Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Metab ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871982

ABSTRACT

Incretin-based therapies are highly successful in combatting obesity and type 2 diabetes1. Yet both activation and inhibition of the glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) in combination with glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) activation have resulted in similar clinical outcomes, as demonstrated by the GIPR-GLP-1R co-agonist tirzepatide2 and AMG-133 (ref. 3) combining GIPR antagonism with GLP-1R agonism. This underlines the importance of a better understanding of the GIP system. Here we show the necessity of ß-arrestin recruitment for GIPR function, by combining in vitro pharmacological characterization of 47 GIPR variants with burden testing of clinical phenotypes and in vivo studies. Burden testing of variants with distinct ligand-binding capacity, Gs activation (cyclic adenosine monophosphate production) and ß-arrestin 2 recruitment and internalization shows that unlike variants solely impaired in Gs signalling, variants impaired in both Gs and ß-arrestin 2 recruitment contribute to lower adiposity-related traits. Endosomal Gs-mediated signalling of the variants shows a ß-arrestin dependency and genetic ablation of ß-arrestin 2 impairs cyclic adenosine monophosphate production and decreases GIP efficacy on glucose control in male mice. This study highlights a crucial impact of ß-arrestins in regulating GIPR signalling and overall preservation of biological activity that may facilitate new developments in therapeutic targeting of the GIPR system.

2.
Sci Signal ; 17(841): eadi4747, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38889226

ABSTRACT

G protein-coupled receptors (GPCRs) regulate cellular signaling processes by coupling to diverse combinations of heterotrimeric G proteins composed of Gα, Gß, and Gγ subunits. Biosensors based on bioluminescence resonance energy transfer (BRET) have advanced our understanding of GPCR functional selectivity. Some BRET biosensors monitor ligand-induced conformational changes in the receptor or G proteins, whereas others monitor the recruitment of downstream effectors to sites of G protein activation. Here, we compared the ability of conformation-and activation-based BRET biosensors to assess the coupling of various class A and B GPCRs to specific Gα proteins in cultured cells. These GPCRs included serotonin 5-HT2A and 5-HT7 receptors, the GLP-1 receptor (GLP-1R), and the M3 muscarinic receptor. We observed different signaling profiles between the two types of sensors, highlighting how data interpretation could be affected by the nature of the biosensor. We also found that the identity of the Gßγ subunits used in the assay could differentially influence the selectivity of a receptor toward Gα subtypes, emphasizing the importance of the receptor-Gßγ pairing in determining Gα coupling specificity. Last, the addition of epitope tags to the receptor could affect stoichiometry and coupling selectivity and yield artifactual findings. These results highlight the need for careful sensor selection and experimental design when probing GPCR-G protein coupling.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques , Biosensing Techniques , Receptors, G-Protein-Coupled , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/chemistry , Bioluminescence Resonance Energy Transfer Techniques/methods , HEK293 Cells , Biosensing Techniques/methods , Protein Conformation , Signal Transduction , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics
3.
Mol Metab ; 85: 101931, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796310

ABSTRACT

OBJECTIVE: Simultaneous activation of ß2- and ß3-adrenoceptors (ARs) improves whole-body metabolism via beneficial effects in skeletal muscle and brown adipose tissue (BAT). Nevertheless, high-efficacy agonists simultaneously targeting these receptors whilst limiting activation of ß1-ARs - and thus inducing cardiovascular complications - are currently non-existent. Therefore, we here developed and evaluated the therapeutic potential of a novel ß2-and ß3-AR, named ATR-127, for the treatment of obesity and its associated metabolic perturbations in preclinical models. METHODS: In the developmental phase, we assessed the impact of ATR-127's on cAMP accumulation in relation to the non-selective ß-AR agonist isoprenaline across various rodent ß-AR subtypes, including neonatal rat cardiomyocytes. Following these experiments, L6 muscle cells were stimulated with ATR-127 to assess the impact on GLUT4-mediated glucose uptake and intramyocellular cAMP accumulation. Additionally, in vitro, and in vivo assessments are conducted to measure ATR-127's effects on BAT glucose uptake and thermogenesis. Finally, diet-induced obese mice were treated with 5 mg/kg ATR-127 for 21 days to investigate the effects on glucose homeostasis, body weight, fat mass, skeletal muscle glucose uptake, BAT thermogenesis and hepatic steatosis. RESULTS: Exposure of L6 muscle cells to ATR-127 robustly enhanced GLUT4-mediated glucose uptake despite low intramyocellular cAMP accumulation. Similarly, ATR-127 markedly increased BAT glucose uptake and thermogenesis both in vitro and in vivo. Prolonged treatment of diet-induced obese mice with ATR-127 dramatically improved glucose homeostasis, an effect accompanied by decreases in body weight and fat mass. These effects were paralleled by an enhanced skeletal muscle glucose uptake, BAT thermogenesis, and improvements in hepatic steatosis. CONCLUSIONS: Our results demonstrate that ATR-127 is a highly effective, novel ß2- and ß3-ARs agonist holding great therapeutic promise for the treatment of obesity and its comorbidities, whilst potentially limiting cardiovascular complications. As such, the therapeutic effects of ATR-127 should be investigated in more detail in clinical studies.


