Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37
Filtrer
1.
Biosensors (Basel) ; 11(3)2021 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-33668313

RÉSUMÉ

The aryl hydrocarbon receptor (AhR) is a highly conserved cellular sensor of a variety of environmental pollutants and dietary-, cell- and microbiota-derived metabolites with important roles in fundamental biological processes. Deregulation of the AhR pathway is implicated in several diseases, including autoimmune diseases and cancer, rendering AhR a promising target for drug development and host-directed therapy. The pharmacological intervention of AhR processes requires detailed information about the ligand binding properties to allow specific targeting of a particular signaling process without affecting the remaining. Here, we present a novel microscale thermophoresis-based approach to monitoring the binding of purified recombinant human AhR to its natural ligands in a cell-free system. This approach facilitates a precise identification and characterization of unknown AhR ligands and represents a screening strategy for the discovery of potential selective AhR modulators.


Sujet(s)
Récepteurs à hydrocarbure aromatique/composition chimique , Facteurs de transcription à motif basique hélice-boucle-hélice , Humains , Ligands , Tumeurs , Liaison aux protéines , Transduction du signal
3.
Science ; 366(6472)2019 12 20.
Article de Anglais | MEDLINE | ID: mdl-31857448

RÉSUMÉ

Pseudomonas aeruginosa rapidly adapts to altered conditions by quorum sensing (QS), a communication system that it uses to collectively modify its behavior through the production, release, and detection of signaling molecules. QS molecules can also be sensed by hosts, although the respective receptors and signaling pathways are poorly understood. We describe a pattern of regulation in the host by the aryl hydrocarbon receptor (AhR) that is critically dependent on qualitative and quantitative sensing of P. aeruginosa quorum. QS molecules bind to AhR and distinctly modulate its activity. This is mirrored upon infection with P. aeruginosa collected from diverse growth stages and with QS mutants. We propose that by spying on bacterial quorum, AhR acts as a major sensor of infection dynamics, capable of orchestrating host defense according to the status quo of infection.


Sujet(s)
Interactions hôte-pathogène , Infections à Pseudomonas/microbiologie , Pseudomonas aeruginosa/pathogénicité , Détection du quorum/physiologie , Récepteurs à hydrocarbure aromatique/physiologie , Cellules A549 , Animaux , Humains , Larve , Macrophages/microbiologie , Souris , Souris knockout , Pseudomonas aeruginosa/génétique , Détection du quorum/génétique , Récepteurs à hydrocarbure aromatique/génétique , Danio zébré
4.
Mol Pharmacol ; 96(4): 452-462, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31399504

RÉSUMÉ

The large TSH-bound ectodomain of the thyrotropin receptor (TSHR) activates the transmembrane domain (TMD) indirectly via an internal agonist (IA). The ectodomain/TMD interface consists of a converging helix, a Cys-Cys-bridge-linked IA, and extracellular loops (ECL). To investigate the intramolecular course of molecular activation, especially details of the indirect activation, we narrowed down allosteric inhibition sites of negative allosteric modulator (NAM) by mutagenesis, homology modeling, and competition studies with positive allosteric modulator (PAM). From the inhibitory effects of NAM S37a on: 1) chimeras with swapped ectodomain, 2) stepwise N-terminal truncations, 3) distinct constitutively active mutations distributed across the hinge region and ECL, but not across the TMD, we conclude that S37a binds at the ectodomain/TMD interface, between the converging helix, ECL1, and the IA. This is also supported by the noncompetitive inhibition of PAM-C2-activation by S37a in the TSHR-TMD construct lacking the ectodomain. Mutagenesis studies on the IA and ECL were guided by our refined model of the ectodomain/TMD interface and indicate an interaction with the TSHR-specific residues E404 (preceding IA) and H478 (ECL1). At this new allosteric interaction site, NAM S37a blocks both TSH- and PAM-induced activation of the TSHR. Our refined models, mutations, and new allosteric binding pocket helped us to gain more detailed insights into the intramolecular course of TSHR activation at the ectodomain/TMD interface, including the delocalization of the converging helix and rearrangement of the conformation of IA. These changes are embedded between the ECL and cooperatively trigger active conformations of TMD. SIGNIFICANCE STATEMENT: The intramolecular activation mechanisms of the TSHR appear to be distinct from those of other G protein-coupled receptors, as the TSHR has a uniquely large N-terminal ectodomain that includes the hormone binding site and an internal agonist sequence. We present new molecular and structural insights into the interface between ectodomain and transmembrane domain in the TSHR, as well as the transfer of activation to the transmembrane domain. This knowledge is critical for understanding activation or inhibition of the receptor by allosteric ligands. We have identified a new allosteric antagonist binding pocket that is located exactly at this interface and possesses specific features that may allow the generation of potent highly TSHR-selective drugs, of potential value for the treatment of Graves' orbitopathy.


