11 research outputs found

    Recurrent EZH1 mutations are a second hit in autonomous thyroid adenomas

    Get PDF
    Autonomous thyroid adenomas (ATAs) are a frequent cause of hyperthyroidism. Mutations in the genes encoding the TSH receptor (TSHR) or the Gs protein alpha subunit (GNAS) are found in approximately 70% of ATAs. The involvement of other genes and the pathogenesis of the remaining cases are presently unknown. Here, we performed whole-exome sequencing in 19 ATAs that were paired with normal DNA samples and identified a recurrent hot-spot mutation (c.1712A>G; p.Gln571Arg) in the enhancer of zeste homolog 1 (EZH1) gene, which codes for a catalytic subunit of the polycomb complex. Targeted screening in an independent cohort confirmed that this mutation occurs with high frequency (27%) in ATAs. EZH1 mutations were strongly associated with known (TSHR, GNAS) or presumed (adenylate cyclase 9 [ADCY9]) alterations in cAMP pathway genes. Furthermore, functional studies revealed that the p.Gln571Arg EZH1 mutation caused increased histone H3 trimethylation and increased proliferation of thyroid cells. In summary, this study revealed that a hot-spot mutation in EZH1 is the second most frequent genetic alteration in ATAs. The association between EZH1 and TSHR mutations suggests a 2-hit model for the pathogenesis of these tumors, whereby constitutive activation of the cAMP pathway and EZH1 mutations cooperate to induce the hyperproliferation of thyroid cells.IZKF Wurzburg [B-281]; ERA-NET E-Rare [01GM1407B]; Deutsche KrebshilfeDeutsche Krebshilfe [109994]; Wilhelm Sander Stiftung [2013.010.1]We wish to thank Eileen Bosenberg, Bianca Klupfel, and Ines Elsner for technical support and Ulrike Zabel for DNA cloning. This study was partially supported by grants from the IZKF Wurzburg (B-281, to DC and MF); the ERA-NET E-Rare (01GM1407B, to MF and DC); the Deutsche Krebshilfe (109994, to ME); and the Wilhelm Sander Stiftung (project 2013.010.1, to RP)

    Efficacy and Safety of Three Antiretroviral Regimens for Initial Treatment of HIV-1: A Randomized Clinical Trial in Diverse Multinational Settings

    Get PDF
    Background:Antiretroviral regimens with simplified dosing and better safety are needed to maximize the efficiency of antiretroviral delivery in resource-limited settings. We investigated the efficacy and safety of antiretroviral regimens with once-daily compared to twice-daily dosing in diverse areas of the world.Methods and Findings:1,571 HIV-1-infected persons (47% women) from nine countries in four continents were assigned with equal probability to open-label antiretroviral therapy with efavirenz plus lamivudine-zidovudine (EFV+3TC-ZDV), atazanavir plus didanosine-EC plus emtricitabine (ATV+DDI+FTC), or efavirenz plus emtricitabine-tenofovir-disoproxil fumarate (DF) (EFV+FTC-TDF). ATV+DDI+FTC and EFV+FTC-TDF were hypothesized to be non-inferior to EFV+3TC-ZDV if the upper one-sided 95% confidence bound for the hazard ratio (HR) was ≤1.35 when 30% of participants had treatment failure.An independent monitoring board recommended stopping study follow-up prior to accumulation of 472 treatment failures. Comparing EFV+FTC-TDF to EFV+3TC-ZDV, during a median 184 wk of follow-up there were 95 treatment failures (18%) among 526 participants versus 98 failures among 519 participants (19%; HR 0.95, 95% CI 0.72-1.27; p = 0.74). Safety endpoints occurred in 243 (46%) participants assigned to EFV+FTC-TDF versus 313 (60%) assigned to EFV+3TC-ZDV (HR 0.64, CI 0.54-0.76; p<0.001) and there was a significant interaction between sex and regimen safety (HR 0.50, CI 0.39-0.64 for women; HR 0.79, CI 0.62-1.00 for men; p = 0.01). Comparing ATV+DDI+FTC to EFV+3TC-ZDV, during a median follow-up of 81 wk there were 108 failures (21%) among 526 participants assigned to ATV+DDI+FTC and 76 (15%) among 519 participants assigned to EFV+3TC-ZDV (HR 1.51, CI 1.12-2.04; p = 0.007).Conclusion: EFV+FTC-TDF had similar high efficacy compared to EFV+3TC-ZDV in this trial population, recruited in diverse multinational settings. Superior safety, especially in HIV-1-infected women, and once-daily dosing of EFV+FTC-TDF are advantageous for use of this regimen for initial treatment of HIV-1 infection in resource-limited countries. ATV+DDI+FTC had inferior efficacy and is not recommended as an initial antiretroviral regimen.Trial Registration:http://www.ClinicalTrials.gov NCT00084136

