TY - JOUR A1 - Lohse, Christian A1 - Bock, Andreas A1 - Maiellaro, Isabella A1 - Hannawacker, Annette A1 - Schad, Lothar R. A1 - Lohse, Martin J. A1 - Bauer, Wolfgang R. T1 - Experimental and mathematical analysis of cAMP nanodomains JF - PLoS ONE N2 - In their role as second messengers, cyclic nucleotides such as cAMP have a variety of intracellular effects. These complex tasks demand a highly organized orchestration of spatially and temporally confined cAMP action which should be best achieved by compartmentalization of the latter. A great body of evidence suggests that cAMP compartments may be established and maintained by cAMP degrading enzymes, e.g. phosphodiesterases (PDEs). However, the molecular and biophysical details of how PDEs can orchestrate cAMP gradients are entirely unclear. In this paper, using fusion proteins of cAMP FRET-sensors and PDEs in living cells, we provide direct experimental evidence that the cAMP concentration in the vicinity of an individual PDE molecule is below the detection limit of our FRET sensors (<100nM). This cAMP gradient persists in crude cytosol preparations. We developed mathematical models based on diffusion-reaction equations which describe the creation of nanocompartments around a single PDE molecule and more complex spatial PDE arrangements. The analytically solvable equations derived here explicitly determine how the capability of a single PDE, or PDE complexes, to create a nanocompartment depend on the cAMP degradation rate, the diffusive mobility of cAMP, and geometrical and topological parameters. We apply these generic models to our experimental data and determine the diffusive mobility and degradation rate of cAMP. The results obtained for these parameters differ by far from data in literature for free soluble cAMP interacting with PDE. Hence, restricted cAMP diffusion in the vincinity of PDE is necessary to create cAMP nanocompartments in cells. KW - fluorescence resonance energy transfer KW - yellow fluorescent protein KW - radii KW - adenylyl cyclase signaling cascade KW - cell fusion KW - cytosol KW - isoproterenol KW - absorption KW - cyclic nucleotides such as cyclic adenosine monophosphate Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-170972 VL - 12 IS - 4 ER - TY - JOUR A1 - Schupp, Nicole A1 - Stopper, Helga A1 - Heidland, August T1 - DNA Damage in Chronic Kidney Disease: Evaluation of Clinical Biomarkers JF - Oxidative Medicine and Cellular Longevity N2 - Patients with chronic kidney disease (CKD) exhibit an increased cancer risk compared to a healthy control population. To be able to estimate the cancer risk of the patients and to assess the impact of interventional therapies thereon, it is of particular interest to measure the patients’ burden of genomic damage. Chromosomal abnormalities, reduced DNA repair, and DNA lesions were found indeed in cells of patients with CKD. Biomarkers for DNA damage measurable in easily accessible cells like peripheral blood lymphocytes are chromosomal aberrations, structural DNA lesions, and oxidatively modified DNA bases. In this review the most common methods quantifying the three parameters mentioned above, the cytokinesis-block micronucleus assay, the comet assay, and the quantification of 8-oxo-7,8-dihydro-2′-deoxyguanosine, are evaluated concerning the feasibility of the analysis and regarding the marker’s potential to predict clinical outcomes. KW - chronic kidney disease KW - cancer risk KW - DNA damage KW - biomarkers Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-166569 VL - 2016 IS - 3592042 ER - TY - JOUR A1 - Fathy, Moustafa A1 - Fawzy, Michael Atef A1 - Hintzsche, Henning A1 - Nikaido, Toshio A1 - Dandekar, Thomas A1 - Othman, Eman M. T1 - Eugenol exerts apoptotic effect and modulates the sensitivity of HeLa cells to cisplatin and radiation JF - Molecules N2 - Eugenol is a phytochemical present in different plant products, e.g., clove oil. Traditionally, it is used against a number of different disorders and it was suggested to have anticancer activity. In this study, the activity of eugenol was evaluated in a human cervical cancer (HeLa) cell line and cell proliferation was examined after treatment with various concentrations of eugenol and different treatment durations. Cytotoxicity was tested using lactate dehydrogenase (LDH) enzyme leakage. In order to