Sujet(s)
Récepteur TSH/composition chimique , Récepteur TSH/métabolisme , Thyréostimuline/métabolisme , Régulation allostérique , Régulation de l'expression des gènes , Cellules HEK293 , Humains , Modèles moléculaires , Mutation , Domaines protéiques , Récepteur TSH/génétique , Similitude de séquences d'acides aminés , Transduction du signal
5.
Sci Rep ; 9(1): 10878, 2019 07 26.
Article de Anglais | MEDLINE | ID: mdl-31350436

RÉSUMÉ

As a first host barrier, the skin is constantly exposed to environmental insults that perturb its integrity. Tight regulation of skin homeostasis is largely controlled by the aryl hydrocarbon receptor (AhR). Here, we demonstrate that Henna and its major pigment, the naphthoquinone Lawsone activate AhR, both in vitro and in vivo. In human keratinocytes and epidermis equivalents, Lawsone exposure enhances the production of late epidermal proteins, impacts keratinocyte differentiation and proliferation, and regulates skin inflammation. To determine the potential use of Lawsone for therapeutic application, we harnessed human, murine and zebrafish models. In skin regeneration models, Lawsone interferes with physiological tissue regeneration and inhibits wound healing. Conversely, in a human acute dermatitis model, topical application of a Lawsone-containing cream ameliorates skin irritation. Altogether, our study reveals how a widely used natural plant pigment is sensed by the host receptor AhR, and how the physiopathological context determines beneficial and detrimental outcomes.


Sujet(s)
Dermatite/traitement médicamenteux , Kératinocytes/métabolisme , Naphtoquinones/métabolisme , Récepteurs à hydrocarbure aromatique/métabolisme , Peau/métabolisme , Animaux , Cellules cultivées , Régénération tissulaire guidée , Homéostasie , Humains , Lawsonia (plante) , Souris , Modèles animaux , Naphtoquinones/usage thérapeutique , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Cicatrisation de plaie , Danio zébré
6.
Thyroid ; 29(1): 111-123, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30351237

RÉSUMÉ

BACKGROUND: The thyrotropin receptor (TSHR) is the target for autoimmune thyroid stimulating antibodies (TSAb) triggering hyperthyroidism. Whereas elevated thyroid hormone synthesis by the thyroid in Graves' disease can be treated by antithyroid agents, for the pathogenic activation of TSHR in retro-orbital fibroblasts of the eye, leading to Graves' orbitopathy (GO), no causal TSHR directed therapy is available. METHODS: Due to the therapeutic gap for severe GO, TSHR inhibitors were identified by high-throughput screening in Chinese hamster ovary cells expressing the TSHR. Stereo-selective synthesis of the screening hits led to the molecule S37, which contains seven chiral centers. Enantiomeric separation of the molecule S37 resulted in the enantiopure molecule S37a-a micro-molar antagonist of thyrotropin-induced cyclic adenosine monophosphate accumulation in HEK 293 cells expressing the TSHR. RESULTS: The unique rigid bent shape of molecule S37a may mediate the observed high TSHR selectivity. Most importantly, the closely related follitropin and lutropin receptors were not affected by this compound. S37a not only inhibits the TSHR activation by thyrotropin itself but also activation by monoclonal TSAb M22 (human), KSAb1 (murine), and the allosteric small-molecule agonist C2. Disease-related ex vivo studies in HEK 293 cells expressing the TSHR showed that S37a also inhibits cyclic adenosine monophosphate formation by oligoclonal TSAb, which are highly enriched in GO patients' sera. Initial in vivo pharmacokinetic studies revealed no toxicity of S37a and a remarkable 53% oral bioavailability in mice. CONCLUSION: In summary, a novel highly selective inhibitor for the TSHR is presented, which has promising potential for further development for the treatment of GO.