    Ein neues Model der GPCR Signaltransduktion am trans-Golgi-Netzwerk von Schilddrüsenzellen

    No full text
    Whereas G-protein coupled receptors (GPCRs) have been long believed to signal through cyclic AMP exclusively at cell surface, our group has previously shown that GPCRs not only signal at the cell surface but can also continue doing so once internalized together with their ligands, leading to persistent cAMP production. This phenomenon, which we originally described for the thyroid stimulating hormone receptor (TSHR) in thyroid cells, has been observed also for other GPCRs. However, the intracellular compartment(s) responsible for such persistent signaling and its consequences on downstream effectors were insufficiently characterized. The aim of this study was to follow by live-cell imaging the trafficking of internalized TSHRs and other involved signaling proteins as well as to understand the consequences of signaling by internalized TSHRs on the downstream activation of protein kinase A (PKA). cAMP and PKA activity was measured in real-time in living thyroid cells using FRET-based sensors Epac1-camp and AKAR2 respectively. The results suggest that TSH co-internalizes with its receptor and that the internalized TSH/TSHR complexes traffic retrogradely to the trans-Golgi network (TGN). This study also provides evidence that these internalized TSH/TSHR complexes meet an intracellular pool of Gs proteins in sorting endosomes and in TGN and activate it there, as visualized in real-time using a conformational biosensor nanobody, Nb37. Acute Brefeldin A-induced Golgi collapse hinders the retrograde trafficking of TSH/TSHR complexes, leading to reduced cAMP production and PKA signaling. BFA pretreatment was also able to attenuate CREB phosphorylation suggesting that an intact Golgi/TGN organisation is essential for an efficient cAMP/PKA signaling by internalized TSH/TSHR complexes. Taken together this data provides evidence that internalized TSH/TSHR complexes meet and activate Gs proteins in sorting endosomes and at the TGN, leading to a local activation of PKA and consequently increased CREB activation. These findings suggest unexpected functions for receptor internalization, with major pathophysiological and pharmacological implications.G-Protein-gekoppelte Rezeptoren sind nur in Eukaryonten vorhandeln und bilden die größte und diverseste Familie von Zellmembranrezeptoren. Sie reagieren auf eine vielfältige Gruppe von Stimuli die verschiedene Effektoren aktivieren und damit nachgelagerte Signalkaskaden auslösen, die letztlich entscheidend für die Zellphysiologie sind. Die Regelung der Ligand-vermittelten Signaltransduktion wird hauptsächlich durch die Desensibilisierung des GPCR mittels Dephosphorylierung (katalysiert durch GRK) und zusätzlich durch Internalisierung des GPCR gesteuert. Die Annahme, dass GPCRs für cAMP nur an der Zellmembran signalisieren und nicht mehr sobald sie in die Zelle internalisiert wurden, konnte durch wegweisende unabhängige Forschung an GPCRs im Besonderen an TSHR und PTHR geändert werden. So konnte gezeigt werden, dass sie für cAMP nicht nur an der Zellmembran signalisieren, sondern auch, wenn sie in intrazelluläre Zellkompartimente internalisiert wurde. Dieses Phänomen („sustained signaling“ hier „anhaltende Signalisierung“) wurde seitdem für andere GPCRs (z.B. 2-AR, V2R und LHR) beschrieben. Aber die Zellkompartimente wurden für nachhaltige intrazelluläre Signale nicht ausreichend charakterisiert. Das Ziel dieser Arbeit war es die Bewegung und die dynamische Natur der möglichen signalisierenden Kompartimente mittels „real-time TIRF“-Mikroskopie und die Signalisierung unter Verwendung von „real-time FRET“ in primären Maus Schilddrüsenzellen zu untersuchen. Die vorliegende Arbeit berichtet, dass TSH/TSHR Komplexe internalisieren und ein signifikanter Teil, welcher vom Retromer Komplex angeführt wird, gelangt über den retrograden (rückwärts gerichteten) Transport in das trans-Golgi-Netzwerk (TGN). Diese TSH/TSHR-Komplexe treffen nicht in den frühen Endosomen auf die Gs-Proteine, sondern in den „Sortierer Endosomen“ und in dem TGN. Ein direkter Beweis für Gs Protein Aktivierung und Signaltransduktion am TGN und in Sortierer Endosomen konnte mittels des nanobody Nb37, einem spezifischen Biosensor für das aktive Gs Protein, erbracht werden. Es konnte gezeigt werden, dass die Sequestrierung von Nb37 an diesen Kompartimenten ein szintillierendes Verhalten in Zeit und Raum zeigt. Die vorliegende Arbeit zeigt, dass die katalytische Untereinheit der PKA am Golgi/TGN angereichert ist. Die Behandlung mit Brefeldin A führt zum Verlust dieser PKA Lokalisation am Golgi. Die Beschädigung und Reorganisation des TGN durch Brefeldin A führt zu a) einer abgeschwächten cAMP Reaktion b) einer dreiphasigen PKA Reaktion charakterisiert durch eine schnelle erste Phase, eine langsame (deutlich abgeschwächte) zweite Phase und eine verzögerte dritte Phase und schließlich c) einer abgeschwächte CREB Phosphorylierung. Es gibt Anzeichen dafür, dass die Reorganisation des TGN Kompartimente betrifft, die verantwortlich für intrazelluläre cAMP- und PKA-Signalisierung sind. Zusammenfassend lässt sich sagen, dass das TGN eines der Kompartimente ist, das für die anhaltende TSHR-Signalisierung verantwortlich ist