Subject(s)
Adipose Tissue, Brown , Mice, Inbred C57BL , Muscle, Skeletal , Animals , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Male , Rats , Obesity/metabolism , Obesity/drug therapy , Fatty Liver/metabolism , Fatty Liver/drug therapy , Thermogenesis/drug effects , Adrenergic Agonists/pharmacology
4.
Nat Microbiol ; 9(6): 1499-1512, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38548922

ABSTRACT

Climate change and population densities accelerated transmission of highly pathogenic viruses to humans, including the Crimean-Congo haemorrhagic fever virus (CCHFV). Here we report that the Low Density Lipoprotein Receptor (LDLR) is a critical receptor for CCHFV cell entry, playing a vital role in CCHFV infection in cell culture and blood vessel organoids. The interaction between CCHFV and LDLR is highly specific, with other members of the LDLR protein family failing to bind to or neutralize the virus. Biosensor experiments demonstrate that LDLR specifically binds the surface glycoproteins of CCHFV. Importantly, mice lacking LDLR exhibit a delay in CCHFV-induced disease. Furthermore, we identified the presence of Apolipoprotein E (ApoE) on CCHFV particles. Our findings highlight the essential role of LDLR in CCHFV infection, irrespective of ApoE presence, when the virus is produced in tick cells. This discovery holds profound implications for the development of future therapies against CCHFV.


Subject(s)
Apolipoproteins E , Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Receptors, LDL , Virus Internalization , Animals , Humans , Mice , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever, Crimean/virology , Hemorrhagic Fever, Crimean/metabolism , Mice, Knockout , Receptors, LDL/metabolism , Receptors, LDL/genetics , Receptors, Virus/metabolism , Ticks/virology , Ticks/metabolism
5.
Nat Commun ; 14(1): 6243, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37813859

ABSTRACT

G protein-coupled receptors are important drug targets that engage and activate signaling transducers in multiple cellular compartments. Delineating therapeutic signaling from signaling associated with adverse events is an important step towards rational drug design. The glucagon-like peptide-1 receptor (GLP-1R) is a validated target for the treatment of diabetes and obesity, but drugs that target this receptor are a frequent cause of adverse events. Using recently developed biosensors, we explored the ability of GLP-1R to activate 15 pathways in 4 cellular compartments and demonstrate that modifications aimed at improving the therapeutic potential of GLP-1R agonists greatly influence compound efficacy, potency, and safety in a pathway- and compartment-selective manner. These findings, together with comparative structure analysis, time-lapse microscopy, and phosphoproteomics, reveal unique signaling signatures for GLP-1R agonists at the level of receptor conformation, functional selectivity, and location bias, thus associating signaling neighborhoods with functionally distinct cellular outcomes and clinical consequences.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Glucagon-Like Peptide-1 Receptor , Incretins , Humans , Glucagon-Like Peptide-1 Receptor/metabolism , Incretins/adverse effects , Signal Transduction
8.
Curr Opin Pharmacol ; 59: 11-18, 2021 08.
Article in English | MEDLINE | ID: mdl-34029832

ABSTRACT

Since the discovery of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019, intense research efforts on an unprecedented scale have focused on the study of viral entry mechanisms and adaptive immunity. While the identification of angiotensin-converting enzyme 2 (ACE2) and other co-receptors has elucidated the molecular and structural basis for viral entry, the pathobiological mechanisms of SARS-CoV-2 in human tissues are less understood. Recent advances in bioengineering have opened opportunities for the use of organotypic human tissue models to investigate host-virus interactions and test antiviral drug candidates in a physiological context. Although it is too early to accurately quantify the added value of these systems compared with conventional cell systems, it can be assumed that these advanced three-dimensional (3D) models contribute toward improved result translation. This mini-review summarizes recent work to study SARS-CoV-2 infection in human 3D tissue models with an emphasis on the pharmacological tools that have been developed to understand and prevent viral entry and replication.