Sujet(s)
Ophtalmopathie basedowienne/traitement médicamenteux , Antihormones/pharmacologie , Récepteur TSH/antagonistes et inhibiteurs , Animaux , Cellules CHO , Cricetinae , Cricetulus , Fibroblastes/effets des médicaments et des substances chimiques , Cellules HEK293 , Antihormones/usage thérapeutique , Humains , Transduction du signal/effets des médicaments et des substances chimiques
7.
Chembiochem ; 18(16): 1639-1649, 2017 08 17.
Article de Anglais | MEDLINE | ID: mdl-28557180

RÉSUMÉ

Unbiased chemoproteomic profiling of small-molecule interactions with endogenous proteins is important for drug discovery. For meaningful results, all protein classes have to be tractable, including G protein-coupled receptors (GPCRs). These receptors are hardly tractable by affinity pulldown from lysates. We report a capture compound (CC)-based strategy to target and identify GPCRs directly from living cells. We synthesized CCs with sertindole attached to the CC scaffold in different orientations to target the dopamine D2 receptor (DRD2) heterologously expressed in HEK 293 cells. The structure-activity relationship of sertindole for DRD2 binding was reflected in the activities of the sertindole CCs in radioligand displacement, cell-based assays, and capture compound mass spectrometry (CCMS). The activity pattern was rationalized by molecular modelling. The most-active CC showed activities very similar to that of unmodified sertindole. A concentration of DRD2 in living cells well below 100 fmol used as an experimental input was sufficient for unambiguous identification of captured DRD2 by mass spectrometry. Our new CCMS workflow broadens the arsenal of chemoproteomic technologies to close a critical gap for the comprehensive characterization of drug-protein interactions.


Sujet(s)
Antagonistes du récepteur D2 de la dopamine/composition chimique , Imidazoles/composition chimique , Indoles/composition chimique , Récepteur D2 de la dopamine/analyse , Animaux , Antagonistes du récepteur D2 de la dopamine/synthèse chimique , Antagonistes du récepteur D2 de la dopamine/effets des radiations , Cellules HEK293 , Humains , Imidazoles/synthèse chimique , Imidazoles/effets des radiations , Indoles/synthèse chimique , Indoles/effets des radiations , Ligands , Simulation de docking moléculaire , Dosage par compétition , Rats , Récepteur D2 de la dopamine/effets des radiations , Spipérone/composition chimique , Relation structure-activité , Suidae , Spectrométrie de masse en tandem , Rayons ultraviolets
8.
Mol Cell Endocrinol ; 443: 163-174, 2017 03 05.
Article de Anglais | MEDLINE | ID: mdl-28108384

RÉSUMÉ

The L-type amino acid transporter 2 (LAT2) imports amino acids (AA) and also certain thyroid hormones (TH), e.g. 3,3'-T2 and T3, but not rT3 and T4. We utilized LAT2 mutations (Y130A, N133S, F242W) that increase 3,3'-T2 import and focus here on import and export capacity for AA, T4, T3, BCH and derivatives thereof to delineate molecular features. Transport studies and analysis of competitive inhibition of import by radiolabelled TH and AA were performed in Xenopus laevis oocytes. Only Y130A, a pocket widening mutation, enabled import for T4 and increased it for T3. Mutant F242W showed increased 3,3'-T2 import but no import rates for other TH derivatives. No export was detected for any TH by LAT2-wild type (WT). Mutations Y130A and N133S enabled only the export of 3,3'-T2, while N133S also increased AA export. Thus, distinct molecular LAT2-features determine bidirectional AA transport but only an unidirectional 3,3'-T2 and T3 import.


Sujet(s)
Système y+ de transport d'acides aminés/génétique , Système y+ de transport d'acides aminés/métabolisme , Acides aminés/métabolisme , Chaines légères de l'antigène CD98/génétique , Chaines légères de l'antigène CD98/métabolisme , Hormones thyroïdiennes/métabolisme , Acides aminés cycliques/pharmacologie , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Membrane cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/métabolisme , Di-iodothyronines/métabolisme , Antigènes CD98/métabolisme , Protéines à fluorescence verte/métabolisme , Cellules HEK293 , Humains , Iode/métabolisme , Cinétique , Souris , Modèles biologiques , Modèles moléculaires , Protéines mutantes/métabolisme , Mutation/génétique , Ovocytes/effets des médicaments et des substances chimiques , Ovocytes/métabolisme , Multimérisation de protéines , Spécificité du substrat/effets des médicaments et des substances chimiques , Xenopus laevis/métabolisme
9.
Article de Anglais | MEDLINE | ID: mdl-26441830