assess eugenol’s potential to act synergistically with chemotherapy and radiotherapy, cell survival was calculated after eugenol treatment in combination with cisplatin and X-rays. To elucidate its mechanism of action, caspase-3 activity was analyzed and the expression of various genes and proteins was checked by RT-PCR and western blot analyses. Eugenol clearly decreased the proliferation rate and increased LDH release in a concentration- and time-dependent manner. It showed synergistic effects with cisplatin and X-rays. Eugenol increased caspase-3 activity and the expression of Bax, cytochrome c (Cyt-c), caspase-3, and caspase-9 and decreased the expression of B-cell lymphoma (Bcl)-2, cyclooxygenase-2 (Cox-2), and interleukin-1 beta (IL-1β) indicating that eugenol mainly induced cell death by apoptosis. In conclusion, eugenol showed antiproliferative and cytotoxic effects via apoptosis and also synergism with cisplatin and ionizing radiation in the human cervical cancer cell line. KW - eugenol KW - HeLa cells KW - cisplatin KW - radiation KW - apoptosis Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-193227 SN - 1420-3049 VL - 24 IS - 21 ER - TY - JOUR A1 - Wölfel, Angela A1 - Sättele, Mathias A1 - Zechmeister, Christina A1 - Nikolaev, Viacheslov O. A1 - Lohse, Martin J. A1 - Boege, Fritz A1 - Jahns, Roland A1 - Boivin-Jahns, Valérie T1 - Unmasking features of the auto-epitope essential for β\(_1\)-adrenoceptor activation by autoantibodies in chronic heart failure JF - ESC Heart Failure N2 - Aims Chronic heart failure (CHF) can be caused by autoantibodies stimulating the heart via binding to first and/or second extracellular loops of cardiac β1-adrenoceptors. Allosteric receptor activation depends on conformational features of the autoantibody binding site. Elucidating these features will pave the way for the development of specific diagnostics and therapeutics. Our aim was (i) to fine-map the conformational epitope within the second extracellular loop of the human β\(_1\)-adrenoceptor (β1ECII) that is targeted by stimulating β\(_1\)-receptor (auto)antibodies and (ii) to generate competitive cyclopeptide inhibitors of allosteric receptor activation, which faithfully conserve the conformational auto-epitope. Methods and results Non-conserved amino acids within the β\(_1\)EC\(_{II}\) loop (compared with the amino acids constituting the ECII loop of the β\(_2\)-adrenoceptor) were one by one replaced with alanine; potential intra-loop disulfide bridges were probed by cysteine–serine exchanges. Effects on antibody binding and allosteric receptor activation were assessed (i) by (auto)antibody neutralization using cyclopeptides mimicking β1ECII ± the above replacements, and (ii) by (auto)antibody stimulation of human β\(_1\)-adrenoceptors bearing corresponding point mutations. With the use of stimulating β\(_1\)-receptor (auto)antibodies raised in mice, rats, or rabbits and isolated from exemplary dilated cardiomyopathy patients, our series of experiments unmasked two features of the β\(_1\)EC\(_{II}\) loop essential for (auto)antibody binding and allosteric receptor activation: (i) the NDPK\(^{211–214}\) motif and (ii) the intra-loop disulfide bond C\(^{209}\)↔C\(^{215}\). Of note, aberrant intra-loop disulfide bond C\(^{209}\)↔C\(^{216}\) almost fully disrupted the functional auto-epitope in cyclopeptides. Conclusions The conformational auto-epitope targeted by cardio-pathogenic β\(_1\)-receptor autoantibodies is faithfully conserved in cyclopeptide homologues of the β\(_1\)EC\(_{II}\) loop bearing the NDPK\(^{211–214}\) motif and the C\(^{209}\)↔C\(^{215}\) bridge while lacking cysteine C216. Such molecules provide promising tools for novel diagnostic and therapeutic approaches in β\(_1\)-autoantibodypositive CHF. KW - antibody/autoantibody KW - β1-adrenoceptor/β1-adrenergic receptor KW - chronic heart failure KW - conformational auto-epitope KW - cyclic peptides/cyclopeptides KW - cyclopeptide therapy Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-235974 VL - 7 IS - 4 ER - TY - JOUR A1 - Scholz, Nicole A1 - Guan, Chonglin A1 - Nieberler, Matthias A1 - Grotmeyer, Alexander A1 - Maiellaro, Isabella A1 - Gao, Shiqiang A1 - Beck, Sebastian A1 - Pawlak, Matthias A1 - Sauer, Markus A1 - Asan, Esther A1 - Rothemund, Sven A1 - Winkler, Jana A1 - Prömel, Simone A1 - Nagel, Georg A1 - Langenhan, Tobias A1 - Kittel, Robert J T1 - Mechano-dependent signaling by Latrophilin/CIRL quenches cAMP in proprioceptive neurons JF - eLife N2 - Adhesion-type G protein-coupled receptors (aGPCRs), a large molecule family with over 30 members in humans, operate in organ development, brain function and govern immunological responses. Correspondingly, this receptor family is linked to a multitude of diverse human diseases. aGPCRs have been suggested to possess mechanosensory properties, though their mechanism of action is fully unknown. Here we show that the Drosophila aGPCR Latrophilin/dCIRL acts in mechanosensory neurons by modulating ionotropic receptor currents, the initiating step of cellular mechanosensation. This process depends on the length of the extended ectodomain and the tethered agonist of the receptor, but not on its autoproteolysis, a characteristic biochemical feature of the aGPCR family. Intracellularly, dCIRL quenches cAMP levels upon mechanical activation thereby specifically increasing the mechanosensitivity of neurons. These results provide direct evidence that the aGPCR dCIRL acts as a molecular sensor and signal transducer that detects and converts mechanical stimuli into a metabotropic response. KW - Latrophilin KW - adhesion GPCR KW - dCIRL KW - sensory physiology KW - metabotropic signalling KW - mechanotransduction Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-170520 VL - 6 IS - e28360 ER - TY - JOUR A1 - Gmach, Philipp A1 - Bathe-Peters, Marc A1 - Telugu, Narasimha A1 - Miller, Duncan C. A1 - Annibale, Paolo T1 - Fluorescence spectroscopy of low-level endogenous β-adrenergic receptor expression at the plasma membrane of differentiating human iPSC-derived cardiomyocytes JF - International Journal of Molecular Sciences N2 - The potential of human-induced pluripotent stem cells (hiPSCs) to be differentiated into cardiomyocytes (CMs) mimicking adult CMs functional morphology, marker genes and signaling characteristics has been investigated since over a decade. The evolution of the membrane localization of CM-specific G protein-coupled receptors throughout differentiation has received, however, only limited attention to date. We employ here advanced fluorescent spectroscopy, namely linescan Fluorescence Correlation Spectroscopy (FCS), to observe how the plasma membrane abundance of the β\(_1\)- and β\(_2\)-adrenergic receptors (β\(_{1/2}\)-ARs), labelled using a bright and photostable fluorescent antagonist, evolves during the long-term monolayer culture of hiPSC-derived CMs. We compare it to the kinetics of observed mRNA levels in wildtype (WT) hiPSCs and in two CRISPR/Cas9 knock-in clones. We conduct these observations against the backdrop of our recent report of cell-to-cell expression variability, as well as of the subcellular localization heterogeneity of β-ARs in adult CMs. KW - GPCR KW - β-adrenergic receptors KW - hiPSC-CM KW - cardiomyocyte KW - fluorescence correlation spectroscopy KW - FCS KW - fluorescence KW - CRISPR/Cas9 KW - differentiation Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-288277 SN - 1422-0067 VL - 23 IS - 18 ER - TY - JOUR A1 - Djelić, Ninoslav A1 - Borozan, Sunčica A1 - Dimitrijević-Srećković, Vesna A1 - Pajović, Nevena A1 - Mirilović, Milorad A1 - Stopper, Helga A1 - Stanimirović, Zoran T1 - Oxidative stress and DNA damage in peripheral blood mononuclear cells from normal, obese, prediabetic and diabetic persons exposed to thyroid hormone in vitro JF - International Journal of Molecular Sciences N2 - Diabetes, a chronic group of medical disorders characterized byhyperglycemia, has become a global pandemic. Some hormones may influence the course and outcome of diabetes, especially if they potentiate the formation of reactive oxygen species (ROS). There is a close relationship between thyroid disorders and diabetes. The main objective of this investigation was to find out whether peripheral blood mononuclear cells (PBMCs) are more prone to DNA damage by triiodothyronine (T\(_3\)) (0.1, 1 and 10 μM) at various stages of progression through diabetes (obese, prediabetics, and type 2 diabetes mellitus—T2DM persons). In addition, some biochemical parameters of oxidative stress (catalase-CAT, thiobarbituric acid reactive substances—TBARS) and lactate dehydrogenase (LDH) were evaluated. PBMCs from prediabetic and diabetic