Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Commun Biol ; 7(1): 501, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664468

ABSTRACT

G protein-coupled receptors naturally oscillate between inactive and active states, often resulting in receptor constitutive activity with important physiological consequences. Among the class C G protein-coupled receptors that typically sense amino-acids and their derivatives, the calcium sensing receptor (CaSR) tightly controls blood calcium levels. Its constitutive activity has not yet been studied. Here, we demonstrate the importance of the inter-subunit disulfide bridges in maintaining the inactive state of CaSR, resulting in undetectable constitutive activity, unlike the other class C receptors. Deletion of these disulfide bridges results in strong constitutive activity that is abolished by mutations preventing amino acid binding. It shows that this inter-subunit disulfide link is necessary to limit the agonist effect of amino acids on CaSR. Furthermore, human genetic mutations deleting these bridges and associated with hypocalcemia result in elevated CaSR constitutive activity. These results highlight the physiological importance of fine tuning the constitutive activity of G protein-coupled receptors.


Subject(s)
Disulfides , Receptors, Calcium-Sensing , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/genetics , Humans , Disulfides/metabolism , Disulfides/chemistry , HEK293 Cells , Calcium/metabolism , Mutation , Animals
2.
Nat Commun ; 15(1): 1990, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38443355

ABSTRACT

G protein-coupled receptors (GPCRs) constitute the largest family of membrane proteins and are important drug targets. The discovery of drugs targeting these receptors and their G protein signaling properties are based on assays mainly performed with modified receptors expressed in heterologous cells. However, GPCR responses may differ in their native environment. Here, by using highly sensitive Gi/o sensors, we reveal specific properties of Gi/o protein-mediated responses triggered by GABAB, α2 adrenergic and cannabinoid CB1 receptors in primary neurons, different from those in heterologous cells. These include different profiles in the Gi/o protein subtypes-mediated responses, and differences in the potencies of some ligands even at similar receptor expression levels. Altogether, our results show the importance of using biosensors compatible with primary cells for evaluating the activities of endogenous GPCRs in their native environment.


Subject(s)
Neurons , Receptors, G-Protein-Coupled , Signal Transduction , Adrenergic Agents , Biological Assay , GTP-Binding Protein alpha Subunits, Gi-Go
3.
J Chem Inf Model ; 64(11): 4436-4461, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38423996

ABSTRACT

The world has witnessed a revolution in therapeutics with the development of biological medicines such as antibodies and antibody fragments, notably nanobodies. These nanobodies possess unique characteristics including high specificity and modulatory activity, making them promising candidates for therapeutic applications. Identifying their binding mode is essential for their development. Experimental structural techniques are effective to get such information, but they are expensive and time-consuming. Here, we propose a computational approach, aiming to identify the epitope of a nanobody that acts as an agonist and a positive allosteric modulator at the rat metabotropic glutamate receptor 5. We employed multiple structure modeling tools, including various artificial intelligence algorithms for epitope mapping. The computationally identified epitope was experimentally validated, confirming the success of our approach. Additional dynamics studies provided further insights on the modulatory activity of the nanobody. The employed methodologies and approaches initiate a discussion on the efficacy of diverse techniques for epitope mapping and later nanobody engineering.


Subject(s)
Deep Learning , Epitopes , Receptor, Metabotropic Glutamate 5 , Single-Domain Antibodies , Receptor, Metabotropic Glutamate 5/chemistry , Receptor, Metabotropic Glutamate 5/metabolism , Receptor, Metabotropic Glutamate 5/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Epitopes/immunology , Epitopes/chemistry , Animals , Rats , Models, Molecular , Epitope Mapping/methods , Molecular Dynamics Simulation , Protein Conformation
4.
Sci Adv ; 10(4): eadg1679, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38277461

ABSTRACT

Metabotropic glutamate receptor 2 (mGlu2) attracts particular attention as a possible target for a new class of antipsychotics. However, the signaling pathways transducing the effects of mGlu2 in the brain remain poorly characterized. Here, we addressed this issue by identifying native mGlu2 interactome in mouse prefrontal cortex. Nanobody-based affinity purification and mass spectrometry identified 149 candidate mGlu2 partners, including the neurotrophin receptor TrkB. The later interaction was confirmed both in cultured cells and prefrontal cortex. mGlu2 activation triggers phosphorylation of TrkB on Tyr816 in primary cortical neurons and prefrontal cortex. Reciprocally, TrkB stimulation enhances mGlu2-operated Gi/o protein activation. Furthermore, TrkB inhibition prevents the rescue of behavioral deficits by glutamatergic antipsychotics in phencyclidine-treated mice. Collectively, these results reveal a cross-talk between TrkB and mGlu2, which is key to the behavioral response to glutamatergic antipsychotics.


