an inhibitory role for GPR17 in the cellular maturation process that enables remyelination of injured axons and suggest that GPR17 may be pharmacologically targeted to treat MS. Replacement of the lost myelin sheath is a therapeutic goal for treating demyelinating diseases of the central nervous system (CNS), such as multiple sclerosis (MS). The G protein (heterotrimeric guanine nucleotide–binding protein)–coupled receptor (GPCR) GPR17, which is phylogenetically closely related to receptors of the “purinergic cluster,” has emerged as a modulator of CNS myelination. However, whether GPR17-mediated signaling positively or negatively regulates this critical process is unresolved. We identified a small-molecule agonist, MDL29,951, that selectively activated GPR17 even in a complex environment of endogenous purinergic receptors in primary oligodendrocytes. MDL29,951-stimulated GPR17 engaged the entire set of intracellular adaptor proteins for GPCRs: G proteins of the Gαi, Gαs, and Gαq subfamily, as well as β-arrestins. This was visualized as alterations in the concentrations of cyclic adenosine monophosphate and inositol phosphate, increased Ca2+ flux, phosphorylation of extracellular signal–regulated kinases 1 and 2 (ERK1/2), as well as multifeatured cell activation recorded with label-free dynamic mass redistribution and impedance biosensors. MDL29,951 inhibited the maturation of primary oligodendrocytes from heterozygous but not GPR17 knockout mice in culture, as well as in cerebellar slices from 4-day-old wild-type mice. Because GPCRs are attractive targets for therapeutic intervention, inhibiting GPR17 emerges as therapeutic strategy to relieve the oligodendrocyte maturation block and promote myelin repair in MS.


British Journal of Pharmacology | 2013

Agonists with supraphysiological efficacy at the muscarinic M2 ACh receptor

Ramona Schrage; Wk Seemann; Jessica Klöckner; Clelia Dallanoce; Kurt Racké; Evi Kostenis; M. De Amici; Ulrike Holzgrabe; Klaus Mohr

Artificial agonists may have higher efficacy for receptor activation than the physiological agonist. Until now, such ‘superagonism’ has rarely been reported for GPCRs. Iperoxo is an extremely potent muscarinic receptor agonist. We hypothesized that iperoxo is a ‘superagonist’.


Journal of Medicinal Chemistry | 2014

Dualsteric Muscarinic Antagonists–Orthosteric Binding Pose Controls Allosteric Subtype Selectivity

Jens Schmitz; Dorina van der Mey; Marcel Bermudez; Jessica Klöckner; Ramona Schrage; Evi Kostenis; Christian Tränkle; Gerhard Wolber; Klaus Mohr; Ulrike Holzgrabe

Bivalent ligands of G protein-coupled receptors have been shown to simultaneously either bind to two adjacent receptors or to bridge different parts of one receptor protein. Recently, we found that bivalent agonists of muscarinic receptors can simultaneously occupy both the orthosteric transmitter binding site and the allosteric vestibule of the receptor protein. Such dualsteric agonists display a certain extent of subtype selectivity, generate pathway-specific signaling, and in addition may allow for designed partial agonism. Here, we want to extend the concept to bivalent antagonism. Using the phthal- and naphthalimide moieties, which bind to the allosteric, extracellular site, and atropine or scopolamine as orthosteric building blocks, both connected by a hexamethonium linker, we were able to prove a bitopic binding mode of antagonist hybrids for the first time. This is demonstrated by structure-activity relationships, site-directed mutagenesis, molecular docking studies, and molecular dynamics simulations. Findings revealed that a difference in spatial orientation of the orthosteric tropane moiety translates into a divergent M2/M5 subtype selectivity of the corresponding bitopic hybrids.


Journal of Medicinal Chemistry | 2015

Rational design of partial agonists for the muscarinic m1 acetylcholine receptor.

Xinyu Chen; Jessika Klöckner; Janine Holze; Cornelia Zimmermann; Wk Seemann; Ramona Schrage; Andreas Bock; Klaus Mohr; Christian Tränkle; Ulrike Holzgrabe; Michael W. Decker

Aiming to design partial agonists for a G-protein-coupled receptor based on dynamic ligand binding, we synthesized three different series of bipharmacophoric ligands composed of the orthosteric building blocks iperoxo and 1 linked to allosteric modulators (BQCA-derived compounds, BQCAd; TBPB-derived compound, TBPBd). Their interactions were studied with the human muscarinic acetylcholine M1-receptor (hM1) with respect to receptor binding and Gq-protein signaling. Results demonstrate that iperoxo/BQCAd (2, 3) and 1/BQCAd hybrids (4) act as M1 partial agonists, whereas 1/TBPBd hybrids (5) did not activate M1-receptors. Among the iperoxo/BQCAd-hybrids, spacer length in conjunction with the pattern of substitution tuned efficacy. Most interestingly, a model of dynamic ligand binding revealed that the spacer length of 2a and 3a controlled the probability of switch between the inactive purely allosteric and the active bitopic orthosteric/allosteric binding pose. In summary, dynamic ligand binding can be exploited in M1 receptors to design partial agonists with graded efficacy.