RÉSUMÉ

The human lutropin (hLH)/choriogonadotropin (hCG) receptor (LHCGR) can be activated by binding two slightly different gonadotropic glycoprotein hormones, choriogonadotropin (CG) - secreted by the placenta, and lutropin (LH) - produced by the pituitary. They induce different signaling profiles at the LHCGR. This cannot be explained by binding to the receptor's leucine-rich-repeat domain (LRRD), as this binding is similar for the two hormones. We therefore speculate that there are previously unknown differences in the hormone/receptor interaction at the extracellular hinge region, which might help to understand functional differences between the two hormones. We have therefore performed a detailed study of the binding and action of LH and CG at the LHCGR hinge region. We focused on a primate-specific additional exon in the hinge region, which is located between LRRD and the serpentine domain. The segment of the hinge region encoded by exon10 was previously reported to be only relevant to hLH signaling, as the exon10-deletion receptor exhibits decreased hLH signaling, but unchanged hCG signaling. We designed an advanced homology model of the hormone/LHCGR complex, followed by experimental characterization of relevant fragments in the hinge region. In addition, we examined predictions of a helical exon10-encoded conformation by block-wise polyalanine (helix supporting) mutations. These helix preserving modifications showed no effect on hormone-induced signaling. However, introduction of a structure-disturbing double-proline mutant LHCGR-Q303P/E305P within the exon10-helix has, in contrast to exon10-deletion, no impact on hLH, but only on hCG signaling. This opposite effect on signaling by hLH and hCG can be explained by distinct sites of hormone interaction in the hinge region. In conclusion, our analysis provides details of the differences between hLH- and hCG-induced signaling that are mainly determined in the L2-beta loop of the hormones and in the hinge region of the receptor.

10.
Nature ; 512(7515): 387-92, 2014 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-25119038

RÉSUMÉ

The aryl hydrocarbon receptor (AhR) is a highly conserved ligand-dependent transcription factor that senses environmental toxins and endogenous ligands, thereby inducing detoxifying enzymes and modulating immune cell differentiation and responses. We hypothesized that AhR evolved to sense not only environmental pollutants but also microbial insults. We characterized bacterial pigmented virulence factors, namely the phenazines from Pseudomonas aeruginosa and the naphthoquinone phthiocol from Mycobacterium tuberculosis, as ligands of AhR. Upon ligand binding, AhR activation leads to virulence factor degradation and regulated cytokine and chemokine production. The relevance of AhR to host defence is underlined by heightened susceptibility of AhR-deficient mice to both P. aeruginosa and M. tuberculosis. Thus, we demonstrate that AhR senses distinct bacterial virulence factors and controls antibacterial responses, supporting a previously unidentified role for AhR as an intracellular pattern recognition receptor, and identify bacterial pigments as a new class of pathogen-associated molecular patterns.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Mycobacterium tuberculosis/immunologie , Pigments biologiques/métabolisme , Pseudomonas aeruginosa/immunologie , Récepteurs à hydrocarbure aromatique/métabolisme , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , Animaux , Antibactériens/métabolisme , Cellules de la moelle osseuse/cytologie , Cytokines/immunologie , Cytokines/métabolisme , Rétrocontrôle physiologique , Humains , Ligands , Activation des macrophages , Souris , Mycobacterium tuberculosis/croissance et développement , Mycobacterium tuberculosis/métabolisme , Phénazines/métabolisme , Pigments biologiques/composition chimique , Infections à Pseudomonas/métabolisme , Pseudomonas aeruginosa/métabolisme , Pyocyanine/métabolisme , Facteurs de virulence/composition chimique , Facteurs de virulence/métabolisme
11.
J Biol Chem ; 289(35): 24250-62, 2014 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-24966326

RÉSUMÉ

G protein-coupled receptors (GPCRs) represent the most important drug targets. Although the smallest functional unit of a GPCR is a monomer, it became clear in the past decades that the vast majority of the receptors form dimers. Only very recently, however, data were presented that some receptors may in fact be expressed as a mixture of monomers and dimers and that the interaction of the receptor protomers is dynamic. To date, equilibrium measurements were restricted to the plasma membrane due to experimental limitations. We have addressed the question as to where this equilibrium is established for the corticotropin-releasing factor receptor type 1. By developing a novel approach to analyze single molecule fluorescence cross-correlation spectroscopy data for intracellular membrane compartments, we show that the corticotropin-releasing factor receptor type 1 has a specific monomer/dimer equilibrium that is already established in the endoplasmic reticulum (ER). It remains constant at the plasma membrane even following receptor activation. Moreover, we demonstrate for seven additional GPCRs that they are expressed in specific but substantially different monomer/dimer ratios. Although it is well known that proteins may dimerize in the ER in principle, our data show that the ER is also able to establish the specific monomer/dimer ratios of GPCRs, which sheds new light on the functions of this compartment.


