Network


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

Hotspot


Dive into the research topics where Ralf Schröder is active.

Publication


Featured researches published by Ralf Schröder.


British Journal of Pharmacology | 2010

GPR55 ligands promote receptor coupling to multiple signalling pathways

Christopher M. Henstridge; Nariman Ab Balenga; Ralf Schröder; Julia Kargl; Wolfgang Platzer; Lene Martini; Simon Arthur; June Penman; Jennifer L. Whistler; Evi Kostenis; Maria Waldhoer; Andrew J. Irving

Background and purpose:  Although GPR55 is potently activated by the endogenous lysophospholipid, L‐α‐lysophosphatidylinositol (LPI), it is also thought to be sensitive to a number of cannabinoid ligands, including the prototypic CB1 receptor antagonists AM251 and SR141716A (Rimonabant®). In this study we have used a range of functional assays to compare the pharmacological activity of selected cannabinoid ligands, AM251, AM281 and SR141716A with LPI in a HEK293 cell line engineered to stably express recombinant, human GPR55.


Cell Research | 2011

GPR55 regulates cannabinoid 2 receptor-mediated responses in human neutrophils

Nariman Balenga; Elma Aflaki; Julia Kargl; Wolfgang Platzer; Ralf Schröder; Stefanie Blättermann; Evi Kostenis; Andrew J. Brown; Akos Heinemann; Maria Waldhoer

The directional migration of neutrophils towards inflammatory mediators, such as chemokines and cannabinoids, occurs via the activation of seven transmembrane G protein coupled receptors (7TM/GPCRs) and is a highly organized process. A crucial role for controlling neutrophil migration has been ascribed to the cannabinoid CB2 receptor (CB2R), but additional modulatory sites distinct from CB2R have recently been suggested to impact CB2R-mediated effector functions in neutrophils. Here, we provide evidence that the recently de-orphanized 7TM/GPCR GPR55 potently modulates CB2R-mediated responses. We show that GPR55 is expressed in human blood neutrophils and its activation augments the migratory response towards the CB2R agonist 2-arachidonoylglycerol (2-AG), while inhibiting neutrophil degranulation and reactive oxygen species (ROS) production. Using HEK293 and HL60 cell lines, along with primary neutrophils, we show that GPR55 and CB2R interfere with each others signaling pathways at the level of small GTPases, such as Rac2 and Cdc42. This ultimately leads to cellular polarization and efficient migration as well as abrogation of degranulation and ROS formation in neutrophils. Therefore, GPR55 limits the tissue-injuring inflammatory responses mediated by CB2R, while it synergizes with CB2R in recruiting neutrophils to sites of inflammation.


Nature Protocols | 2011

Applying label-free dynamic mass redistribution technology to frame signaling of G protein-coupled receptors noninvasively in living cells

Ralf Schröder; Johannes Schmidt; Stefanie Blättermann; Lucas Peters; Nicole Janssen; Manuel Grundmann; Wk Seemann; Dorina Kaufel; Nicole Merten; Christel Drewke; Jesus Gomeza; Graeme Milligan; Klaus Mohr; Evi Kostenis

Label-free dynamic mass redistribution (DMR) is a cutting-edge assay technology that enables real-time detection of integrated cellular responses in living cells. It relies on detection of refractive index alterations on biosensor-coated microplates that originate from stimulus-induced changes in the total biomass proximal to the sensor surface. Here we describe a detailed protocol to apply DMR technology to frame functional behavior of G protein–coupled receptors that are traditionally examined with end point assays on the basis of detection of individual second messengers, such as cAMP, Ca2+ or inositol phosphates. The method can be readily adapted across diverse cellular backgrounds (adherent or suspension), including primary human cells. Real-time recordings can be performed in 384-well microtiter plates and be completed in 2 h, or they can be extended to several hours depending on the biological question to be addressed. The entire procedure, including cell harvesting and DMR detection, takes 1–2 d.


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.


Journal of Biological Chemistry | 2009

The C-terminal Tail of CRTH2 Is a Key Molecular Determinant That Constrains Gαi and Downstream Signaling Cascade Activation

Ralf Schröder; Nicole Merten; Jesper Mosolff Mathiesen; Lene Martini; Anamarija Kruljac-Letunic; Friederike Krop; Andree Blaukat; Ye Fang; Elizabeth J. Tran; Trond Ulven; Christel Drewke; Jennifer L. Whistler; Leonardo Pardo; Jesus Gomeza; Evi Kostenis