    Internalized TSH receptors en route to the TGN induce local GS_{S}-protein signaling and gene transcription

    Get PDF
    A new paradigm of G-protein-coupled receptor (GPCR) signaling at intracellular sites has recently emerged, but the underlying mechanisms and functional consequences are insufficiently understood. Here, we show that upon internalization in thyroid cells, endogenous TSH receptors traffic retrogradely to the trans-Golgi network (TGN) and activate endogenous Gs-proteins in the retromer-coated compartment that brings them to the TGN. Receptor internalization is associated with a late cAMP/protein kinase A (PKA) response at the Golgi/TGN. Blocking receptor internalization, inhibiting PKA II/interfering with its Golgi/TGN localization, silencing retromer or disrupting Golgi/TGN organization all impair efficient TSH-dependent cAMP response element binding protein (CREB) phosphorylation. These results suggest that retrograde trafficking to the TGN induces local GS_{S}-protein activation and cAMP/PKA signaling at a critical position near the nucleus, which appears required for efficient CREB phosphorylation and gene transcription. This provides a new mechanism to explain the functional consequences of GPCR signaling at intracellular sites and reveals a critical role for the TGN in GPCR signaling

    Recurrent <em>EZH1 </em>mutations are a second hit in autonomous thyroid adenomas.

    Get PDF
    Autonomous thyroid adenomas (ATAs) are a frequent cause of hyperthyroidism. Mutations in the genes encoding the TSH receptor (TSHR) or the Gs protein &alpha; subunit (GNAS) are found in approximately 70% of ATAs. The involvement of other genes and the pathogenesis of the remaining cases are presently unknown. Here, we performed whole-exome sequencing in 19 ATAs that were paired with normal DNA samples and identified a recurrent hot-spot mutation (c.1712A&gt;G; p.Gln571Arg) in the enhancer of zeste homolog 1 (EZH1) gene, which codes for a catalytic subunit of the polycomb complex. Targeted screening in an independent cohort confirmed that this mutation occurs with high frequency (27%) in ATAs. EZH1 mutations were strongly associated with known (TSHR, GNAS) or presumed (adenylate cyclase 9 [ADCY9]) alterations in cAMP pathway genes. Furthermore, functional studies revealed that the p.Gln571Arg EZH1 mutation caused increased histone H3 trimethylation and increased proliferation of thyroid cells. In summary, this study revealed that a hot-spot mutation in EZH1 is the second most frequent genetic alteration in ATAs. The association between EZH1 and TSHR mutations suggests a 2-hit model for the pathogenesis of these tumors, whereby constitutive activation of the cAMP pathway and EZH1 mutations cooperate to induce the hyperproliferation of thyroid cells
    corecore