Sujet(s)
Réticulum endoplasmique/métabolisme , Récepteur CRH/métabolisme , Animaux , Membrane cellulaire/métabolisme , Dimérisation , Cellules HEK293 , Humains , Rats , Récepteur CRH/composition chimique
12.
FEBS J ; 281(5): 1479-1492, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24438591

RÉSUMÉ

The human lutropin/choriogonadotropin receptor (hLHR) for the gonadotropic hormones human luteinizing hormone (hLH; lutropin) and human choriogonadotropin (hCG) is crucial for normal sexual development and fertility. We aimed to unravel differences between the two hLHR hormones in molecular activation mechanisms at hLHR. We utilized a specific hLHR variant that lacks exon 10 (hLHR-delExon10), which maintains full cAMP signaling by hCG, but decreases hLH-induced receptor signaling, resulting in a pathogenic phenotype. Exon 10 encodes 27 amino acids within the hinge region, which is an extracellular segment that is important for signaling and hormone interaction. Initially, we assumed that the lack of exon 10 might disturb intermolecular trans-activation of hLH, a mechanism that has been reported for hCG at hLHR. Coexpression of signaling-deficient hLHR and binding-deficient hLHR can be used to examine the mechanisms of receptor signaling, in particular intermolecular cooperation and intramolecular cis-activation. Therefore, hLHR-delExon10 was combined with the hLHR Lys605→Glu mutant, in which signaling is abolished, and the hLHR mutant Cys131→Arg, in which binding is deficient. We found that hCG signaling was partially rescued, indicating trans-activation. However, the hLH signal could not be restored via forced trans-activation with any construct. Fluorescence cross-correlation spectroscopy detected oligomerization in all combinations, indicating that these functional differences cannot be explained by monomerization of hLHR-delExon10. Thus, our data demonstrate not only that the different behavior of hLH at hLHR-delExon10 is unlikely to be related to modified intermolecular receptor activation, but also that hLH may exclusively stimulate the targeted hLHR by cis-activation, whereas hCG is also capable of inducing trans-activation.


Sujet(s)
Gonadotrophine chorionique/métabolisme , Hormone lutéinisante/métabolisme , Récepteur LH/métabolisme , Substitution d'acide aminé , Membrane cellulaire/métabolisme , AMP cyclique/métabolisme , Exons , Cellules HEK293 , Humains , Mutagenèse dirigée , Protéines mutantes/composition chimique , Protéines mutantes/génétique , Protéines mutantes/métabolisme , Structure quaternaire des protéines , Récepteur LH/composition chimique , Récepteur LH/génétique , Délétion de séquence , Transduction du signal , Activation de la transcription
13.
J Am Soc Nephrol ; 24(5): 744-58, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23559583

RÉSUMÉ

In the principal cells of the renal collecting duct, arginine vasopressin (AVP) stimulates the synthesis of cAMP, leading to signaling events that culminate in the phosphorylation of aquaporin-2 water channels and their redistribution from intracellular domains to the plasma membrane via vesicular trafficking. The molecular mechanisms that control aquaporin-2 trafficking and the consequent water reabsorption, however, are not completely understood. Here, we used a cell-based assay and automated immunofluorescence microscopy to screen 17,700 small molecules for inhibitors of the cAMP-dependent redistribution of aquaporin-2. This approach identified 17 inhibitors, including 4-acetyldiphyllin, a selective blocker of vacuolar H(+)-ATPase that increases the pH of intracellular vesicles and causes accumulation of aquaporin-2 in the Golgi compartment. Although 4-acetyldiphyllin did not inhibit forskolin-induced increases in cAMP formation and downstream activation of protein kinase A (PKA), it did prevent cAMP/PKA-dependent phosphorylation at serine 256 of aquaporin-2, which triggers the redistribution to the plasma membrane. It did not, however, prevent cAMP-induced changes to the phosphorylation status at serines 261 or 269. Last, we identified the fungicide fluconazole as an inhibitor of cAMP-mediated redistribution of aquaporin-2, but its target in this pathway remains unknown. In conclusion, our screening approach provides a method to begin dissecting molecular mechanisms underlying AVP-mediated water reabsorption, evidenced by our identification of 4-acetyldiphyllin as a modulator of aquaporin-2 trafficking.