Subject(s)
Antipsychotic Agents , Mice , Animals , Antipsychotic Agents/pharmacology , Receptor, trkB/metabolism , Prefrontal Cortex/metabolism , Cells, Cultured , Neurons/metabolism
5.
Sci Adv ; 9(22): eadf1378, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37267369

ABSTRACT

Allosteric modulators bear great potential to fine-tune neurotransmitter action. Promising targets are metabotropic glutamate (mGlu) receptors, which are associated with numerous brain diseases. Orthosteric and allosteric ligands act in synergy to control the activity of these multidomain dimeric GPCRs. Here, we analyzed the effect of such molecules on the concerted conformational changes of full-length mGlu2 at the single-molecule level. We first established FRET sensors through genetic code expansion combined with click chemistry to monitor conformational changes on live cells. We then used single-molecule FRET and show that orthosteric agonist binding leads to the stabilization of most of the glutamate binding domains in their closed state, while the reorientation of the dimer into the active state remains partial. Allosteric modulators, interacting with the transmembrane domain, are required to stabilize the fully reoriented active dimer. These results illustrate how concerted conformational changes within multidomain proteins control their activity, and how these are modulated by allosteric ligands.


Subject(s)
Receptors, Metabotropic Glutamate , Allosteric Regulation , Ligands , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Glutamates
6.
Am J Physiol Cell Physiol ; 325(1): C79-C89, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37184233

ABSTRACT

G protein-coupled receptors (GPCRs) represent the largest family of membrane proteins and are important drug targets. GPCRs are allosteric machines that transduce an extracellular signal to the cell by activating heterotrimeric G proteins. Herein, we summarize the recent advancements in the molecular activation mechanism of the γ-aminobutyric acid type B (GABAB) and metabotropic glutamate (mGlu) receptors, the most important class C GPCRs that modulate synaptic transmission in the brain. Both are mandatory dimers, this quaternary structure being needed for their function The structures of these receptors in different conformations and in complexes with G proteins have revealed their asymmetric activation. This asymmetry is further highlighted by the recent discovery of mGlu heterodimers, where the eight mGlu subunits can form specific and functional heterodimers. Finally, the development of allosteric modulators has revealed new possibilities for regulating the function of these receptors by targeting the transmembrane dimer interface. This family of receptors never ceases to astonish and serve as models to better understand the diversity and asymmetric functioning of GPCRs.NEW & NOTEWORTHY γ-aminobutyric acid type B (GABAB) and metabotropic glutamate (mGlu) receptors form constitutive dimers, which are required for their function. They serve as models to better understand the diversity and activation of G protein-coupled receptors (GPCRs). The structures of these receptors in different conformations and in complexes with G proteins have revealed their asymmetric activation. This asymmetry is further highlighted by the recent discovery of specific and functional mGlu heterodimers. Allosteric modulators can be developed to target the transmembrane interface and modulate the asymmetry.


Subject(s)
Receptors, Metabotropic Glutamate , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Receptors, G-Protein-Coupled , Synaptic Transmission , Glutamic Acid , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism
7.
Int J Mol Sci ; 24(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36768953

ABSTRACT

Treatments for central nervous system diseases with therapeutic antibodies have been increasingly investigated over the last decades, leading to some approved monoclonal antibodies for brain disease therapies. The detection of biomarkers for diagnosis purposes with non-invasive antibody-based imaging approaches has also been explored in brain cancers. However, antibodies generally display a low capability of reaching the brain, as they do not efficiently cross the blood-brain barrier. As an alternative, recent studies have focused on single-domain antibodies (sdAbs) that correspond to the antigen-binding fragment. While some reports indicate that the brain uptake of these small antibodies is still low, the number of studies reporting brain-penetrating sdAbs is increasing. In this review, we provide an overview of methods used to assess or evaluate brain penetration of sdAbs and discuss the pros and cons that could affect the identification of brain-penetrating sdAbs of therapeutic or diagnostic interest.