patients exhibited increased sensitivity for T\(_3\) regarding elevated level of DNA damage, inhibition of catalase, and increase of TBARS and LDH. PBMCs from obese patients reacted in the same manner, except for DNA damage. The results of this study should contribute to a better understanding of the role of thyroid hormones in the progression of T2DM. KW - diabetes KW - oxidative stress KW - DNA damage KW - lymphocytes KW - thyroid hormone Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-285988 SN - 1422-0067 VL - 23 IS - 16 ER - TY - JOUR A1 - Sedaghat-Hamedani, Farbod A1 - Rebs, Sabine A1 - Kayvanpour, Elham A1 - Zhu, Chenchen A1 - Amr, Ali A1 - Müller, Marion A1 - Haas, Jan A1 - Wu, Jingyan A1 - Steinmetz, Lars M. A1 - Ehlermann, Philipp A1 - Streckfuss-Bömeke, Katrin A1 - Frey, Norbert A1 - Meder, Benjamin T1 - Genotype complements the phenotype: identification of the pathogenicity of an LMNA splice variant by nanopore long-read sequencing in a large DCM family JF - International Journal of Molecular Sciences N2 - Dilated cardiomyopathy (DCM) is a common cause of heart failure (HF) and is of familial origin in 20–40% of cases. Genetic testing by next-generation sequencing (NGS) has yielded a definite diagnosis in many cases; however, some remain elusive. In this study, we used a combination of NGS, human-induced pluripotent-stem-cell-derived cardiomyocytes (iPSC-CMs) and nanopore long-read sequencing to identify the causal variant in a multi-generational pedigree of DCM. A four-generation family with familial DCM was investigated. Next-generation sequencing (NGS) was performed on 22 family members. Skin biopsies from two affected family members were used to generate iPSCs, which were then differentiated into iPSC-CMs. Short-read RNA sequencing was used for the evaluation of the target gene expression, and long-read RNA nanopore sequencing was used to evaluate the relevance of the splice variants. The pedigree suggested a highly penetrant, autosomal dominant mode of inheritance. The phenotype of the family was suggestive of laminopathy, but previous genetic testing using both Sanger and panel sequencing only yielded conflicting evidence for LMNA p.R644C (rs142000963), which was not fully segregated. By re-sequencing four additional affected family members, further non-coding LMNA variants could be detected: rs149339264, rs199686967, rs201379016, and rs794728589. To explore the roles of these variants, iPSC-CMs were generated. RNA sequencing showed the LMNA expression levels to be significantly lower in the iPSC-CMs of the LMNA variant carriers. We demonstrated a dysregulated sarcomeric structure and altered calcium homeostasis in the iPSC-CMs of the LMNA variant carriers. Using targeted nanopore long-read sequencing, we revealed the biological significance of the variant c.356+1G>A, which generates a novel 5′ splice site in exon 1 of the cardiac isomer of LMNA, causing a nonsense mRNA product with almost complete RNA decay and haploinsufficiency. Using novel molecular analysis and nanopore technology, we demonstrated the pathogenesis of the rs794728589 (c.356+1G>A) splice variant in LMNA. This study highlights the importance of precise diagnostics in the clinical management and workup of cardiomyopathies. KW - familial DCM KW - laminopathy KW - long-read sequencing KW - nanopore KW - induced pluripotent stem cell cardiomyocytes Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-290415 SN - 1422-0067 VL - 23 IS - 20 ER - TY - THES A1 - Nemec, Katarina T1 - Modulation of parathyroid hormone 1 receptor (PTH1R) signaling by receptor activity-modifying proteins (RAMPs) T1 - Regulierung der Signalübertragung des Parathormon 1-Rezeptors (PTH1R) durch Rezeptoraktivitäts-modifizierende Proteine (RAMPs) N2 - The receptor activity-modifying proteins (RAMPs) are ubiquitously expressed membrane proteins that interact with several G protein-coupled receptors (GPCRs), the largest and pharmacologically most important family of cell surface receptors. RAMPs can regulate GPCR function in terms of ligand-binding, G-protein coupling, downstream signaling, trafficking, and recycling. The integrity of their interactions translates to many physiological functions or pathological conditions. Regardless of numerous reports on its essential importance for cell biology and pivotal role in (patho-)physiology, the molecular mechanism of how RAMPs modulate GPCR activation remained largely elusive. This work presents new insights that add to the common understanding of the allosteric regulation of receptor activation and will help interpret how accessory proteins - RAMPs - modulate activation dynamics and how this affects the fundamental aspects of cellular signaling. Using a prototypical class B GPCR, the parathyroid hormone 1 receptor (PTH1R) in the form of advanced genetically encoded optical biosensors, I examined RAMP's impact on the PTH1R activation and signaling in intact cells. A panel of single-cell FRET and confocal microscopy experiments as well canonical and non-canonical functional assays were performed to get a holistic picture of the signaling initiation and transduction of that clinically and therapeutically relevant GPCR. Finally, structural modeling was performed to add molecular mechanistic details to that novel art of modulation. I describe here that RAMP2 acts as a specific allosteric modulator of PTH1R, shifting PTH1R to a unique pre-activated state that permits faster activation in a ligand-specific manner. Moreover, RAMP2 modulates PTH1R downstream signaling in an agonist-dependent manner, most notably increasing the PTH-mediated Gi3 signaling sensitivity and kinetics of cAMP accumulation. Additionally, RAMP2 increases PTH- and PTHrP-triggered β-arrestin2 recruitment to PTH1R and modulates cytosolic ERK1/2 phosphorylation. Structural homology modeling shows that structural motifs governing GPCR-RAMP interaction originate in allosteric hotspots and rationalize functional modulation. Moreover, to interpret the broader role of RAMP's modulation in GPCRs pharmacology, different fluorescent tools to investigate RAMP's spatial organization were developed, and novel conformational biosensors for class B GPCRs were engineered. Lastly, a high throughput assay is proposed and prototyped to expand the repertoire of RAMPs or other membrane protein interactors. These data uncover the critical role of RAMPs in GPCR activation and signaling and set up a novel platform for studying GPCR modulation. Furthermore, these insights may provide a new venue for precise modulation of GPCR function and advanced drug design. N2 - G Protein-gekoppelte Rezeptoren (GPCRs) bilden die größte und pharmakologisch wichtigste Familie von Zelloberflächenrezeptoren, die zahlreiche (patho-)physiologische Prozesse im menschlichen Körper steuern. GPCRs übertragen während des Rezeptoraktivierungsprozesses extrazelluläre Signale in das Zellinnere, wo durch die extrazelluläre Stimulation Konformationsänderungen des Rezeptorkerns auslöst und die Bindung intrazellulärer Bindungspartner – G Proteine, G Protein-gekoppelte Rezeptorkinase und Arrestine - ermöglicht. Es handelt sich also um einen kritischen Prozess in der Signaltransduktion, der durch einige endogene Moleküle wie Ionen, Lipide oder andere Proteine moduliert werden kann und Auswirkungen auf nachgeschaltete Signalkaskaden hat. GPCRs bilden gewebeabhängige Oligomere mit ihren interagierenden Partnern, Rezeptor-Aktivitäts-modifizierende Proteinen (RAMPs), ubiquitär exprimierten Membranproteinen. Bekannt ist, dass sie die Ligandenbindung, die G- Protein-Kopplung, die nachgeschaltete Signalisierung, das Trafficking und das Recycling einiger GPCRs modulieren. Ihre Rolle im kritischsten Prozess der Signaltransduktion - der Rezeptoraktivierung - wurde jedoch nur begrenzt erforscht. Anhand des physiologisch und therapeutisch wichtigen Parathormon-Rezeptors (PTH1R), einem GPCR der Klasse B, wurden die Modulationseffekte von RAMPs auf den Prozess der Rezeptoraktivierung und ihre Folgen für die nachgeschaltete Signalübertragung analysiert. Hierzu wurden verschiedene optische Biosensoren zur Messung der Aktivierung des PTH1R und seiner Signalkaskade entwickelt und in verschiedenen Versuchsanordnungen eingesetzt, mit dem Ziel einen holistischen Blick auf die Interaktion zwischen PTH1R und RAMPs und ihre funktionellen Auswirkungen zu erhalten. Die Interaktion zwischen PTH1R und RAMPs erwies sich als besonders ausgeprägt für RAMP2, und RAMP2 zeigte eine spezifische allosterische Modulation der PTH1R-Konformation, sowohl im basalen als auch im Liganden- aktivierten Zustand. Ein einzigartiger voraktivierter oder (meta-stabiler) Zustand ermöglichte eine