Sujet(s)
Aquaporine-2/métabolisme , Benzodioxoles/pharmacologie , Benzofuranes/pharmacologie , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Animaux , Cellules cultivées , Colforsine/pharmacologie , AMP cyclique/physiologie , Fluconazole/pharmacologie , Appareil de Golgi/métabolisme , Tests de criblage à haut débit , Humains , Souris , Microscopie de fluorescence , Phosphorylation , Transport des protéines/effets des médicaments et des substances chimiques , Rats
14.
FEBS Lett ; 586(16): 2351-9, 2012 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-22659187

RÉSUMÉ

The protease-activated receptor 1 (PAR1) is activated by thrombin cleavage releasing the physiologically-relevant parstatin peptide (residues 1-41). However, the actual length of parstatin was unclear since the receptor may also possess a cleavable signal peptide (residues 1-21) according to prediction programs. Here, we show that this putative signal peptide is indeed functional and removed from the PAR1 resolving the question of parstatin length. Moreover, we show that the sequence encoding the signal peptide may surprisingly play a role in stabilization of the PAR1 mRNA, a function which would be novel for a G protein-coupled receptor.


Sujet(s)
Régulation de l'expression des gènes , Signaux de triage des protéines , Récepteur de type PAR-1/physiologie , ADN complémentaire/métabolisme , Cellules HEK293 , Humains , Microscopie confocale/méthodes , Microscopie de fluorescence/méthodes , Conformation d'acide nucléique , Fragments peptidiques/composition chimique , Plasmides/métabolisme , Structure tertiaire des protéines , ARN messager/métabolisme , Récepteur de type PAR-1/composition chimique , Récepteurs couplés aux protéines G/métabolisme , Protéines de fusion recombinantes/composition chimique , Thrombine/composition chimique
15.
FEBS Lett ; 586(6): 784-91, 2012 Mar 23.
Article de Anglais | MEDLINE | ID: mdl-22306118

RÉSUMÉ

In this study we demonstrate that the photoconvertible monomeric Kikume green-red (mKikGR) protein is suitable to study trafficking of G protein-coupled receptors. Taking mKikGR-tagged mutants of the vasopressin V(2) receptor (V(2)R) as models, we analyzed whether the V(2)R-specific pharmacological chaperone SR121463B influences receptor folding on a co- or post-translational level. Misfolded mKikGR-tagged V(2)Rs were completely photoconverted in the early secretory pathway yielding a red receptor population (already synthesized receptors) and an arising green receptor population (newly synthesized receptors). Trafficking of both receptor populations could be rescued by treatment with SR121463B demonstrating that the substance can act co- and post-translationally.


Sujet(s)
Protéines luminescentes/métabolisme , Chaperons moléculaires/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Séquence d'acides aminés , Antagonistes des récepteurs de l'hormone antidiurétique , Couleur , Cellules HEK293 , Humains , Protéines luminescentes/génétique , Chaperons moléculaires/composition chimique , Données de séquences moléculaires , Morpholines/composition chimique , Morpholines/métabolisme , Pliage des protéines , Récepteurs couplés aux protéines G/composition chimique , Récepteurs couplés aux protéines G/génétique , Récepteurs à la vasopressine/composition chimique , Récepteurs à la vasopressine/génétique , Récepteurs à la vasopressine/métabolisme , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Spiranes/composition chimique , Spiranes/métabolisme
16.
J Biol Chem ; 286(41): 35588-35600, 2011 Oct 14.
Article de Anglais | MEDLINE | ID: mdl-21808059