Subject(s)
COVID-19 Drug Treatment , Models, Biological , SARS-CoV-2 , Drug Development , Humans , Intestines , Kidney , Liver , Organoids , Respiratory System
9.
Curr Opin Struct Biol ; 69: 142-149, 2021 08.
Article in English | MEDLINE | ID: mdl-34048988

ABSTRACT

It should come as no surprise that G protein-coupled receptors (GPCRs) continue to occupy the focus of drug discovery efforts. Their widespread expression and broad role in signal transduction underline their importance in human physiology. Despite more than 800 GPCRs sharing a common architecture, unique differences govern ligand specificity and pathway selectivity. From the relatively simplified view offered by classical radioligand binding assays and contractility responses in organ baths, the road from ligand binding to biological action has become more and more complex as we learn about the molecular mediators that underly GPCR activation and translate it to physiological outcomes. In particular, the development of biosensors has evolved over the years to dissect the capacity of a given receptor to activate individual pathways. Here, we discuss how recent biosensor development has reinforced the idea that biased signaling may become mainstream in drug discovery programs.


Subject(s)
Biosensing Techniques , Receptors, G-Protein-Coupled , Drug Discovery , Humans , Ligands , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33990469

ABSTRACT

G protein-coupled receptors (GPCRs) are gatekeepers of cellular homeostasis and the targets of a large proportion of drugs. In addition to their signaling activity at the plasma membrane, it has been proposed that their actions may result from translocation and activation of G proteins at endomembranes-namely endosomes. This could have a significant impact on our understanding of how signals from GPCR-targeting drugs are propagated within the cell. However, little is known about the mechanisms that drive G protein movement and activation in subcellular compartments. Using bioluminescence resonance energy transfer (BRET)-based effector membrane translocation assays, we dissected the mechanisms underlying endosomal Gq trafficking and activity following activation of Gq-coupled receptors, including the angiotensin II type 1, bradykinin B2, oxytocin, thromboxane A2 alpha isoform, and muscarinic acetylcholine M3 receptors. Our data reveal that GPCR-promoted activation of Gq at the plasma membrane induces its translocation to endosomes independently of ß-arrestin engagement and receptor endocytosis. In contrast, Gq activity at endosomes was found to rely on both receptor endocytosis-dependent and -independent mechanisms. In addition to shedding light on the molecular processes controlling subcellular Gq signaling, our study provides a set of tools that will be generally applicable to the study of G protein translocation and activation at endosomes and other subcellular organelles, as well as the contribution of signal propagation to drug action.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Endocytosis/physiology , Endosomes/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Receptors, G-Protein-Coupled/physiology , HEK293 Cells , Humans , Rho Guanine Nucleotide Exchange Factors/physiology , Signal Transduction/physiology , beta-Arrestins/physiology
11.
Biochim Biophys Acta Mol Cell Res ; 1867(12): 118849, 2020 12.
Article in English | MEDLINE | ID: mdl-32916203