Prostaglandin D2 activation of the seven-transmembrane receptor CRTH2 regulates numerous cell functions that are important in inflammatory diseases, such as asthma. Despite its disease implication, no studies to date aimed at identifying receptor domains governing signaling and surface expression of human CRTH2. We tested the hypothesis that CRTH2 may take advantage of its C-tail to silence its own signaling and that this mechanism may explain the poor functional responses observed with CRTH2 in heterologous expression systems. Although the C terminus is a critical determinant for retention of CRTH2 at the plasma membrane, the presence of this domain confers a signaling-compromised conformation onto the receptor. Indeed, a mutant receptor lacking the major portion of its C-terminal tail displays paradoxically enhanced Gαi and ERK1/2 activation despite enhanced constitutive and agonist-mediated internalization. Enhanced activation of Gαi proteins and downstream signaling cascades is probably due to the inability of the tail-truncated receptor to recruit β-arrestin2 and undergo homologous desensitization. Unexpectedly, CRTH2 is not phosphorylated upon agonist-stimulation, a primary mechanism by which GPCR activity is regulated. Dynamic mass redistribution assays, which allow label-free monitoring of all major G protein pathways in real time, confirm that the C terminus inhibits Gαi signaling of CRTH2 but does not encode G protein specificity determinants. We propose that intrinsic CRTH2 inhibition by its C terminus may represent a rather unappreciated strategy employed by a GPCR to specify the extent of G protein activation and that this mechanism may compensate for the absence of the classical phosphorylation-dependent signal attenuation.


Nature Chemical Biology | 2012

A biased ligand for OXE-R uncouples Gα and Gβγ signaling within a heterotrimer

Stefanie Blättermann; Lucas Peters; Philipp A. Ottersbach; Andreas Bock; Viktoria Konya; C. David Weaver; Angel Gonzalez; Ralf Schröder; Rahul Tyagi; Petra Luschnig; Jürgen Gäb; Stephanie Hennen; Trond Ulven; Leonardo Pardo; Klaus Mohr; Michael Gütschow; Akos Heinemann; Evi Kostenis

Differential targeting of heterotrimeric G protein versus β-arrestin signaling are emerging concepts in G protein-coupled receptor (GPCR) research and drug discovery, and biased engagement by GPCR ligands of either β-arrestin or G protein pathways has been disclosed. Herein we report on a new mechanism of ligand bias to titrate the signaling specificity of a cell-surface GPCR. Using a combination of biomolecular and virtual screening, we identified the small-molecule modulator Gue1654, which inhibits Gβγ but not Gα signaling triggered upon activation of Gα(i)-βγ by the chemoattractant receptor OXE-R in both recombinant and human primary cells. Gue1654 does not interfere nonspecifically with signaling directly at or downstream of Gβγ. This hitherto unappreciated mechanism of ligand bias at a GPCR highlights both a new paradigm for functional selectivity and a potentially new strategy to develop pathway-specific therapeutics.


British Journal of Pharmacology | 2014

Heteromerization of GPR55 and cannabinoid CB2 receptors modulates signalling

Nariman Ab Balenga; E Martínez-Pinilla; Julia Kargl; Ralf Schröder; Miriam Peinhaupt; Wolfgang Platzer; Zoltán Bálint; M Zamarbide; I G Dopeso-Reyes; A Ricobaraza; J M Pérez-Ortiz; Evi Kostenis; Maria Waldhoer; Akos Heinemann; Rafael Franco

Heteromerization of GPCRs is key to the integration of extracellular signals and the subsequent cell response via several mechanisms including heteromer‐selective ligand binding, trafficking and/or downstream signalling. As the lysophosphatidylinositol GPCR 55 (GPR55) has been shown to affect the function of the cannabinoid receptor subtype 2 (CB2 receptor) in human neutrophils, we investigated the possible heteromerization of CB2 receptors with GPR55.


Journal of Medicinal Chemistry | 2012

Free Fatty Acid Receptor 1 (FFA1/GPR40) Agonists: Mesylpropoxy Appendage Lowers Lipophilicity and Improves ADME Properties

Elisabeth Christiansen; Maria E. Due-Hansen; Christian Urban; Manuel Grundmann; Ralf Schröder; Brian D. Hudson; Graeme Milligan; Michael A. Cawthorne; Evi Kostenis; Matthias U. Kassack; Trond Ulven

FFA1 (GPR40) is a new target for treatment of type 2 diabetes. We recently identified the potent FFA1 agonist TUG-469 (5). Inspired by the structurally related TAK-875, we explored the effects of a mesylpropoxy appendage on 5. The appendage significantly lowers lipophilicity and improves metabolic stability while preserving potency, resulting in discovery of the potent FFA1 agonist 13.