RÉSUMÉ

The specific inhibition of the biosynthesis of target proteins is a relatively novel strategy in pharmacology and is based mainly on antisense approaches (e.g. antisense oligonucleotides or RNA interference). Recently, a novel class of substances was described acting at a later step of protein biosynthesis. The cyclic heptadepsipeptides CAM741 and cotransin were shown to inhibit selectively the biosynthesis of a small subset of secretory proteins by preventing stable insertion of the nascent chains into the Sec61 translocon complex at the endoplasmic reticulum membrane (Besemer, J., Harant, H., Wang, S., Oberhauser, B., Marquardt, K., Foster, C. A., Schreiner, E. P., de Vries, J. E., Dascher-Nadel, C., and Lindley, I. J. (2005) Nature 436, 290-293; Garrison, J. L., Kunkel, E. J., Hegde, R. S., and Taunton, J. (2005) Nature 436, 285-289). These peptides act in a signal sequence-discriminatory manner, which explains their selectivity. Here, we have analyzed the cotransin sensitivity of various G protein-coupled receptors in transfected HEK 293 cells. We show that the biosynthesis of the human endothelin B receptor (ET(B)R) is highly sensitive to cotransin, in contrast to that of the other G protein-coupled receptors analyzed. Using a novel biosynthesis assay based on fusions with the photoconvertible Kaede protein, we show that the IC(50) value of cotransin action on ET(B)R biosynthesis is 5.4 µm and that ET(B)R signaling could be completely blocked by treating cells with 30 µm cotransin. Taken together, our data add an integral membrane protein, namely the ET(B)R, to the small group of cotransin-sensitive proteins.


Sujet(s)
Peptides cycliques/pharmacologie , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Récepteur de l'endothéline de type B/biosynthèse , Cellules HEK293 , Humains , Protéines luminescentes/biosynthèse , Protéines luminescentes/génétique , Biosynthèse des protéines/génétique , Récepteur de l'endothéline de type B/génétique , Protéines de fusion recombinantes/biosynthèse , Protéines de fusion recombinantes/génétique
17.
J Biol Chem ; 286(29): 25859-71, 2011 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-21586576

RÉSUMÉ

Transmembrane helices (TMHs) 5 and 6 are known to be important for signal transduction by G-protein-coupled receptors (GPCRs). Our aim was to characterize the interface between TMH5 and TMH6 of the thyrotropin receptor (TSHR) to gain molecular insights into aspects of signal transduction and regulation. A proline at TMH5 position 5.50 is highly conserved in family A GPCRs and causes a twist in the helix structure. Mutation of the TSHR-specific alanine (Ala-5935·5°) at this position to proline resulted in a 20-fold reduction of cell surface expression. This indicates that TMH5 in the TSHR might have a conformation different from most other family A GPCRs by forming a regular α-helix. Furthermore, linking our own and previous data from directed mutagenesis with structural information led to suggestions of distinct pairs of interacting residues between TMH5 and TMH6 that are responsible for stabilizing either the basal or the active state. Our insights suggest that the inactive state conformation is constrained by a core set of polar interactions among TMHs 2, 3, 6, and 7 and in contrast that the active state conformation is stabilized mainly by non-polar interactions between TMHs 5 and 6. Our findings might be relevant for all family A GPCRs as supported by a statistical analysis of residue properties between the TMHs of a vast number of GPCR sequences.


Sujet(s)
Membrane cellulaire/métabolisme , Récepteurs couplés aux protéines G/composition chimique , Récepteurs couplés aux protéines G/métabolisme , Récepteur TSH/composition chimique , Récepteur TSH/métabolisme , Transduction du signal , Animaux , Séquence conservée , AMP cyclique/métabolisme , Cellules HEK293 , Humains , Inositol phosphates/métabolisme , Modèles moléculaires , Mutagenèse dirigée , Mutation , Structure secondaire des protéines , Récepteur TSH/génétique
18.
J Biol Chem ; 286(11): 9079-96, 2011 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-21177871

RÉSUMÉ

A-kinase anchoring proteins (AKAPs) tether protein kinase A (PKA) and other signaling proteins to defined intracellular sites, thereby establishing compartmentalized cAMP signaling. AKAP-PKA interactions play key roles in various cellular processes, including the regulation of cardiac myocyte contractility. We discovered small molecules, 3,3'-diamino-4,4'-dihydroxydiphenylmethane (FMP-API-1) and its derivatives, which inhibit AKAP-PKA interactions in vitro and in cultured cardiac myocytes. The molecules bind to an allosteric site of regulatory subunits of PKA identifying a hitherto unrecognized region that controls AKAP-PKA interactions. FMP-API-1 also activates PKA. The net effect of FMP-API-1 is a selective interference with compartmentalized cAMP signaling. In cardiac myocytes, FMP-API-1 reveals a novel mechanism involved in terminating ß-adrenoreceptor-induced cAMP synthesis. In addition, FMP-API-1 leads to an increase in contractility of cultured rat cardiac myocytes and intact hearts. Thus, FMP-API-1 represents not only a novel means to study compartmentalized cAMP/PKA signaling but, due to its effects on cardiac myocytes and intact hearts, provides the basis for a new concept in the treatment of chronic heart failure.