Subject(s)
Single-Domain Antibodies , Diagnostic Imaging , Brain
8.
Elife ; 122023 01 23.
Article in English | MEDLINE | ID: mdl-36688536

ABSTRACT

Amyloid-ß precursor protein (APP) regulates neuronal activity through the release of secreted APP (sAPP) acting at cell surface receptors. APP and sAPP were reported to bind to the extracellular sushi domain 1 (SD1) of GABAB receptors (GBRs). A 17 amino acid peptide (APP17) derived from APP was sufficient for SD1 binding and shown to mimic the inhibitory effect of sAPP on neurotransmitter release and neuronal activity. The functional effects of APP17 and sAPP were similar to those of the GBR agonist baclofen and blocked by a GBR antagonist. These experiments led to the proposal that sAPP activates GBRs to exert its neuronal effects. However, whether APP17 and sAPP influence classical GBR signaling pathways in heterologous cells was not analyzed. Here, we confirm that APP17 binds to GBRs with nanomolar affinity. However, biochemical and electrophysiological experiments indicate that APP17 does not influence GBR activity in heterologous cells. Moreover, APP17 did not regulate synaptic GBR localization, GBR-activated K+ currents, neurotransmitter release, or neuronal activity in vitro or in vivo. Our results show that APP17 is not a functional GBR ligand and indicate that sAPP exerts its neuronal effects through receptors other than GBRs.


Subject(s)
Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , gamma-Aminobutyric Acid/metabolism
9.
Nat Commun ; 13(1): 6365, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289206

ABSTRACT

G protein-coupled receptors (GPCRs) are important drug targets that mediate various signaling pathways by activating G proteins and engaging ß-arrestin proteins. Despite its importance for the development of therapeutics with fewer side effects, the underlying mechanism that controls the balance between these signaling modes of GPCRs remains largely unclear. Here, we show that assembly into dimers and oligomers can largely influence the signaling mode of the platelet-activating factor receptor (PAFR). Single-particle analysis results show that PAFR can form oligomers at low densities through two possible dimer interfaces. Stabilization of PAFR oligomers through cross-linking increases G protein activity, and decreases ß-arrestin recruitment and agonist-induced internalization significantly. Reciprocally, ß-arrestin prevents PAFR oligomerization. Our results highlight a mechanism involved in the control of receptor signaling, and thereby provide important insights into the relationship between GPCR oligomerization and downstream signaling.


Subject(s)
Platelet Activating Factor , Receptors, G-Protein-Coupled , Platelet Activating Factor/metabolism , Platelet Activating Factor/pharmacology , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism , Signal Transduction , beta-Arrestin 1/metabolism , GTP-Binding Proteins/metabolism
10.
Nat Chem Biol ; 18(8): 894-903, 2022 08.
Article in English | MEDLINE | ID: mdl-35681029

ABSTRACT

Membrane proteins, including ion channels, receptors and transporters, are often composed of multiple subunits and can form large complexes. Their specific composition in native tissues is difficult to determine and remains largely unknown. In this study, we developed a method for determining the subunit composition of endogenous cell surface protein complexes from isolated native tissues. Our method relies on nanobody-based sensors, which enable proximity detection between subunits in time-resolved Förster resonance energy transfer (FRET) measurements. Additionally, given conformation-specific nanobodies, the activation of these complexes can be recorded in native brain tissue. Applied to the metabotropic glutamate receptors in different brain regions, this approach revealed the clear existence of functional metabotropic glutamate (mGlu)2-mGlu4 heterodimers in addition to mGlu2 and mGlu4 homodimers. Strikingly, the mGlu4 subunits appear to be mainly heterodimers in the brain. Overall, these versatile biosensors can determine the presence and activity of endogenous membrane proteins in native tissues with high fidelity and convenience.


