@article{SubramanianDoeringKollertetal.2016, author = {Subramanian, Hariharan and D{\"o}ring, Frank and Kollert, Sina and Rukoyatkina, Natalia and Sturm, Julia and Gambaryan, Stepan and Stellzig-Eisenhauer, Angelika and Meyer-Marcotty, Philipp and Eigenthaler, Martin and Wischmeyer, Erhard}, title = {PTH1R Mutants Found in Patients with Primary Failure of Tooth Eruption Disrupt G-Protein Signaling}, series = {PLoS One}, volume = {11}, journal = {PLoS One}, number = {11}, doi = {10.1371/journal.pone.0167033}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-147967}, pages = {e0167033}, year = {2016}, abstract = {Aim Primary failure of tooth eruption (PFE) is causally linked to heterozygous mutations of the parathyroid hormone receptor (PTH1R) gene. The mutants described so far lead to exchange of amino acids or truncation of the protein that may result in structural changes of the expressed PTH1R. However, functional effects of these mutations have not been investigated yet. Materials and Methods In HEK293 cells, PTH1R wild type was co-transfected with selected PTH1R mutants identified in patients with PFE. The effects on activation of PTH-regulated intracellular signaling pathways were analyzed by ELISA and Western immunoblotting. Differential effects of wild type and mutated PTH1R on TRESK ion channel regulation were analyzed by electrophysiological recordings in Xenopus laevis oocytes. Results In HEK293 cells, activation of PTH1R wild type increases cAMP and in response activates cAMP-stimulated protein kinase as detected by phosphorylation of the vasodilator stimulated phosphoprotein (VASP). In contrast, the PTH1R mutants are functionally inactive and mutant PTH1R/Gly452Glu has a dominant negative effect on the signaling of PTH1R wild type. Confocal imaging revealed that wild type PTH1R is expressed on the cell surface, whereas PTH1R/Gly452Glu mutant is mostly retained inside the cell. Furthermore, in contrast to wild type PTH1R which substantially augmented K+ currents of TRESK channels, coupling of mutated PTH1R to TRESK channels was completely abolished. Conclusions PTH1R mutations affect intracellular PTH-regulated signaling in vitro. In patients with primary failure of tooth eruption defective signaling of PTH1R mutations is suggested to occur in dento-alveolar cells and thus may lead to impaired tooth movement.}, language = {en} } @article{SchaeferFriedrichJorgensenetal.2018, author = {Sch{\"a}fer, Nadine and Friedrich, Maximilian and J{\o}rgensen, Morten Egevang and Kollert, Sina and Koepsell, Hermann and Wischmeyer, Erhard and Lesch, Klaus-Peter and Geiger, Dietmar and D{\"o}ring, Frank}, title = {Functional analysis of a triplet deletion in the gene encoding the sodium glucose transporter 3, a potential risk factor for ADHD}, series = {PLoS ONE}, volume = {13}, journal = {PLoS ONE}, number = {10}, doi = {10.1371/journal.pone.0205109}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-176495}, pages = {e0205109}, year = {2018}, abstract = {Sodium-glucose transporters (SGLT) belong to the solute carrier 5 family, which is characterized by sodium dependent transport of sugars and other solutes. In contrast, the human SGLT3 (hSGLT3) isoform, encoded by SLC5A4, acts as a glucose sensor that does not transport sugar but induces membrane depolarization by Na\(^{+}\) currents upon ligand binding. Whole-exome sequencing (WES) of several extended pedigrees with high density of