ABSTRACT

FPR2, a member of the family of G protein-coupled receptors (GPCRs), mediates neutrophil migration, a response that has been linked to ß-arrestin recruitment. ß-Arrestin regulates GPCR endocytosis and can also elicit non-canonical receptor signaling. To determine the poorly understood role of ß-arrestin in FPR2 endocytosis and in NADPH-oxidase activation in neutrophils, Barbadin was used as a research tool in this study. Barbadin has been shown to bind the clathrin adaptor protein (AP2) and thereby prevent ß-arrestin/AP2 interaction and ß-arrestin-mediated GPCR endocytosis. In agreement with this, AP2/ß-arrestin interaction induced by an FPR2-specific agonist was inhibited by Barbadin. Unexpectedly, however, Barbadin did not inhibit FPR2 endocytosis, indicating that a mechanism independent of ß-arrestin/AP2 interaction may sustain FPR2 endocytosis. This was confirmed by the fact, that FPR2 also underwent agonist-promoted endocytosis in ß-arrestin deficient cells, albeit at a diminished level as compared to wild type cells. Dissection of the Barbadin effects on FPR2-mediated neutrophil functions including NADPH-oxidase activation mediated release of reactive oxygen species (ROS) and chemotaxis revealed that Barbadin had no effect on chemotactic migration whereas the release of ROS was potentiated/primed. The effect of Barbadin on ROS production was reversible, independent of ß-arrestin recruitment, and similar to that induced by latrunculin A. Taken together, our data demonstrate that endocytic uptake of FPR2 occurs independently of ß-arrestin, while Barbadin selectively augments FPR2-mediated ROS production independently of receptor endocytosis. Given that Barbadin binds to AP2 and prevents the AP2/ß-arrestin interaction, our results indicate a role for AP2 in FPR2-mediated ROS release from neutrophils.


Subject(s)
Endocytosis/genetics , Pyrimidines/pharmacology , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , beta-Arrestin 1/genetics , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/genetics , Clathrin/chemistry , Endocytosis/drug effects , HEK293 Cells , Humans , NADPH Oxidases/genetics , Neutrophils/drug effects , Protein Binding/drug effects , Pyrimidines/chemistry , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Lipoxin/chemistry , Signal Transduction/drug effects , beta-Arrestin 1/chemistry
12.
Proc Natl Acad Sci U S A ; 117(35): 21723-21730, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817560

ABSTRACT

G proteins are activated when they associate with G protein-coupled receptors (GPCRs), often in response to agonist-mediated receptor activation. It is generally thought that agonist-induced receptor-G protein association necessarily promotes G protein activation and, conversely, that activated GPCRs do not interact with G proteins that they do not activate. Here we show that GPCRs can form agonist-dependent complexes with G proteins that they do not activate. Using cell-based bioluminescence resonance energy transfer (BRET) and luminescence assays we find that vasopressin V2 receptors (V2R) associate with both Gs and G12 heterotrimers when stimulated with the agonist arginine vasopressin (AVP). However, unlike V2R-Gs complexes, V2R-G12 complexes are not destabilized by guanine nucleotides and do not promote G12 activation. Activating V2R does not lead to signaling responses downstream of G12 activation, but instead inhibits basal G12-mediated signaling, presumably by sequestering G12 heterotrimers. Overexpressing G12 inhibits G protein receptor kinase (GRK) and arrestin recruitment to V2R and receptor internalization. Formyl peptide (FPR1 and FPR2) and Smoothened (Smo) receptors also form complexes with G12 that are insensitive to nucleotides, suggesting that unproductive GPCR-G12 complexes are not unique to V2R. These results indicate that agonist-dependent receptor-G protein association does not always lead to G protein activation and may in fact inhibit G protein activation.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Bioluminescence Resonance Energy Transfer Techniques/methods , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/physiology , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/physiology , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Protein Binding/physiology , Receptors, Vasopressin/metabolism , Signal Transduction/physiology , Vasopressins/metabolism , beta-Arrestins/metabolism
13.
Nat Commun ; 10(1): 667, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737406

ABSTRACT

Class F receptors are considered valuable therapeutic targets due to their role in human disease, but structural changes accompanying receptor activation remain unexplored. Employing population and cancer genomics data, structural analyses, molecular dynamics simulations, resonance energy transfer-based approaches and mutagenesis, we identify a conserved basic amino acid in TM6 in Class F receptors that acts as a molecular switch to mediate receptor activation. Across all tested Class F receptors (FZD4,5,6,7, SMO), mutation of the molecular switch confers an increased potency of agonists by stabilizing an active conformation as assessed by engineered mini G proteins as conformational sensors. Disruption of the switch abrogates the functional interaction between FZDs and the phosphoprotein Dishevelled, supporting conformational selection as a prerequisite for functional selectivity. Our studies reveal the molecular basis of a common activation mechanism conserved in all Class F receptors, which facilitates assay development and future discovery of Class F receptor-targeting drugs.


