8 research outputs found

    Functional Analysis of Nuclear beta-Adrenergic Receptors in the Myocardium

    Full text link
    Récemment plusieurs récepteurs couplés aux protéines G (RCPGs) ont été caractérisés au niveau des membranes intracellulaires, dont la membrane nucléaire. Notre objectif était de déterminer si les sous-types de récepteurs β-adrénergiques (βAR) et leurs machineries de signalisation étaient fonctionnels et localisés à la membrane nucléaire des cardiomyocytes. Nous avons démontré la présence des β1AR et β3AR, mais pas du β2AR à la membrane nucléaire de myocytes ventriculaires adultes par immunobuvardage, par microscopie confocale, et par des essais fonctionnels. De plus, certains partenaires de signalisation comme les protéines GαS, Gαi, l’adénylate cyclase II, et V/VI y étaient également localisés. Les sous-types de βAR nucléaires étaient fonctionnels puisqu'ils pouvaient lier leurs ligands et activer leurs effecteurs. En utilisant des noyaux isolés, nous avons observé que l'agoniste non-sélectif isoprotérénol (ISO), et que le BRL37344, un ligand sélectif du β3AR, stimulaient l'initiation de la synthèse de l’ARN, contrairement à l'agoniste sélectif du β1AR, le xamotérol. Cette synthèse était abolie par la toxine pertussique (PTX). Cependant, la stimulation des récepteurs nucléaires de type B de l’endothéline (ETB) causaient une réduction de l'initiation de la synthèse d’ARN. Les voies de signalisations impliquées dans la régulation de la synthèse d’ARN par les RCPGs ont ensuite été étudiées en utilisant des noyaux isolés stimulés par des agonistes en présence ou absence de différents inhibiteurs des voies MAP Kinases (proteines kinases activées par mitogènes) et de la voie PI3K/PKB. Les protéines impliquées dans les voies de signalisation de p38, JNK, ERK MAP Kinase et PKB étaient présents dans les noyaux isolés. L'inhibition de PKB par la triciribine, inhibait la synthèse d’ARN. Nous avons ensuite pu mettre en évidence par qPCR que la stimulation par l’ISO entrainait une augmentation du niveau d'ARNr 18S ainsi qu’une diminution de l'expression d’ARNm de NFκB. En contraste, l’ET-1 n’avait aucun effet sur le niveau d’expression de l’ARNr 18S. Nous avons ensuite montré que la stimulation par l’ISO réduisait l’expression de plusieurs gènes impliqués dans l'activation de NFκB, tandis que l’inhibition de ERK1/2 et PKB renversait cet effet. Un microarray global nous a ensuite permis de démontrer que les βARs et les ETRs nucléaires régulaient un grand nombre de gènes distincts. Finalement, les βARs et ETRs nucléaires augmentaient aussi une production de NO de noyaux isolés, ce qui pouvait être inhibée par le LNAME. Ces résultats ont été confirmés dans des cardiomyocytes intacts en utilisant des analogues cagés et perméables d’ISO et de l'ET-1: l'augmentation de NO nucléaire détectée par DAF2-DA, causée par l'ET-1 et l'ISO, pouvait être prévenue par le LNAME. Finalement, l’augmentation de l’initiation de la transcription induite par l'ISO était aussi bloquée par le L-NAME ou par un inbitheur de PKG, le KT5823, suggérant que la voie NO-GC-PKG est impliquée dans la régulation de la transcription par les βAR. En conclusion, les βARs et les ETRs nucléaires utilisent des voies de signalisation différentes et exercent ainsi des effets distincts sur l’expression des gènes cardiaques. Ils représentent donc une avenue intéressante pour le développement de drogues pharmacologiques.Recently several G protein-coupled receptors (GPCRs) have been shown to localize to intracellular membranes, in particular the nuclear membrane. As such, we sought to determine if the β-adrenergic receptor (βAR) subtypes and their associated signalling machinery are functionally localized to nuclear membranes. We demonstrated the presence of β1AR and the β3AR, but not the β2AR, in adult ventricular myocyte nuclei by western blotting, confocal microscopy and functional assays. Downstream signalling partners such as GαS, Gαi and adenylyl cyclase II and V/VI were also present. Nuclear-localized βARs were functional with respect to ligand binding and effector activation. In isolated nuclei, the non-selective βAR agonist isoproterenol (ISO) and the β3AR-selective ligand BRL37344, but not the β1AR-selective xamoterol, stimulated transcription initiation in a pertussis toxin (PTX)-sensitive manner. In contrast, stimulation of type B endothelin receptors (ETB), another GPCR family shown to be present on the nuclear membrane, decreased de novo RNA synthesis. To investigate the signalling pathway(s) involved in GPCR-mediated regulation of RNA synthesis, nuclei were isolated from intact adult rat hearts and treated with receptor agonists in the presence or absence of inhibitors of the PI3K/PKB and mitogen-activated protein kinase (MAPK) pathways. Components of p38, JNK, and ERK1/2 MAPK cascades as well as PKB were detected in nuclear preparations. Inhibition of PKB with triciribine converted the activation of the βAR from stimulatory to inhibitory with regards to transcription initiation. Analysis by qPCR indicated isoproterenol treatment increased 18S rRNA but decreased NFκB mRNA. In contrast, ET-1 had no effect on 18S rRNA expression. Further investigation using pathway-specific PCR arrays revealed that isoproterenol treatment also reduced the expression of several other genes involved in the activation of NFκB and that ERK1/2 and PKB inhibitors attenuated this effect. Subsequent genome-wide microarray analysis has revealed that nuclear βAR and ETB regulated a host of genes in an overlapping but distinct manner. Moreover, both ET-1 and ISO produced an L-NAME-sensitive increase in NO production in isolated cardiac nuclei. These observations were confirmed in intact cardiomyocytes using novel caged analogues of ISO and ET-1 and the cell-permable NO-sensitive fluorescent dye, DAF-2 DA. Briefly, both ET-1 and isoproterenol increased NO production, and this increase was prevented upon preincubation with L-NAME. Moreover, the ability of isoproterenol to increase transcription initiation in isolated nuclei was blocked by L-NAME or the PKG inhibitor KT5823, indicating the NO-GC-PKG pathway is involved in the regulation of gene expression by nuclear βARs. Hence, we have shown that βARs and ETRs in the nuclear membrane activate distinct signalling pathways, resulting in different effects on gene transcription and thus represent potentially important targets for drug development