attention-deficit/hyperactivity disorder (ADHD) identified a triplet ATG deletion in SLC5A4 leading to a single amino acid loss (ΔM500) in the hSGLT3 protein imperfectly co-segregating with the clinical phenotype of ADHD. Since mutations in homologous domains of hSGLT1 and hSGLT2 were found to affect intestinal and renal function, respectively, we analyzed the functional properties of hSGLT3[wt] and [ΔM500] by voltage clamp and current clamp recordings from cRNA-injected Xenopus laevis oocytes. The cation conductance of hSGLT3[wt] was activated by application of glucose or the specific agonist 1-desoxynojirimycin (DNJ) as revealed by inward currents in the voltage clamp configuration and cell depolarization in the current clamp mode. Almost no currents and changes in membrane potential were observed when glucose or DNJ were applied to hSGLT3[ΔM500]-injected oocytes, demonstrating a loss of function by this amino acid deletion in hSGLT3. To monitor membrane targeting of wt and mutant hSGLT3, fusion constructs with YFP were generated, heterologously expressed in Xenopus laevis oocytes and analyzed for membrane fluorescence by confocal microscopy. In comparison to hSGLT3[wt] the fluorescent signal of mutant [ΔM500] was reduced by 43\% indicating that the mutant phenotype might mainly result from inaccurate membrane targeting. As revealed by homology modeling, residue M500 is located in TM11 suggesting that in addition to the core structure (TM1-TM10) of the transporter, the surrounding TMs are equally crucial for transport/sensor function. In conclusion, our findings indicate that the deletion [ΔM500] in hSGLT3 inhibits membrane targeting and thus largely disrupts glucose-induced sodium conductance, which may, in interaction with other ADHD risk-related gene variants, influence the risk for ADHD in deletion carriers.}, language = {en} } @article{KollertDombertDoeringetal.2015, author = {Kollert, Sina and Dombert, Benjamin and D{\"o}ring, Frank and Wischmeyer, Erhard}, title = {Activation of TRESK channels by the inflammatory mediator lysophosphatidic acid balances nociceptive signalling}, series = {Scientific Reports}, volume = {5}, journal = {Scientific Reports}, number = {12548}, doi = {10.1038/srep12548}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-148312}, year = {2015}, abstract = {In dorsal root ganglia (DRG) neurons TRESK channels constitute a major current component of the standing outward current IK\(_{SO}\). A prominent physiological role of TRESK has been attributed to pain sensation. During inflammation mediators of pain e.g. lysophosphatidic acid (LPA) are released and modulate nociception. We demonstrate co-expression of TRESK and LPA receptors in DRG neurons. Heterologous expression of TRESK and LPA receptors in Xenopus oocytes revealed augmentation of basal K\(^{+}\) currents upon LPA application. In DRG neurons nociception can result from TRPV\(_{1}\) activation by capsaicin or LPA. Upon co-expression in Xenopus oocytes LPA simultaneously increased both depolarising TRPV\(_{1}\) and hyperpolarising TRESK currents. Patch-clamp recordings in cultured DRG neurons from TRESK[wt] mice displayed increased IK\(_{SO}\) after application of LPA whereas under these conditions IK\(_{SO}\) in neurons from TRESK[ko] mice remained unaltered. Under current-clamp conditions LPA application differentially modulated excitability in these genotypes upon depolarising pulses. Spike frequency was attenuated in TRESK[wt] neurons and, in contrast, augmented in TRESK[ko] neurons. Accordingly, excitation of nociceptive neurons by LPA is balanced by co-activation of TRESK channels. Hence excitation of sensory neurons is strongly controlled by the activity of TRESK channels, which therefore are good candidates for the treatment of pain disorders.