Sujet(s)
Protéines d'ancrage aux protéines kinases A/métabolisme , Cyclic AMP-Dependent Protein Kinases/antagonistes et inhibiteurs , Cyclic AMP-Dependent Protein Kinases/métabolisme , AMP cyclique/métabolisme , Myocytes cardiaques/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Systèmes de seconds messagers/physiologie , Protéines d'ancrage aux protéines kinases A/génétique , Régulation allostérique/effets des médicaments et des substances chimiques , Régulation allostérique/génétique , Animaux , Maladie chronique , AMP cyclique/génétique , Cyclic AMP-Dependent Protein Kinases/génétique , Activation enzymatique/effets des médicaments et des substances chimiques , Activation enzymatique/génétique , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Mâle , Contraction myocardique/effets des médicaments et des substances chimiques , Rats , Rats de lignée WKY , Systèmes de seconds messagers/effets des médicaments et des substances chimiques
19.
Cell Mol Life Sci ; 68(1): 159-67, 2011 Jan.
Article de Anglais | MEDLINE | ID: mdl-20652618

RÉSUMÉ

The thyrotropin receptor (TSHR) exhibits elevated cAMP signaling in the basal state and becomes fully activated by thyrotropin. Previously we presented evidence that small-molecule ligands act allosterically within the transmembrane region in contrast to the orthosteric extracellular hormone-binding sites. Our goal in this study was to identify positions that surround the allosteric pocket and that are sensitive for inactivation of TSHR. Homology modeling combined with site-directed mutagenesis and functional characterization revealed seven mutants located in the allosteric binding site that led to a decrease of basal cAMP signaling activity. The majority of these silencing mutations, which constrain the TSHR in an inactive conformation, are found in two clusters when mapped onto the 3D structural model. We suggest that the amino acid positions identified herein are indicating locations where small-molecule antagonists, both neutral antagonists and inverse agonists, might interfere with active TSHR conformations.


Sujet(s)
Mutation , Récepteur TSH/génétique , Transduction du signal/génétique , Sites de fixation , AMP cyclique/métabolisme , Cellules HEK293 , Humains , Ligands , Modèles moléculaires , Structure tertiaire des protéines/génétique , Récepteur TSH/composition chimique , Récepteur TSH/physiologie
20.
J Biol Chem ; 285(43): 32878-32887, 2010 Oct 22.
Article de Anglais | MEDLINE | ID: mdl-20682782

RÉSUMÉ

The corticotropin-releasing factor receptor type 2a (CRF(2(a))R) belongs to the family of G protein-coupled receptors. The receptor possesses an N-terminal pseudo signal peptide that is unable to mediate targeting of the nascent chain to the endoplasmic reticulum membrane during early receptor biogenesis. The pseudo signal peptide remains uncleaved and consequently forms an additional hydrophobic receptor domain with unknown function that is unique within the large G protein-coupled receptor protein family. Here, we have analyzed the functional significance of this domain in comparison with the conventional signal peptide of the homologous corticotropin-releasing factor receptor type 1 (CRF(1)R). We show that the presence of the pseudo signal peptide leads to a very low cell surface receptor expression of the CRF(2(a))R in comparison with the CRF(1)R. Moreover, whereas the presence of the pseudo signal peptide did not affect coupling to the G(s) protein, G(i)-mediated inhibition of adenylyl cyclase activity was abolished. The properties mediated by the pseudo signal peptide were entirely transferable to the CRF(1)R in signal peptide exchange experiments. Taken together, our results show that signal peptides do not only influence early protein biogenesis. In the case of the corticotropin-releasing factor receptor subtypes, the use of conventional and pseudo signal peptides have an unexpected influence on signal transduction.


Sujet(s)
Adenylate Cyclase/métabolisme , Sous-unités alpha Gi-Go des protéines G/métabolisme , Régulation de l'expression des gènes/physiologie , Signaux de triage des protéines , Récepteur CRH/biosynthèse , Transduction du signal/physiologie , Adenylate Cyclase/génétique , Animaux , Lignée cellulaire , Sous-unités alpha Gi-Go des protéines G/génétique , Humains , Rats , Récepteur CRH/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...