Subject(s)
Glutamic Acid , Receptors, Metabotropic Glutamate , Brain/metabolism , Fluorescence Resonance Energy Transfer/methods , Receptors, Metabotropic Glutamate/metabolism
12.
Elife ; 102021 12 06.
Article in English | MEDLINE | ID: mdl-34866572

ABSTRACT

G protein-coupled receptors (GPCRs) are among the most promising drug targets. They often form homo- and heterodimers with allosteric cross-talk between receptor entities, which contributes to fine-tuning of transmembrane signaling. Specifically controlling the activity of GPCR dimers with ligands is a good approach to clarify their physiological roles and validate them as drug targets. Here, we examined the mode of action of positive allosteric modulators (PAMs) that bind at the interface of the transmembrane domains of the heterodimeric GABAB receptor. Our site-directed mutagenesis results show that mutations of this interface impact the function of the three PAMs tested. The data support the inference that they act at the active interface between both transmembrane domains, the binding site involving residues of the TM6s of the GABAB1 and the GABAB2 subunit. Importantly, the agonist activity of these PAMs involves a key region in the central core of the GABAB2 transmembrane domain, which also controls the constitutive activity of the GABAB receptor. This region corresponds to the sodium ion binding site in class A GPCRs that controls the basal state of the receptors. Overall, these data reveal the possibility of developing allosteric compounds able to specifically modulate the activity of GPCR homo- and heterodimers by acting at their transmembrane interface.


Subject(s)
Receptors, GABA-B/genetics , Allosteric Regulation , HEK293 Cells , Humans , Ligands , Protein Domains , Receptors, GABA-B/metabolism
13.
iScience ; 24(11): 103311, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34778730

ABSTRACT

Neurotransmitter receptors are involved in cancer progression. Among them, the heterodimeric GABAB receptor, activated by the main inhibitory neurotransmitter GABA, is composed of the transmembrane GABAB1 and GABAB2 subunits. The oncogenic role of the isoform GABAB1e (GB1e) containing only the extracellular domain of GABAB1 remains unclear. We revealed that GB1e is largely expressed in human breast cancer (BrCa) cell lines as well as in BrCa tissues where it is upregulated. Moreover, GB1e promoted the malignancy of BrCa cells both in vitro and in vivo. We propose that GB1e favors EGFR signaling by interacting with PTPN12 to disrupt the interaction between EGFR and PTPN12, and phosphorylation of Y230 and Y404 on GB1e is required in this process. Our data highlight that the GABBR1 gene through the expression of the GB1e isoform might play an important oncogenic role in BrCa and that GB1e is of interest for the treatment of some cancers.

14.
Sci Signal ; 14(705): eaaz4112, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34665640

ABSTRACT

G protein­coupled receptors (GPCRs) activate various mitogen-activated protein kinase (MAPK) pathways to regulate critical cell functions. ß-Arrestins mediate this mechanism for most GPCRs but not the GABAB receptor (GABABR). When coupled to the G protein Gi/o, GABABR phosphorylates the kinases ERK1 and ERK2. Here, we uncovered a distinct ß-arrestin­independent mechanism of MAPK pathway activation by GABABR. We found that GABABR also phosphorylated the kinase JNK downstream of activation of the small guanosine triphosphatases (GTPases) RhoA and Rac1 in primary mouse neurons. However, instead of Gi/o proteins, activation of this RhoA/Rac1-JNK pathway was mediated by G13. This pathway promoted the phosphorylation and accumulation of the postsynaptic scaffolding protein PSD95 and GABABR-mediated neuroprotection in granule neurons. In addition, this pathway synergized with a previously reported GABABR-mediated neuroprotection mediated by a Gi/o-dependent mechanism. GABABR agonists activated G13 with slower kinetics and lower potency than with which they activated Gi/o. Our findings reveal distinct, ß-arrestin­independent, context-specific synergistic mechanisms of MAPK activation by G protein­mediated GPCR signaling.