    Enhanced anti-metastatic bioactivity of an IGF-TRAP re-engineered to improve physicochemical properties

    No full text
    Abstract The insulin-like growth factor (IGF) axis has been implicated in the progression of malignant disease and identified as a clinically important therapeutic target. Several IGF-1 receptor (IGF-1R) targeting drugs including humanized monoclonal antibodies have advanced to phase II/III clinical trials, but to date, have not progressed to clinical use, due, at least in part, to interference with insulin receptor signalling. We previously reported on the production of a soluble fusion protein consisting of the extracellular domain of human IGF-1R fused to the Fc portion of human IgG1 (first generation IGF-TRAP) that bound human IGF-1 and IGF-2 with a 3 log higher affinity than insulin. We showed that the IGF-TRAP had potent anti-cancer activity in several pre-clinical models of aggressive carcinomas. Here we report on the re-engineering of the IGF-TRAP with the aim of improving physicochemical properties and suitability for clinical applications. We show that cysteine-serine substitutions in the Fc hinge region of IGF-TRAP eliminated high-molecular-weight oligomerized species, while a further addition of a flexible linker, not only improved the pharmacokinetic profile, but also enhanced the therapeutic profile of the IGF-TRAP, as evaluated in an experimental colon carcinoma metastasis model. Dose-response profiles of the modified IGF-TRAPs correlated with their bio-availability profiles, as measured by the IGF kinase-receptor-activation (KIRA) assay, providing a novel, surrogate biomarker for drug efficacy. This study provides a compelling example of structure-based re-engineering of Fc-fusion-based biologics for better manufacturability that also significantly improved pharmacological parameters. It identifies the re-engineered IGF-TRAP as a potent anti-cancer therapeutic

    Regulation of cardiac nitric oxide signaling by nuclear β-adrenergic and endothelin receptors.

    No full text
    International audienceAt the cell surface, βARs and endothelin receptors can regulate nitric oxide (NO) production. β-adrenergic receptors (βARs) and type B endothelin receptors (ETB) are present in cardiac nuclear membranes and regulate transcription. The present study investigated the role of the NO pathway in the regulation of gene transcription by these nuclear G protein-coupled receptors. Nitric oxide production and transcription initiation were measured in nuclei isolated from the adult rat heart. The cell-permeable fluorescent dye 4,5-diaminofluorescein diacetate (DAF2 DA) was used to provide a direct assessment of nitric oxide release. Both isoproterenol and endothelin increased NO production in isolated nuclei. Furthermore, a β3AR-selective agonist, BRL 37344, increased NO synthesis whereas the β1AR-selective agonist xamoterol did not. Isoproterenol increased, whereas ET-1 reduced, de novo transcription. The NO synthase inhibitor l-NAME prevented isoproterenol from increasing either NO production or de novo transcription. l-NAME also blocked ET-1-induced NO-production but did not alter the suppression of transcription initiation by ET-1. Inhibition of the cGMP-dependent protein kinase (PKG) using KT5823 also blocked the ability of isoproterenol to increase transcription initiation. Furthermore, immunoblotting revealed eNOS, but not nNOS, in isolated nuclei. Finally, caged, cell-permeable isoproterenol and endothelin-1 analogs were used to selectively activate intracellular β-adrenergic and endothelin receptors in intact adult cardiomyocytes. Intracellular release of caged ET-1 or isoproterenol analogs increased NO production in intact adult cardiomyocytes. Hence, activation of the NO synthase/guanylyl cyclase/PKG pathway is necessary for nuclear β3ARs to increase de novo transcription. Furthermore, we have demonstrated the potential utility of caged receptor ligands in selectively modulating signaling via endogenous intracellular G protein-coupled receptors

    G protein-coupled receptor signalling in the cardiac nuclear membrane: evidence and possible roles in physiological and pathophysiological function

    No full text
    corecore