schnellere Rezeptoraktivierung auf Liganden-spezifische Weise. Außerdem beeinflusste RAMP2 die G Protein- und Nicht-G Protein-vermittelte Signalübertragung indem es die PTH-vermittelte Gi3-Signalempfindlichkeit und die Kinetik der cAMP-Akkumulation modulierte. Weiterhin erhöhte RAMP2 die Menge der β-Arrestin2-Rekrutierung an PTH1R auf Liganden-spezifische Weise. Dies könnte mit einer erhöhten zytosolischen ERK-Menge zusammenhängen, die hat sich von der nukleären ERK-Phosphorylierung unterscheidet. Um einen molekularen Mechanismus für die vorgestellten Ergebnisse vorzuschlagen, wurden mehrere strukturelle Modelle entwickelt und analysiert. Diese Arbeit liefert den Beweis, dass RAMP die GPCR-Aktivierung mit funktionellen Auswirkungen auf die zelluläre Signalübertragung reguliert. Die Ergebnisse sollten im Zusammenhang mit zellspezifischen Koexpressionsmustern interpretiert werden und können zur Entwicklung von fortschrittlichen Therapeutika positiv beitragen. Da GPCRs praktisch alle Zellfunktionen koordinieren und seit jeher wichtigen Angriffspunkten für Medikamente sind, tragen die vorgestellten Erkenntnisse zum universellen Verständnis der molekularen Mechanismen bei, die den menschlichen Körper orchestrieren. KW - G-Protein gekoppelter Rezeptor KW - GPCR KW - RAMP KW - PTH1R KW - FRET KW - BRET KW - pharmacology KW - Fluoreszenz-Resonanz-Energie-Transfer KW - Förster Resonanz Energie Transfer Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-288588 ER - TY - THES A1 - Eppli, Nenad T1 - Untersuchung des Einflusses der ERK1/2-Autophosphorylierung an Threonin 188 auf Mausherzen mittels transgener Mäuse mit ubiquitärer Überexpression von ERK2\(^{T188D}\) T1 - Study on the effects of autophosphorylation of ERK1/2 at threonine 188 on murine hearts by means of transgenic mice ubiquitiously overexpressing ERK2\(^{T188D}\) N2 - Die ERK2Thr188-Autophosphoylierung stellt einen regulatorischen Signalweg dar, der infolge einer hypertrophen Stimulation die kardiale Hypertrophie begünstigt. Eine Hemmung dieser Phosphorylierung in Kardiomyozyten verhindert die Ausbildung der kardialen Hypertrophie ohne Beeinflussung der kardioprotektiven Funktionen von ERK1/2. Demgegenüber führt die dauerhafte Simulation zu einem gain-of-function-Phänotypen mit ausgeprägter Hypertophie, Fibrose und einer reduzierten Herzfunktion. In dieser Arbeit wurde die dauerhafte Simulation ERK2Thr188-Phosphorylierung (T188D) in einem Mausmodell mit ubiquitärer Expression dieser Mutation untersucht. Dabei konnte gezeigt werden, dass sich nach Stimulation durch TAC in diesen Tieren ein etwas stärkerer hypertropher Phänotyp mit vergrößerten Kardiomyozyten, gesteigerter interstitieller Fibrosierung und reduzierter Herzfunktion ausbildet als in Mäusen mit kardiomyozyten-spezifischer Überexpression diese Mutante. In Fibroblasten- und VSMC-Zelllinien wurde eine gesteigerte Proliferation der T188D-überexprimierenden Zellen im Vergleich zu Kontrollen festgestellt. Somit scheint die ERK2Thr188-Phosphorylierung auch in kardialen Nicht-Myozyten einen maladaptiven Einfluss auf das Herz auszuüben. N2 - Autophosphorylation of ERK2 on threonine 188 is a regulatory signaling pathway facilitating cardiac hypertrophy due to a hypertrophic stimulation. The formation of cardiac hypertrophy can be impaired by inhibition of the phosphorylation without interfering with the cardioprotective functions of ERK1/2. In contrast, permanent stimulation of ERK2Thr188 phosphorylation leads to a gain-of-function phenotype with distinct hypertrophy fibrosis and a reduced cardiac function. Here, we examined a murine model with ubiquitous overexpression of ERK2Thr188 phosphorylation (T188D). We point out that mice showed an increased hypertrophic phenotype with augmented cardiomyocytes, enhanced interstitial fibrosis and reduced cardiac function in response to TAC compared to mice with overexpression of this mutant limited to cardiomyocytes. Fibroblasts and vascular smooth muscle cells overexpressing T188D showed an enhanced proliferation compared to controls. Taken together, the phosphorylation of ERK2 on threonine 188 exerts a maladaptive influence on non-myocytes in the heart as well. KW - Herzhypertrophie KW - cardiac hypertrophy KW - ERK1/2-Autophosphorylierung KW - autophosphorylation of ERK1/2 Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-216558 ER -