Network


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

Hotspot


Dive into the research topics where Gerhard Trube is active.

Publication


Featured researches published by Gerhard Trube.


Neuron | 1990

The effect of subunit composition of rat brain GABAA receptors on channel function

Erwin Sigel; Roland Baur; Gerhard Trube; Hanms Möhler; Pari Malherbe

Different combinations of cloned rat brain subunit isoforms of the GABAA receptor channel were expressed in Xenopus oocytes. The voltage-clamp technique was then used to measure properties of the GABA-induced membrane currents and to study the effects of various modulators of the GABAA receptor channel (diazepam, DMCM, pentobarbital, and picrotoxin). This approach was used to obtain information on the minimal structural requirements for several functional properties of the ion channel. The combination alpha 5 beta 2 gamma 2 was identified as the minimal requirement reproducing consensus properties of the vertebrate GABAA receptor channel, including cooperativity of GABA-dependent channel gating with a Ka in the range of 10 microM, modulation by various drugs acting at the benzodiazepine binding site, picrotoxin sensitivity, and barbiturate effects.


The Journal of Physiology | 1996

A novel mechanism of activity‐dependent NMDA receptor antagonism describes the effect of ifenprodil in rat cultured cortical neurones.

James N.C. Kew; Gerhard Trube; John A. Kemp

1. Ifenprodil is a selective, atypical non‐competitive antagonist of NMDA receptors that contain the NR2B subunit with an undefined mechanism of action. Ifenprodil is neuroprotective in in vivo models of cerebral ischaemia but lacks many of the undesirable side‐effects associated with NMDA antagonist. 2. Using whole‐cell voltage‐clamp recordings, we have studied the mechanism of inhibition of NMDA‐evoked currents by ifenprodil in rat cultured cortical neurones in the presence of saturating concentrations of glycine. 3. Ifenprodil antagonized NMDA receptors in an activity‐dependent manner, whilst also increasing the receptor affinity for glutamate recognition‐site agonists. Ifenprodil inhibition curves against 10 and 100 microM NMDA‐evoked currents yielded IC50 values of 0.88 and 0.17 microM, respectively. Thus, the apparent affinity of ifenprodil for the NMDA receptor is increased in an NMDA concentration‐dependent manner. 4. Currents evoked by 0.3 and 1 microM NMDA were potentiated to approximately 200% of control levels in the presence of 3 microM ifenprodil. Thus, with increasing concentration of NMDA the effect of ifenprodil on NMDA‐evoked currents changed from one of potentiation to one of increasing inhibition. 5. These results are predicted by a reaction scheme in which ifenprodil exhibits a 39‐ and 50‐fold higher affinity for the agonist‐bound activated and desensitized states of the NMDA receptor, respectively, relative to the resting, agonist‐unbound state. Furthermore, ifenprodil binding to the NMDA receptor results in a 6‐fold higher affinity for glutamate site agonists. 6. This represents a novel mechanism of NMDA receptor antagonism that, together with the subunit selectivity, probably contributes to the attractive neuropharmacological profile of this and related compounds.


Proceedings of the National Academy of Sciences of the United States of America | 2011

TAAR1 activation modulates monoaminergic neurotransmission, preventing hyperdopaminergic and hypoglutamatergic activity

Florent G. Revel; Jean-Luc Moreau; Raul R. Gainetdinov; Amyaouch Bradaia; Tatyana D. Sotnikova; Roland Mory; Sean Durkin; Katrin Groebke Zbinden; Roger David Norcross; Claas Aiko Meyer; Veit Metzler; Sylvie Chaboz; Laurence Ozmen; Gerhard Trube; Bruno Pouzet; Bernhard Bettler; Marc G. Caron; Joseph G. Wettstein; Marius C. Hoener