Current Opinion in Pharmacology | 2017

Functional selectivity and dualsteric/bitopic GPCR targeting

Ramona Schrage; Evi Kostenis

HIGHLIGHTSGPCR‐linked signaling is pleiotropic and involves multiple intracellular pathways.Biased ligands favor particular signaling pathways at the expense of others.Functional selectivity can avoid non‐desired on‐target effects.Dualsteric/bitopic ligands restrain conformational flexibility of GPCRs.Dualsteric/bitopic compounds are ideally suited to induce functional selectivity. &NA; Functional selectivity provides a new avenue to selectively engage particular pathways of the pleiotropic signaling repertoire of a G protein‐coupled receptor. First examples for signaling biased compounds at the angiotensin II receptor and the &mgr; opioid receptor have progressed to clinical trials and are promising in regard to selective activation of signaling pathways that can be linked to beneficial clinical outcomes. Dualsteric/bitopic hybrid compounds which consist of at least two pharmacophores combined in one single ligand are more recent examples for functionally selective ligands. Their binding topography makes them ideally suited to disrupt receptor flexibility and rationally induce signaling bias. Therefore, the dualsteric/bitopic design principle is most promising to facilitate generation of structurally diverse biased agonists at G protein‐coupled receptors.


Biochemical Pharmacology | 2014

New insight into active muscarinic receptors with the novel radioagonist [3H]iperoxo

Ramona Schrage; Janine Holze; Jessica Klöckner; Aileen Balkow; Anne S. Klause; Anna-Lena Schmitz; Marco De Amici; Evi Kostenis; Christian Tränkle; Ulrike Holzgrabe; Klaus Mohr

Activation of G protein-coupled receptors involves major conformational changes of the receptor protein ranging from the extracellular transmitter binding site to the intracellular G protein binding surface. GPCRs such as the muscarinic acetylcholine receptors are commonly probed with radioantagonists rather than radioagonists due to better physicochemical stability, higher affinity, and indifference towards receptor coupling states of the former. Here we introduce tritiated iperoxo, a superagonist at muscarinic M₂ receptors with very high affinity. In membrane suspensions of transfected CHO-cells, [³H]iperoxo - unlike the common radioagonists [³H]acetylcholine and [³H]oxotremorine M - allowed labelling of each of the five muscarinic receptor subtypes in radioagonist displacement and saturation binding studies. [³H]iperoxo revealed considerable differences in affinity between the even- and the odd-numbered muscarinic receptor subtypes with affinities for the M₂ and M₄ receptor in the picomolar range. Probing ternary complex formation on the M₂ receptor, [³H]iperoxo dissociation was not influenced by an archetypal allosteric inverse agonist, reflecting activation-related rearrangement of the extracellular loop region. At the inner side of M₂, the preferred Gi protein acted as a positive allosteric modulator of [³H]iperoxo binding, whereas Gs and Gq were neutral in spite of their robust coupling to the activated receptor. In intact CHO-hM₂ cells, endogenous guanylnucleotides promoted receptor/G protein-dissociation resulting in low-affinity agonist binding which, nevertheless, was still reported by [³H]iperoxo. Taken together, the muscarinic superagonist [³H]iperoxo is the best tool currently available for direct probing activation-related conformational transitions of muscarinic receptors.