}, language = {en} } @phdthesis{Kollert2015, author = {Kollert, Sina}, title = {Kaliumkan{\"a}le der K2P-Familie kontrollieren die Aktivit{\"a}t neuronaler Zellen - TRESK als Regulator inflammatorischer Hyperalgesie}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119077}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Das Empfinden von Schmerz ist f{\"u}r uns {\"u}berlebenswichtig. Chronischer Schmerz hingegen hat seine physiologische Bedeutung verloren und wird als eigenes Krankheitsbild angesehen. Schmerzempfindung beginnt mit der Nozizeption. Die Zellk{\"o}rper nozizeptiver Neurone befinden sich in den Spinalganglien (Hinterwurzelganglion, dorsal root ganglion DRG) und Trigeminalganglien (TG). In den DRG-Neuronen macht der Zwei-Poren-Kaliumkanal (K2P) TRESK die Hauptkomponente eines Kaliumstromes, des „standing outward currents" IKSO, aus. Die physiologische Hauptaufgabe der TRESK-Kan{\"a}le liegt in der Regulation der zellul{\"a}ren Erregbarkeit nozizeptiver Neurone. W{\"a}hrend einer Entz{\"u}ndungsreaktion werden Entz{\"u}ndungsmediatoren wie Histamin, Bradykinin, Serotonin und Lysophosphatids{\"a}ure (LPA) ausgesch{\"u}ttet und k{\"o}nnen durch die Aktivierung ihrer G-Protein gekoppelten Rezeptoren (GPCR) oder direkte Interaktion mit Ionenkan{\"a}len die nozizeptive Erregung beeinflussen. Durch Anwendung von RT-PCR und eines neu entwickelten Antik{\"o}rpers wurde die Ko-Expression von TRESK-Kan{\"a}len zusammen mit Kan{\"a}len der Transient-Receptor-Potential-Kationenkanalfamilie (TRP) und LPA-Rezeptoren in DRG-Neuronen nachgewiesen. Durch rekombinante Ko-Expression von TRESK-Kan{\"a}len und LPA2-Rezeptoren in Xenopus Oozyten konnte durch Zugabe von LPA eine fast 10-fache Aktivierung des basalen K+-Stromes erzielt werden. Die Auswertung der Dosis-Wirkungskurve ergab einen EC50-Wert von 0,2 µM LPA. Die LPA-induzierte TRESK-Stromaktivierung konnte durch die Verwendung des mutierten Kanals TRESK[PQAVAD] oder durch die Zugabe des Phospholipase C (PLC) Inhibitors U73122 verhindert werden. Dies zeigt die Beteiligung des PLC-Signalwegs und die Bindung von Calcineurin an den TRESK-Kanal bei der Stromaktivierung. TRESK ist das einzige Mitglied der K2P-Familie, das eine LPA-induzierte Aktivierung des Stromes zeigt. TREK- und TASK-1-Str{\"o}me werden durch LPA inhibiert. In DRG-Neuronen mit kleinem Durchmesser wird Nozizeption durch die Aktivierung von TRPV1-Kan{\"a}len durch Hitze oder Capsaicin, dem Inhaltsstoff des Chilis, und zus{\"a}tzlich durch die Substanz LPA verursacht. Ein weiteres Mitglied der TRP-Familie, der TRPA1-Kanal, ist bei der verst{\"a}rkten Nozizeption w{\"a}hrend einer Entz{\"u}ndung involviert. Werden TRESK- und TRP-Kan{\"a}le in Xenopus Oozyten ko-exprimiert, verursacht LPA gleichzeitig einen Kationeneinw{\"a}rts- wie auch -ausw{\"a}rtsstrom. Unter diesen Bedingungen verschob sich das Umkehrpotenzial in einen Bereich zwischen den Umkehrpotenzialen von Oozyten, die nur den K+-Kanal exprimieren und von Oozyten, die nur den unspezifischen Kationenkanal exprimieren. Durch diese Experimente konnte gezeigt werden, dass die