Subject(s)
Neuroprotection , Receptors, GABA-B , gamma-Aminobutyric Acid
15.
Nat Commun ; 12(1): 5426, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521824

ABSTRACT

Much hope in drug development comes from the discovery of positive allosteric modulators (PAM) that display target subtype selectivity and act by increasing agonist potency and efficacy. How such compounds can allosterically influence agonist action remains unclear. Metabotropic glutamate receptors (mGlu) are G protein-coupled receptors that represent promising targets for brain diseases, and for which PAMs acting in the transmembrane domain have been developed. Here, we explore the effect of a PAM on the structural dynamics of mGlu2 in optimized detergent micelles using single molecule FRET at submillisecond timescales. We show that glutamate only partially stabilizes the extracellular domains in the active state. Full activation is only observed in the presence of a PAM or the Gi protein. Our results provide important insights on the role of allosteric modulators in mGlu activation, by stabilizing the active state of a receptor that is otherwise rapidly oscillating between active and inactive states.


Subject(s)
Glutamic Acid/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/chemistry , Allosteric Regulation/drug effects , Allosteric Site , Amino Acids/chemistry , Amino Acids/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Catalytic Domain , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholesterol Esters/chemistry , Cholesterol Esters/pharmacology , Diosgenin/analogs & derivatives , Diosgenin/chemistry , Diosgenin/pharmacology , Disaccharides/chemistry , Disaccharides/pharmacology , Fluorescence Resonance Energy Transfer , Gene Expression , Glucosides/chemistry , Glucosides/pharmacology , Glycolipids/chemistry , Glycolipids/pharmacology , HEK293 Cells , Humans , Indans/chemistry , Indans/pharmacology , Micelles , Octoxynol/chemistry , Octoxynol/pharmacology , Protein Binding , Protein Conformation , Protein Multimerization , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single Molecule Imaging , Xanthenes/chemistry , Xanthenes/pharmacology
16.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Article in English | MEDLINE | ID: mdl-34385321

ABSTRACT

There is growing interest in developing biologics due to their high target selectivity. The G protein-coupled homo- and heterodimeric metabotropic glutamate (mGlu) receptors regulate many synapses and are promising targets for the treatment of numerous brain diseases. Although subtype-selective allosteric small molecules have been reported, their effects on the recently discovered heterodimeric receptors are often not known. Here, we describe a nanobody that specifically and fully activates homodimeric human mGlu4 receptors. Molecular modeling and mutagenesis studies revealed that the nanobody acts by stabilizing the closed active state of the glutamate binding domain by interacting with both lobes. In contrast, this nanobody does not activate the heterodimeric mGlu2-4 but acts as a pure positive allosteric modulator. These data further reveal how an antibody can fully activate a class C receptor and bring further evidence that nanobodies represent an alternative way to specifically control mGlu receptor subtypes.


Subject(s)
Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Single-Domain Antibodies , Gene Expression Regulation/drug effects , Humans , Models, Biological , Mutation , Protein Binding , Protein Conformation , Receptors, Metabotropic Glutamate/genetics
17.
Nature ; 594(7864): 589-593, 2021 06.
Article in English | MEDLINE | ID: mdl-34135509

ABSTRACT

The metabotropic glutamate receptors (mGlus) are involved in the modulation of synaptic transmission and neuronal excitability in the central nervous system1. These receptors probably exist as both homo- and heterodimers that have unique pharmacological and functional properties2-4. Here we report four cryo-electron microscopy structures of the human mGlu subtypes mGlu2 and mGlu7, including inactive mGlu2 and mGlu7 homodimers; mGlu2 homodimer bound to an agonist and a positive allosteric modulator; and inactive mGlu2-mGlu7 heterodimer. We observed a subtype-dependent dimerization mode for these mGlus, as a unique dimer interface that is mediated by helix IV (and that is important for limiting receptor activity) exists only in the inactive mGlu2 structure. The structures provide molecular details of the inter- and intra-subunit conformational changes that are required for receptor activation, which distinguish class C G-protein-coupled receptors from those in classes A and B. Furthermore, our structure and functional studies of the mGlu2-mGlu7 heterodimer suggest that the mGlu7 subunit has a dominant role in controlling dimeric association and G-protein activation in the heterodimer. These insights into mGlu homo- and heterodimers highlight the complex landscape of mGlu dimerization and activation.