Subject(s)
Models, Theoretical , Molecular Dynamics Simulation , Receptors, G-Protein-Coupled/metabolism , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Models, Molecular , Phylogeny , Protein Binding , Protein Conformation , Receptors, G-Protein-Coupled/genetics
14.
Sci Signal ; 11(559)2018 12 04.
Article in English | MEDLINE | ID: mdl-30514810

ABSTRACT

Frizzleds (FZDs) are a group of seven transmembrane-spanning (7TM) receptors that belong to class F of the G protein-coupled receptor (GPCR) superfamily. FZDs bind WNT proteins to stimulate diverse signaling cascades involved in embryonic development, stem cell regulation, and adult tissue homeostasis. Frizzled 5 (FZD5) is one of the most studied class F GPCRs that promote the functional inactivation of the ß-catenin destruction complex in response to WNTs. However, whether FZDs function as prototypical GPCRs has been heavily debated and, in particular, FZD5 has not been shown to activate heterotrimeric G proteins. Here, we show that FZD5 exhibited a conformational change after the addition of WNT-5A, which is reminiscent of class A and class B GPCR activation. In addition, we performed several live-cell imaging and spectrometric-based approaches, such as dual-color fluorescence recovery after photobleaching (dcFRAP) and resonance energy transfer (RET)-based assays that demonstrated that FZD5 activated Gαq and its downstream effectors upon stimulation with WNT-5A. Together, these findings suggest that FZD5 is a 7TM receptor with a bona fide GPCR activation profile and suggest novel targets for drug discovery in WNT-FZD signaling.


Subject(s)
Cell Proliferation , Frizzled Receptors/metabolism , Pancreatic Neoplasms/pathology , Wnt-5a Protein/metabolism , Calcium/metabolism , Diglycerides/metabolism , Frizzled Receptors/chemistry , HEK293 Cells , Humans , Pancreatic Neoplasms/metabolism , Protein Conformation , Protein Kinase C/metabolism , Signal Transduction , Tumor Cells, Cultured , Wnt-5a Protein/chemistry
15.
Trends Pharmacol Sci ; 39(9): 828-842, 2018 09.
Article in English | MEDLINE | ID: mdl-30049420

ABSTRACT

For more than 30 years, WNT/ß-catenin and planar cell polarity signaling has formed the basis for what we understand to be the primary output of the interaction between WNTs and their cognate receptors known as Frizzleds (FZDs). In the shadow of these pathways, evidence for the involvement of heterotrimeric G proteins in WNT signaling has grown substantially over the years - redefining the complexity of the WNT signaling network. Moreover, the distinct characteristics of FZD paralogs are becoming better understood, and we can now apply concepts valid for classical GPCRs to grasp FZDs as molecular machines at the interface of ligand binding and intracellular effects. This review discusses recent developments in the field of WNT/FZD signaling in the context of GPCR pharmacology, and identifies remaining mysteries with an emphasis on structural and kinetic components that support this dogma shift.


Subject(s)
Frizzled Receptors/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Frizzled Receptors/chemistry , Humans , Models, Molecular , Receptors, G-Protein-Coupled/chemistry , Wnt Signaling Pathway
16.
Nat Commun ; 8(1): 226, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28790300

ABSTRACT

G protein-coupled receptors (GPCRs) constitute the largest family of cell surface receptors. They can exist and act as dimers, but the requirement of dimers for agonist-induced signal initiation and structural dynamics remains largely unknown. Frizzled 6 (FZD6) is a member of Class F GPCRs, which bind WNT proteins to initiate signaling. Here, we show that FZD6 dimerizes and that the dimer interface of FZD6 is formed by the transmembrane α-helices four and five. Most importantly, we present the agonist-induced dissociation/re-association of a GPCR dimer through the use of live cell imaging techniques. Further analysis of a dimerization-impaired FZD6 mutant indicates that dimer dissociation is an integral part of FZD6 signaling to extracellular signal-regulated kinases1/2. The discovery of agonist-dependent dynamics of dimers as an intrinsic process of receptor activation extends our understanding of Class F and other dimerizing GPCRs, offering novel targets for dimer-interfering small molecules.Frizzled 6 (FZD6) is a G protein-coupled receptor (GPCR) involved in several cellular processes. Here, the authors use live cell imaging and spectroscopy to show that FZD6 forms dimers, whose association is regulated by WNT proteins and that dimer dissociation is crucial for FZD6 signaling.


Subject(s)
Frizzled Receptors/metabolism , Dimerization , HEK293 Cells , Humans , Wnt-5a Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...