LPA-induzierte Ko-Aktivierung von TRP-Kan{\"a}len und TRESK zu einer Begrenzung des exzitatorischen Effekts f{\"u}hren kann. Die DRG-{\"a}hnlichen F11-Zellen exprimieren keine TRESK-Kan{\"a}le. Sie sind in der Lage durch Strompulse Aktionspotenziale zu generieren. Mit TRESK transfizierte F11-Zellen zeigten eine Verschiebung des Umkehrpotenzials in negative Richtung, einen gr{\"o}ßeren Ausw{\"a}rtsstrom und den Verlust von spannungsgesteuerten Natriumkan{\"a}len. Auch hohe Strompulse konnten keine Aktionspotenziale mehr ausl{\"o}sen. Bei Spannungs-Klemme-Messungen von prim{\"a}ren DRG-Neuronen von TRESK[wt]-M{\"a}usen erh{\"o}hte sich der IKSO nach Zugabe von LPA um {\"u}ber 20 \%. Im Gegensatz dazu zeigten DRG-Neurone von TRESK[ko]-M{\"a}usen unter diesen Bedingungen eine leichte Hemmung des IKSO von etwa 10 \%. In Neuronen, die TRPV1 exprimieren, f{\"u}hrte LPA nicht nur zum Anstieg des IKSO, sondern auch zur Aktivierung eines Einw{\"a}rtsstromes (TRPV1). Im Vergleich dazu wurde in TRESK[ko]-Neuronen durch LPA nur der Einw{\"a}rtsstrom aktiviert. In Strom-Klemme-Experimenten f{\"u}hrte LPA-Applikation zur Entstehung von Aktionspotenzialen mit h{\"o}herer Frequenz in Zellen von TRESK[ko]-M{\"a}usen im Vergleich zu Zellen von TRESK[wt]-M{\"a}usen. Zus{\"a}tzlich wurde die Erregung, die durch Strompulse von 100 pA ausgel{\"o}st wurde, in den beiden Genotypen durch LPA unterschiedlich moduliert. Die Aktionspotenzialfrequenz in TRESK[wt]-Neuronen wurde gesenkt, in TRESK[ko]-Neuronen wurde sie erh{\"o}ht. Die vorliegende Arbeit zeigt, dass die Erregung nozizeptiver Neurone durch LPA aufgrund der Ko-Aktivierung der TRESK-Kan{\"a}le abgeschw{\"a}cht werden kann. Die Erregbarkeit von sensorischen Neuronen wird strak durch die Aktivit{\"a}t und Expression der TRESK-Kan{\"a}le kontrolliert. Deswegen sind TRESK-Kan{\"a}le gute Kandidaten f{\"u}r die pharmakologische Behandlung von Schmerzkrankheiten.}, subject = {Kaliumkanal}, language = {de} } @article{JanschGuentherWaideretal.2018, author = {Jansch, Charline and G{\"u}nther, Katharina and Waider, Jonas and Ziegler, Georg C. and Forero, Andrea and Kollert, Sina and Svirin, Evgeniy and P{\"u}hringer, Dirk and Kwok, Chee Keong and Ullmann, Reinhard and Maierhofer, Anna and Flunkert, Julia and Haaf, Thomas and Edenhofer, Frank and Lesch, Klaus-Peter}, title = {Generation of a human induced pluripotent stem cell (iPSC) line from a 51-year-old female with attention-deficit/hyperactivity disorder (ADHD) carrying a duplication of SLC2A3}, series = {Stem Cell Research}, volume = {28}, journal = {Stem Cell Research}, doi = {10.1016/j.scr.2018.02.005}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-176654}, pages = {136-140}, year = {2018}, abstract = {Fibroblasts were isolated from a skin biopsy of a clinically diagnosed 51-year-old female attention-deficit/hyperactivity disorder (ADHD) patient carrying a duplication of SLC2A3, a gene encoding neuronal glucose transporter-3 (GLUT3). Patient fibroblasts were infected with Sendai virus, a single-stranded RNA virus, to generate transgene-free human induced pluripotent stem cells (iPSCs). SLC2A3-D2-iPSCs showed expression of pluripotency-associated markers, were able to differentiate into cells of the three germ layers in vitro and had a normal female karyotype. This in vitro cellular model can be used to study the role of risk genes in the pathogenesis of ADHD, in a patient-specific manner.}, language = {en} }