Subject(s)
Receptors, Metabotropic Glutamate/chemistry , Cryoelectron Microscopy , Humans , Protein Multimerization , Protein Structure, Tertiary
18.
Nature ; 594(7864): 594-598, 2021 06.
Article in English | MEDLINE | ID: mdl-33911284

ABSTRACT

G-protein-coupled receptors (GPCRs) have central roles in intercellular communication1,2. Structural studies have revealed how GPCRs can activate G proteins. However, whether this mechanism is conserved among all classes of GPCR remains unknown. Here we report the structure of the class-C heterodimeric GABAB receptor, which is activated by the inhibitory transmitter GABA, in its active form complexed with Gi1 protein. We found that a single G protein interacts with the GB2 subunit of the GABAB receptor at a site that mainly involves intracellular loop 2 on the side of the transmembrane domain. This is in contrast to the G protein binding in a central cavity, as has been observed with other classes of GPCR. This binding mode results from the active form of the transmembrane domain of this GABAB receptor being different from that of other GPCRs, as it shows no outside movement of transmembrane helix 6. Our work also provides details of the inter- and intra-subunit changes that link agonist binding to G-protein activation in this heterodimeric complex.


Subject(s)
GTP-Binding Proteins/chemistry , Receptors, GABA-B/chemistry , Cryoelectron Microscopy , Humans , Protein Binding , Protein Domains , Protein Multimerization , Protein Structure, Tertiary
19.
FASEB J ; 34(12): 16348-16363, 2020 12.
Article in English | MEDLINE | ID: mdl-33058267

ABSTRACT

Cell surface trafficking of many G protein-coupled receptors is tightly regulated. Among them, the mandatory heterodimer GABAB receptor for the main inhibitory neurotransmitter, γ-aminobutyric acid (GABA), is a model. In mammals, its cell surface trafficking is highly controlled by an endoplasmic reticulum retention signal in the C-terminal intracellular region of the GB1 subunit that is masked through a coiled-coil interaction with the GB2 subunit. Here, we investigate the molecular basis for the export of its homolog in Drosophila melanogaster that regulates the circadian rhythm and sleep. In contrast to mammals, the endoplasmic retention signal is carried by GB2, while GB1 reaches the cell surface alone. NMR analysis showed that the coiled-coil domain that controls GABAB heterodimer formation is structurally conserved between flies and mammals, despite specific features. These findings show the adaptation of a similar quality control system during evolution for maintaining the subunit composition of a functional heterodimeric receptor.


Subject(s)
Receptors, GABA/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Circadian Rhythm/physiology , Dimerization , Drosophila melanogaster/metabolism , Endoplasmic Reticulum/metabolism , Fishes/metabolism , HEK293 Cells , Humans , Mammals/metabolism , Protein Subunits , Protein Transport/physiology , Quality Control , Rats , Sleep/physiology , gamma-Aminobutyric Acid/metabolism
20.
Proc Natl Acad Sci U S A ; 117(35): 21711-21722, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817431

ABSTRACT

Many membrane receptors are regulated by nutrients. However, how these nutrients control a single receptor remains unknown, even in the case of the well-studied calcium-sensing receptor CaSR, which is regulated by multiple factors, including ions and amino acids. Here, we developed an innovative cell-free Förster resonance energy transfer (FRET)-based conformational CaSR biosensor to clarify the main conformational changes associated with activation. By allowing a perfect control of ambient nutrients, this assay revealed that Ca2+ alone fully stabilizes the active conformation, while amino acids behave as pure positive allosteric modulators. Based on the identification of Ca2+ activation sites, we propose a molecular basis for how these different ligands cooperate to control CaSR activation. Our results provide important information on CaSR function and improve our understanding of the effects of genetic mutations responsible for human diseases. They also provide insights into how a receptor can integrate signals from various nutrients to better adapt to the cell response.


Subject(s)
Calcium/metabolism , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/ultrastructure , Allosteric Regulation/physiology , Binding Sites/genetics , Calcium/physiology , Fluorescence Resonance Energy Transfer/methods , Humans , Ligands , Molecular Conformation , Receptors, Calcium-Sensing/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...