Molecular Pharmacology | 2017

The Orphan Receptor GPR17 Is Unresponsive to Uracil Nucleotides and Cysteinyl Leukotrienes

Katharina Simon; Nicole Merten; Ralf Schröder; Stephanie Hennen; Philip Preis; Nina-Katharina Schmitt; Lucas Peters; Ramona Schrage; Céline Vermeiren; Michel Gillard; Klaus Mohr; Jesus Gomeza; Evi Kostenis

Pairing orphan G protein–coupled receptors (GPCRs) with their cognate endogenous ligands is expected to have a major impact on our understanding of GPCR biology. It follows that the reproducibility of orphan receptor ligand pairs should be of fundamental importance to guide meaningful investigations into the pharmacology and function of individual receptors. GPR17 is an orphan receptor characterized by some as a dualistic uracil nucleotide/cysteinyl leukotriene receptor and by others as inactive toward these stimuli altogether. Whereas regulation of central nervous system myelination by GPR17 is well established, verification of activity of its putative endogenous ligands has proven elusive so far. Herein we report that uracil nucleotides and cysteinyl leukotrienes do not activate human, mouse, or rat GPR17 in various cellular backgrounds, including primary cells, using eight distinct functional assay platforms based on label-free pathway-unbiased biosensor technologies, as well as canonical second-messenger or biochemical assays. Appraisal of GPR17 activity can be accomplished with neither the coapplication of both ligand classes nor the exogenous transfection of partner receptors nucleotide purinergic G protein–coupled receptor, cysteinyl leukotriene 1, to reconstitute the elusive pharmacology. Moreover, our study does not support the inhibition of GPR17 by the marketed antiplatelet drugs cangrelor and ticagrelor, previously suggested to antagonize GPR17. Whereas our data do not disagree with a role of GPR17 per se as an orchestrator of central nervous system functions, they challenge the utility of the proposed (ant)agonists as tools to imply direct contribution of GPR17 in complex biologic settings.


Molecular Pharmacology | 2017

A new molecular mechanism to engineer protean agonism at a G protein-coupled receptor

Anna De Min; Carlo Matera; Andreas Bock; Janine Holze; Jessica Kloeckner; Mathias Muth; Christian Traenkle; Marco De Amici; Terry P. Kenakin; Ulrike Holzgrabe; Clelia Dallanoce; Evi Kostenis; Klaus Mohr; Ramona Schrage

Protean agonists are of great pharmacological interest as their behavior may change in magnitude and direction depending on the constitutive activity of a receptor. Yet, this intriguing phenomenon has been poorly described and understood, due to the lack of stable experimental systems and design strategies. In this study, we overcome both limitations: First, we demonstrate that modulation of the ionic strength in a defined experimental set-up allows for analysis of G protein–coupled receptor activation in the absence and presence of a specific amount of spontaneous receptor activity using the muscarinic M2 acetylcholine receptor as a model. Second, we employ this assay system to show that a dualsteric design principle, that is, molecular probes, carrying two pharmacophores to simultaneously adopt orthosteric and allosteric topography within a G protein–coupled receptor, may represent a novel approach to achieve protean agonism. We pinpoint three molecular requirements within dualsteric compounds that elicit protean agonism at the muscarinic M2 acetylcholine receptor. Using radioligand-binding and functional assays, we posit that dynamic ligand binding may be the mechanism underlying protean agonism of dualsteric ligands. Our findings provide both new mechanistic insights into the still enigmatic phenomenon of protean agonism and a rationale for the design of such compounds for a G protein–coupled receptor.


Journal of Pharmacology and Experimental Therapeutics | 2017

Engineered Context-Sensitive Agonism: Tissue-Selective Drug Signaling through a G Protein-Coupled Receptor

Wk Seemann; Daniela Wenzel; Ramona Schrage; Justine Etscheid; Theresa Bödefeld; Anna Bartol; Mareille Warnken; Philipp Sasse; Jessica Klöckner; Ulrike Holzgrabe; Marco DeAmici; Eberhard Schlicker; Kurt Racké; Evi Kostenis; Rainer Meyer; Bernd K. Fleischmann; Klaus Mohr

Drug discovery strives for selective ligands to achieve targeted modulation of tissue function. Here we introduce engineered context-sensitive agonism as a postreceptor mechanism for tissue-selective drug action through a G protein-coupled receptor. Acetylcholine M2-receptor activation is known to mediate, among other actions, potentially dangerous slowing of the heart rate. This unwanted side effect is one of the main reasons that limit clinical application of muscarinic agonists. Herein we show that dualsteric (orthosteric/allosteric) agonists induce less cardiac depression ex vivo and in vivo than conventional full agonists. Exploration of the underlying mechanism in living cells employing cellular dynamic mass redistribution identified context-sensitive agonism of these dualsteric agonists. They translate elevation of intracellular cAMP into a switch from full to partial agonism. Designed context-sensitive agonism opens an avenue toward postreceptor pharmacologic selectivity, which even works in target tissues operated by the same subtype of pharmacologic receptor.

Collaboration


Dive into the Ramona Schrage's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andreas Bock

University of Würzburg

View shared research outputs
Researchain Logo
Decentralizing Knowledge