The trace amine-associated receptor 1 (TAAR1), activated by endogenous metabolites of amino acids like the trace amines p-tyramine and β-phenylethylamine, has proven to be an important modulator of the dopaminergic system and is considered a promising target for the treatment of neuropsychiatric disorders. To decipher the brain functions of TAAR1, a selective TAAR1 agonist, RO5166017, was engineered. RO5166017 showed high affinity and potent functional activity at mouse, rat, cynomolgus monkey, and human TAAR1 stably expressed in HEK293 cells as well as high selectivity vs. other targets. In mouse brain slices, RO5166017 inhibited the firing frequency of dopaminergic and serotonergic neurons in regions where Taar1 is expressed (i.e., the ventral tegmental area and dorsal raphe nucleus, respectively). In contrast, RO5166017 did not change the firing frequency of noradrenergic neurons in the locus coeruleus, an area devoid of Taar1 expression. Furthermore, modulation of TAAR1 activity altered the desensitization rate and agonist potency at 5-HT1A receptors in the dorsal raphe, suggesting that TAAR1 modulates not only dopaminergic but also serotonergic neurotransmission. In WT but not Taar1−/− mice, RO5166017 prevented stress-induced hyperthermia and blocked dopamine-dependent hyperlocomotion in cocaine-treated and dopamine transporter knockout mice as well as hyperactivity induced by an NMDA antagonist. These results tie TAAR1 to the control of monoamine-driven behaviors and suggest anxiolytic- and antipsychotic-like properties for agonists such as RO5166017, opening treatment opportunities for psychiatric disorders.


Proceedings of the National Academy of Sciences of the United States of America | 2009

The selective antagonist EPPTB reveals TAAR1-mediated regulatory mechanisms in dopaminergic neurons of the mesolimbic system.

Amyaouch Bradaia; Gerhard Trube; Henri Stalder; Roger David Norcross; Laurence Ozmen; Joseph G. Wettstein; Audrée Pinard; Danièle Buchy; Martin Gassmann; Marius C. Hoener; Bernhard Bettler

Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) that is nonselectively activated by endogenous metabolites of amino acids. TAAR1 is considered a promising drug target for the treatment of psychiatric and neurodegenerative disorders. However, no selective ligand to identify TAAR1-specific signaling mechanisms is available yet. Here we report a selective TAAR1 antagonist, EPPTB, and characterize its physiological effects at dopamine (DA) neurons of the ventral tegmental area (VTA). We show that EPPTB prevents the reduction of the firing frequency of DA neurons induced by p-tyramine (p-tyr), a nonselective TAAR1 agonist. When applied alone, EPPTB increases the firing frequency of DA neurons, suggesting that TAAR1 either exhibits constitutive activity or is tonically activated by ambient levels of endogenous agonist(s). We further show that EPPTB blocks the TAAR1-mediated activation of an inwardly rectifying K+ current. When applied alone, EPPTB induces an apparent inward current, suggesting the closure of tonically activated K+ channels. Importantly, these EPPTB effects were absent in Taar1 knockout mice, ruling out off-target effects. We additionally found that both the acute application of EPPTB and the constitutive genetic lack of TAAR1 increase the potency of DA at D2 receptors in DA neurons. In summary, our data support that TAAR1 tonically activates inwardly rectifying K+ channels, which reduces the basal firing frequency of DA neurons in the VTA. We hypothesize that the EPPTB-induced increase in the potency of DA at D2 receptors is part of a homeostatic feedback mechanism compensating for the lack of inhibitory TAAR1 tone.


European Journal of Pharmacology | 1993

Dextromethorphan blocks N-methyl-D-aspartate-induced currents and voltage-operated inward currents in cultured cortical neurons

Rainer Netzer; Pascal Pflimlin; Gerhard Trube

The effect of dextromethorphan on several types of cation currents in cultured rat cortical neurons and PC12 cells was studied by using the whole-cell configuration of the patch-clamp technique. The Ba2+ current through L- and N-type Ca2+ channels was blocked with similar potencies (52-71 microM) in both types of cells. The effect was not voltage-dependent, in contrast to that of amlodipine (a dihydropyridine). Dextromethorphan was able to block the Ba2+ current completely unlike amlodipine and omega-conotoxin (an N-type channel blocker) which produced only partial inhibition. The voltage-activated Na+ and Ca2+ channels in cortical neurons were inhibited by similar concentrations of dextromethorphan (IC50 approximately 80 microM). The morphinan was at least 100 times more potent (IC50 = 0.55 microM) as a blocker of the current induced by N-methyl-D-aspartate (NMDA) in cortical neurons. Currents induced by (RS)-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid ((RS)-AMPA) or kainic acid were not significantly affected even at 1 mM. The results suggest that the neuroprotective effect of dextromethorphan, previously found to occur in a concentration range of 10-100 microM, may be due to a complete blockade of the NMDA receptor channel and a partial inhibition of voltage-dependent Ca2+ and Na+ channels.