The Journal of Allergy and Clinical Immunology | 2012

D-type prostanoid receptor enhances the signaling of chemoattractant receptor–homologous molecule expressed on TH2 cells

Miriam Sedej; Ralf Schröder; Kathrin Bell; Wolfgang Platzer; Anela Vukoja; Evi Kostenis; Akos Heinemann; Maria Waldhoer

BACKGROUND Prostaglandin (PG) D(2) is substantially involved in allergic responses and signals through the 7 transmembrane-spanning/G protein-coupled receptors, chemoattractant receptor-homologous molecule expressed on T(H)2 cells (CRTH2), and D-type prostanoid (DP) receptor. OBJECTIVE Although the proinflammatory function of CRTH2 is well recognized and CRTH2 is hence considered an important emerging pharmacotherapeutic target, the role of the DP receptor in mediating the biological effects of PGD(2) in patients with allergic inflammation has remained unclear. METHODS The cross-talk of CRTH2 and DP receptors was investigated by using both a recombinant HEK293 cell model and human eosinophils in Ca(2+) mobilization assays, coimmunoprecipitation, Western blotting, radioligand binding, and immunofluorescence. RESULTS We show that CRTH2 and DP receptors modulate one anothers signaling properties and form CRTH2/DP heteromers without altering their ligand-binding capacities. We find that the DP receptor amplifies the CRTH2-induced Ca(2+) release from intracellular stores and coincidentally forfeits its own signaling potency. Moreover, desensitization or pharmacologic blockade of the DP receptor hinders CRTH2-mediated signal transduction. However, CRTH2 internalization occurs independently of the DP receptor. In cells that express both receptors, pharmacologic blockade of Gα(q/11) proteins abolishes the Ca(2+) response to both CRTH2 and DP agonists, whereas inhibition of Gα(i) proteins selectively attenuates the CRTH2-mediated response but not the DP signal. CONCLUSION Our data demonstrate the capacity of DP receptors to amplify the biological response to CRTH2 activation. Therefore the CRTH2/DP heteromer might not only represent a functional signaling unit for PGD(2) but also a potential target for the development of heteromer-directed therapies to treat allergic diseases.


Science Translational Medicine | 2017

Targeted inhibition of Gq signaling induces airway relaxation in mouse models of asthma

Michaela Matthey; Richard E. Roberts; Alexander Seidinger; Annika Simon; Ralf Schröder; Markus Kuschak; Suvi Annala; Gabriele M. König; Christa E. Müller; Ian P. Hall; Evi Kostenis; Bernd K. Fleischmann; Daniela Wenzel

Local pharmacological inhibition of Gq results in airway relaxation in mouse models of asthma. Breathing freely Obstructive lung diseases are common disorders characterized by airway narrowing. Because some patients do not respond well to current therapies or suffer from side effects, new drugs are needed. Matthey et al. now report that the selective Gq inhibitor FR900359 reduces airway tone in mouse, pig, and human airways ex vivo and decreases airway resistance in mouse models of asthma in vivo. The compound has the advantage that it can be locally applied to the lung via inhalation and shows promising properties that may prove useful for treating obstructive airway disease. Obstructive lung diseases are common causes of disability and death worldwide. A hallmark feature is aberrant activation of Gq protein–dependent signaling cascades. Currently, drugs targeting single G protein (heterotrimeric guanine nucleotide–binding protein)–coupled receptors (GPCRs) are used to reduce airway tone. However, therapeutic efficacy is often limited, because various GPCRs contribute to bronchoconstriction, and chronic exposure to receptor-activating medications results in desensitization. We therefore hypothesized that pharmacological Gq inhibition could serve as a central mechanism to achieve efficient therapeutic bronchorelaxation. We found that the compound FR900359 (FR), a membrane-permeable inhibitor of Gq, was effective in silencing Gq signaling in murine and human airway smooth muscle cells. Moreover, FR both prevented bronchoconstrictor responses and triggered sustained airway relaxation in mouse, pig, and human airway tissue ex vivo. Inhalation of FR in healthy wild-type mice resulted in high local concentrations of the compound in the lungs and prevented airway constriction without acute effects on blood pressure and heart rate. FR administration also protected against airway hyperreactivity in murine models of allergen sensitization using ovalbumin and house dust mite as allergens. Our findings establish FR as a selective Gq inhibitor when applied locally to the airways of mice in vivo and suggest that pharmacological blockade of Gq proteins may be a useful therapeutic strategy to achieve bronchorelaxation in asthmatic lung disease.

Collaboration


Dive into the Ralf Schröder's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Akos Heinemann

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Maria Waldhoer

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wolfgang Platzer

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julia Kargl

Medical University of Graz

View shared research outputs
Researchain Logo
Decentralizing Knowledge