ChemBioChem | 2002

A Virtual Screening Method for Prediction of the hERG Potassium Channel Liability of Compound Libraries

Olivier Roche; Gerhard Trube; Jochen Zuegge; Pascal Pflimlin; Alexander Alanine; Gisbert Schneider

A computer‐based method has been developed for prediction of the hERG (human ether‐à‐go‐go related gene) K+‐channel affinity of low molecular weight compounds. hERG channel blockage is a major concern in drug design, as such blocking agents can cause sudden cardiac death. Various techniques were applied to finding appropriate molecular descriptors for modeling structure–activity relationships: substructure analysis, self‐organizing maps (SOM), principal component analysis (PCA), partial least squares fitting (PLS), and supervised neural networks. The most accurate prediction system was based on an artificial neural network. In a validation study, 93 % of the nonblocking agents and 71 % of the hERG channel blockers were correctly classified. This virtual screening method can be used for general compound‐library shaping and combinatorial library design.


British Journal of Pharmacology | 1998

State‐dependent NMDA receptor antagonism by Ro 8‐4304, a novel NR2B selective, non‐competitive, voltage‐independent antagonist

James N.C. Kew; Gerhard Trube; John A. Kemp

Subunit‐selective blockade of N‐methyl‐D‐aspartate (NMDA) receptors provides a potentially attractive strategy for neuroprotection in the absence of undesirable side effects. Here, we describe a novel NR2B‐selective NMDA antagonist, 4‐{3‐[4‐(4‐fluoro‐phenyl)‐3,6‐dihydro‐2H‐pyridin‐1‐yl]‐2‐hydroxy‐propoxy}‐benzamide (Ro 8‐4304), which exhibits >100 fold higher affinity for recombinant NR1001/NR2B than NR1001/NR2A receptors. Ro 8‐4304 is a voltage‐independent, non‐competitive antagonist of NMDA receptors in rat cultured cortical neurones and exhibits a state‐dependent mode of action similar to that described for ifenprodil. The apparent affinity of Ro 8‐4304 for the NMDA receptor increased in an NMDA concentration‐dependent manner so that Ro 8‐4304 inhibited 10 and 100 μM NMDA responses with IC50s of 2.3 and 0.36 μM, respectively. Currents elicited by 1 μM NMDA were slightly potentiated in the presence of 10 μM Ro 8‐4304, and Ro 8‐4304 binding slowed the rate of glutamate dissociation from NMDA receptors. These results were predicted by a reaction scheme in which Ro 8‐4304 exhibits a 14 and 23 fold higher affinity for the activated and desensitized states of the NMDA receptor, respectively, relative to the agonist‐unbound resting state. Additionally, Ro 8‐4304 binding resulted in a 3–4 fold increase in receptor affinity for glutamate site agonists. Surprisingly, whilst exhibiting a similar affinity for NR2B‐containing NMDA receptors as ifenprodil, Ro 8‐4304 exhibited markedly faster kinetics of binding and unbinding to the NMDA receptor. This spectrum of kinetic behaviour reveals a further important feature of this emerging class of NR2B‐selective compounds.


Bioorganic & Medicinal Chemistry Letters | 2009

The discovery and unique pharmacological profile of RO4938581 and RO4882224 as potent and selective GABAA α5 inverse agonists for the treatment of cognitive dysfunction

Henner Knust; Guido Achermann; Theresa M. Ballard; Bernd Buettelmann; Rodolfo Gasser; Holger Fischer; Maria-Clemencia Hernandez; Frédéric Knoflach; Andreas Koblet; Heinz Stadler; Andrew William Thomas; Gerhard Trube; Pius Waldmeier

Lead optimisation of the imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepine class led to the identification of two clinical leads [RO4882224 (11) and RO4938581 (44)] functioning as novel potent and selective GABAA alpha5 inverse agonists. The unique pharmacological profiles and optimal pharmacokinetic profiles resulted in in vivo activity in selected cognition models.


European Journal of Neuroscience | 1992

Pharmacological and electrophysiological properties of recombinant GABAA receptors comprising the α3, β1 and γ2 subunits

F. Knoflach; Kurt Harald Backus; T. Giller; Pari Malherbe; P. Pflimlin; H. Möhler; Gerhard Trube

To assess the role of subunits for channel function and drug modulation in recombinant GABAA receptors, the α3β1γ2 subunits and the dual combinations α3β1, β1γ2 and α3γ2 were expressed by transfection of human embryonic kidney cells and by RNA injection in Xenopus oocytes (α3β1γ2 combination). GABA‐induced chloride currents were recorded using the whole‐cell configuration of the patch‐clamp technique (transfected cells) or the voltage‐clamp technique (oocytes). The currents recorded from the α3β1Γ2 subunit combination in transfected cells were reduced by bicuculline and picrotoxin, enhanced by flunitrazepam in a flumazenil‐sensitive manner and reduced by β‐carboline‐3‐carboxylic acid methyl ester (β‐CCM). The GABA‐induced current was reduced by β‐CCM in all combinations containing the γ2 subunit, but potentiation by flunitrazepam was only obtained when the 72 subunit was coexpressed in the presence of the α3 subunit (α3β1γ2 or α3γ2). The GABA sensitivities of the receptors were similar when the α3β1γ2 combination was expressed in oocytes (half‐maximum effective concentration = 240 μM) or in the kidney cell line (270 μM). However, the currents were less potentiated by flunitrazepam in oocytes (129% of controls) than in transfected cells (189%). These results suggest that the α3β1γ2 subunit combination, which is coexpressed in various brain regions as shown by in situ hybridization histochemistry, may represent a building block of functional GABAA receptors in situ.


Brain Research | 1991

Action of diazepam on the voltage-dependent Na+ current. Comparison with the effects of phenytoin, carbamazepine, lidocaine and flumazenil.

Kurt Harald Backus; Pascal Pflimlin; Gerhard Trube

The influence of diazepam, an agonist, and flumazenil (Ro 15-1788), an antagonist of the benzodiazepine receptor, on repetitive firing of action potentials in cultured spinal neurons and on voltage-dependent Na+ currents in cultured N2A neuroblastoma cells was examined. The effects were compared to those of the antiepileptics phenytoin and carbamazepine and the local anesthetic lidocaine. The whole-cell configuration of the patch-clamp technique was used for potential and current recording. Diazepam (10 microM) or phenytoin (10 microM) reduced the duration of repetitive action potential discharges in 50 or 67% of the spinal neurons, respectively. At a concentration of 100 microM repetitive firing was completely blocked. Flumazenil (100 microM) had no effect. In N2A neuroblastoma cells diazepam, phenytoin, carbamazepine and lidocaine, but not flumazenil, at a concentration of 100 microM reduced the Na+ current to 60-67% of control. At 10 microM no or only a weak depression was seen with any drug. In the presence of diazepam (100 microM) the Na+ channel inactivation curve was shifted in the hyperpolarizing direction by -4.8 +/- 0.5 mV. Phenytoin, carbamazepine and lidocaine (all 100 microM) caused stronger shifts of -17.4 +/- 2.1, -10.6 +/- 0.9 and -17.0 +/- 2.1 mV, respectively. Inhibition of the Na+ current by diazepam increased use-dependently over 9 depolarizing pulses repeated at high frequency (200 Hz), whereas use-dependent effects of the other compounds developed less rapidly. At a low stimulation rate (7 Hz) use-dependent block was pronounced with lidocaine, but weak or absent with diazepam and carbamazepine.(ABSTRACT TRUNCATED AT 250 WORDS)

Collaboration


Dive into the Gerhard Trube'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

Marie-Paule Heitz

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge