Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
Cell ; 185(21): 3950-3965.e25, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36170854

ABSTRACT

The G protein-coupled receptor cascade leading to production of the second messenger cAMP is replete with pharmacologically targetable proteins, with the exception of the Gα subunit, Gαs. GTPases remain largely undruggable given the difficulty of displacing high-affinity guanine nucleotides and the lack of other drug binding sites. We explored a chemical library of 1012 cyclic peptides to expand the chemical search for inhibitors of this enzyme class. We identified two macrocyclic peptides, GN13 and GD20, that antagonize the active and inactive states of Gαs, respectively. Both macrocyclic peptides fine-tune Gαs activity with high nucleotide-binding-state selectivity and G protein class-specificity. Co-crystal structures reveal that GN13 and GD20 distinguish the conformational differences within the switch II/α3 pocket. Cell-permeable analogs of GN13 and GD20 modulate Gαs/GƟƎĀ³ signaling in cells through binding to crystallographically defined pockets. The discovery of cyclic peptide inhibitors targeting Gαs provides a path for further development of state-dependent GTPase inhibitors.


Subject(s)
Peptides , Receptors, G-Protein-Coupled , GTP Phosphohydrolases , Guanine Nucleotides , Nucleotides , Peptides/chemistry , Peptides, Cyclic/pharmacology
2.
Cell ; 185(24): 4560-4573.e19, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36368322

ABSTRACT

Binding of arrestin to phosphorylated G protein-coupled receptors (GPCRs) is crucial for modulating signaling. Once internalized, some GPCRs remain complexed with Ɵ-arrestins, while others interact only transiently; this difference affects GPCR signaling and recycling. Cell-based and inĀ vitro biophysical assays reveal the role of membrane phosphoinositides (PIPs) in Ɵ-arrestin recruitment and GPCR-Ɵ-arrestin complexĀ dynamics. We find that GPCRs broadly stratify into two groups, one that requires PIP binding for Ɵ-arrestin recruitment and one that does not. Plasma membrane PIPs potentiate an active conformation of Ɵ-arrestin and stabilize GPCR-Ɵ-arrestin complexes by promoting a fully engaged state of the complex. As allosteric modulators of GPCR-Ɵ-arrestin complex dynamics, membrane PIPs allow for additional conformational diversity beyond that imposed by GPCR phosphorylation alone. For GPCRs that require membrane PIP binding for Ɵ-arrestin recruitment, this provides a mechanism for Ɵ-arrestin release upon translocation of the GPCR to endosomes, allowing for its rapid recycling.


Subject(s)
Arrestins , Phosphatidylinositols , beta-Arrestins/metabolism , Phosphatidylinositols/metabolism , Arrestins/metabolism , beta-Arrestin 1/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
Annu Rev Biochem ; 90: 709-737, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33606955

ABSTRACT

Intricate relationships between endocytosis and cellular signaling, first recognized nearly 40 years ago through the study of tyrosine kinase growth factor receptors, are now known to exist for multiple receptor classes and to affect myriad physiological and developmental processes. This review summarizes our present understanding of how endocytosis orchestrates cellular signaling networks, with an emphasis on mechanistic underpinnings and focusing on two receptor classes-tyrosine kinase and G protein-coupled receptors-that have been investigated in particular detail. Together, these examples provide a useful survey of the current consensus, uncertainties, and controversies in this rapidly advancing area of cell biology.


Subject(s)
Endocytosis/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Membrane/metabolism , Endosomes/metabolism , Humans , Lysosomes/metabolism , Protein Transport , Signal Transduction
4.
Cell ; 174(3): 505-520, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30053424

ABSTRACT

Although gene discovery in neuropsychiatric disorders, including autism spectrum disorder, intellectual disability, epilepsy, schizophrenia, and Tourette disorder, has accelerated, resulting in a large number of molecular clues, it has proven difficult to generate specific hypotheses without the corresponding datasets at the protein complex and functional pathway level. Here, we describe one path forward-an initiative aimed at mapping the physical and genetic interaction networks of these conditions and then using these maps to connect the genomic data to neurobiology and, ultimately, the clinic. These efforts will include a team of geneticists, structural biologists, neurobiologists, systems biologists, and clinicians, leveraging a wide array of experimental approaches and creating a collaborative infrastructure necessary for long-term investigation. This initiative will ultimately intersect with parallel studies that focus on other diseases, as there is a significant overlap with genes implicated in cancer, infectious disease, and congenital heart defects.


Subject(s)
Chromosome Mapping/methods , Neurodevelopmental Disorders/genetics , Systems Biology/methods , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Genomics/methods , Humans , Neurobiology/methods , Neuropsychiatry
5.
Cell ; 169(2): 350-360.e12, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388416

ABSTRACT

Cells operate through protein interaction networks organized in space and time. Here, we describe an approach to resolve both dimensions simultaneously by using proximity labeling mediated by engineered ascorbic acid peroxidase (APEX). APEX has been used to capture entire organelle proteomes with high temporal resolution, but its breadth of labeling is generally thought to preclude the higher spatial resolution necessary to interrogate specific protein networks. We provide a solution to this problem by combining quantitative proteomics with a system of spatial references. As proof of principle, we apply this approach to interrogate proteins engaged by G-protein-coupled receptors as they dynamically signal and traffic in response to ligand-induced activation. The method resolves known binding partners, as well as previously unidentified network components. Validating its utility as a discovery pipeline, we establish that two of these proteins promote ubiquitin-linked receptor downregulation after prolonged activation.


Subject(s)
Ascorbate Peroxidases/chemistry , Protein Interaction Maps , Staining and Labeling/methods , Animals , Humans , Lysosomes/metabolism , Protein Transport , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid/metabolism , Ubiquitin/metabolism
6.
Nat Chem Biol ; 20(3): 323-332, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37749347

ABSTRACT

Many G protein-coupled receptors (GPCRs) initiate a second phase of stimulatory heterotrimeric G protein (Gs)-coupled cAMP signaling after endocytosis. The prevailing current view is that the endosomal signal is inherently Ɵ-arrestin-dependent because Ɵ-arrestin is necessary for receptor internalization and, for some GPCRs, to prolong the endosomal signal. Here we revise this view by showing that the vasoactive intestinal peptide receptor 1 (VIPR1), a secretin-family polypeptide hormone receptor, does not require Ɵ-arrestin to internalize or to generate an endosomal signal. Ɵ-Arrestin instead resolves the plasma membrane and endosomal signaling phases into sequential cAMP peaks by desensitizing the plasma membrane phase without affecting the endosomal phase. This appears to occur through the formation of functionally distinct VIPR1-Ɵ-arrestin complexes at each location that differ in their phosphorylation dependence. We conclude that endosomal GPCR signaling can occur in the absence of Ɵ-arrestin and that Ɵ-arrestin sculpts the spatiotemporal profile of cellular GPCR-G protein signaling through location-specific remodeling of GPCR-Ɵ-arrestin complexes.


Subject(s)
Peptide Hormones , Signal Transduction , beta-Arrestins , beta-Arrestin 1 , Cell Membrane
7.
Nat Chem Biol ; 20(9): 1133-1143, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38528119

ABSTRACT

The Āµ-opioid receptor (ĀµOR) represents an important target of therapeutic and abused drugs. So far, most understanding of ĀµOR activity has focused on a subset of known signal transducers and regulatory molecules. Yet ĀµOR signaling is coordinated by additional proteins in the interaction network of the activated receptor, which have largely remained invisible given the lack of technologies to interrogate these networks systematically. Here we describe a proteomics and computational approach to map the proximal proteome of the activated ĀµOR and to extract subcellular location, trafficking and functional partners of G-protein-coupled receptor (GPCR) activity. We demonstrate that distinct opioid agonists exert differences in the ĀµOR proximal proteome mediated by endocytosis and endosomal sorting. Moreover, we identify two new ĀµOR network components, EYA4 and KCTD12, which are recruited on the basis of receptor-triggered G-protein activation and might form a previously unrecognized buffering system for G-protein activity broadly modulating cellular GPCR signaling.


Subject(s)
Proteome , Proteomics , Receptors, Opioid, mu , Humans , Endocytosis , HEK293 Cells , Proteome/metabolism , Proteomics/methods , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/agonists , Signal Transduction
8.
Mol Syst Biol ; 20(8): 952-971, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951684

ABSTRACT

Proximity labeling (PL) via biotinylation coupled with mass spectrometry (MS) captures spatial proteomes in cells. Large-scale processing requires a workflow minimizing hands-on time and enhancing quantitative reproducibility. We introduced a scalable PL pipeline integrating automated enrichment of biotinylated proteins in a 96-well plate format. Combining this with optimized quantitative MS based on data-independent acquisition (DIA), we increased sample throughput and improved protein identification and quantification reproducibility. We applied this pipeline to delineate subcellular proteomes across various compartments. Using the 5HT2A serotonin receptor as a model, we studied temporal changes of proximal interaction networks induced by receptor activation. In addition, we modified the pipeline for reduced sample input to accommodate CRISPR-based gene knockout, assessing dynamics of the 5HT2A network in response to perturbation of selected interactors. This PL approach is universally applicable to PL proteomics using biotinylation-based PL enzymes, enhancing throughput and reproducibility of standard protocols.


Subject(s)
Biotinylation , Proteome , Proteomics , Proteomics/methods , Reproducibility of Results , Humans , Proteome/metabolism , Mass Spectrometry/methods , HEK293 Cells
9.
Cell ; 143(5): 761-73, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21111236

ABSTRACT

The functional consequences of signaling receptor endocytosis are determined by the endosomal sorting of receptors between degradation and recycling pathways. How receptors recycle efficiently, in a sequence-dependent manner that is distinct from bulk membrane recycling, is not known. Here, in live cells, we visualize the sorting of a prototypical sequence-dependent recycling receptor, the beta-2 adrenergic receptor, from bulk recycling proteins and the degrading delta-opioid receptor. Our results reveal a remarkable diversity in recycling routes at the level of individual endosomes, and indicate that sequence-dependent recycling is an active process mediated by distinct endosomal subdomains distinct from those mediating bulk recycling. We identify a specialized subset of tubular microdomains on endosomes, stabilized by a highly localized but dynamic actin machinery, that mediate this sorting, and provide evidence that these actin-stabilized domains provide the physical basis for a two-step kinetic and affinity-based model for protein sorting into the sequence-dependent recycling pathway.


Subject(s)
Actins/metabolism , Endosomes/metabolism , Protein Transport , Cell Line , Cell Membrane/metabolism , Cytoskeleton/metabolism , Humans , Kinetics , Protein Structure, Tertiary , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Receptors, Opioid, delta/metabolism
10.
J Biol Chem ; 299(11): 105293, 2023 11.
Article in English | MEDLINE | ID: mdl-37774973

ABSTRACT

Ɵ-arrestins play a key role in G protein-coupled receptor (GPCR) internalization, trafficking, and signaling. Whether Ɵ-arrestins act independently of G protein-mediated signaling has not been fully elucidated. Studies using genome-editing approaches revealed that whereas G proteins are essential for mitogen-activated protein kinase activation by GPCRs., Ɵ-arrestins play a more prominent role in signal compartmentalization. However, in the absence of G proteins, GPCRs may not activate Ɵ-arrestins, thereby limiting the ability to distinguish G protein from Ɵ-arrestin-mediated signaling events. We used Ɵ2-adrenergic receptor (Ɵ2AR) and its Ɵ2AR-C tail mutant expressed in human embryonic kidney 293Ā cells wildtype or CRISPR-Cas9 gene edited for Gαs, Ɵ-arrestin1/2, or GPCR kinases 2/3/5/6 in combination with arrestin conformational sensors to elucidate the interplay between Gαs and Ɵ-arrestins in controlling gene expression. We found that Gαs is not required for Ɵ2AR and Ɵ-arrestin conformational changes, Ɵ-arrestin recruitment, and receptor internalization, but that Gαs dictates the GPCR kinase isoforms involved in Ɵ-arrestin recruitment. By RNA-Seq analysis, we found that protein kinase A and mitogen-activated protein kinase gene signatures were activated by stimulation of Ɵ2AR in wildtype and Ɵ-arrestin1/2-KO cells but absent in Gαs-KO cells. These results were validated by re-expressing Gαs in the corresponding KO cells and silencing Ɵ-arrestins in wildtype cells. These findings were extended to cellular systems expressing endogenous levels of Ɵ2AR. Overall, our results support that Gs is essential for Ɵ2AR-promoted protein kinase A and mitogen-activated protein kinase gene expression signatures, whereas Ɵ-arrestins initiate signaling events modulating Gαs-driven nuclear transcriptional activity.


Subject(s)
GTP-Binding Proteins , Gene Expression Regulation , Receptors, Adrenergic, beta-2 , beta-Arrestins , Humans , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , beta-Arrestins/genetics , beta-Arrestins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/genetics , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , HEK293 Cells , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Protein Structure, Tertiary , Protein Isoforms , Enzyme Activation/genetics
11.
Nature ; 557(7705): 381-386, 2018 05.
Article in English | MEDLINE | ID: mdl-29720660

ABSTRACT

Ɵ-arrestins are critical regulator and transducer proteins for G-protein-coupled receptors (GPCRs). Ɵ-arrestin is widely believed to be activated by forming a stable and stoichiometric GPCR-Ɵ-arrestin scaffold complex, which requires and is driven by the phosphorylated tail of the GPCR. Here we demonstrate a distinct and additional mechanism of Ɵ-arrestin activation that does not require stable GPCR-Ɵ-arrestin scaffolding or the GPCR tail. Instead, it occurs through transient engagement of the GPCR core, which destabilizes a conserved inter-domain charge network in Ɵ-arrestin. This promotes capture of Ɵ-arrestin at the plasma membrane and its accumulation in clathrin-coated endocytic structures (CCSs) after dissociation from the GPCR, requiring a series of interactions with membrane phosphoinositides and CCS-lattice proteins. Ɵ-arrestin clustering in CCSs in the absence of the upstream activating GPCR is associated with a Ɵ-arrestin-dependent component of the cellular ERK (extracellular signal-regulated kinase) response. These results delineate a discrete mechanism of cellular Ɵ-arrestin function that is activated catalytically by GPCRs.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism , Animals , Biocatalysis , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , HEK293 Cells , Humans , Phosphatidylinositols/metabolism , Protein Transport , Receptors, G-Protein-Coupled/chemistry , beta-Arrestins/chemistry
12.
Nat Methods ; 17(11): 1147-1155, 2020 11.
Article in English | MEDLINE | ID: mdl-32895537

ABSTRACT

Genetically encoded dopamine sensors based on green fluorescent protein (GFP) enable high-resolution imaging of dopamine dynamics in behaving animals. However, these GFP-based variants cannot be readily combined with commonly used optical sensors and actuators, due to spectral overlap. We therefore engineered red-shifted variants of dopamine sensors called RdLight1, based on mApple. RdLight1 can be combined with GFP-based sensors with minimal interference and shows high photostability, permitting prolonged continuous imaging. We demonstrate the utility of RdLight1 for receptor-specific pharmacological analysis in cell culture, simultaneous assessment of dopamine release and cell-type-specific neuronal activity and simultaneous subsecond monitoring of multiple neurotransmitters in freely behaving rats. Dual-color photometry revealed that dopamine release in the nucleus accumbens evoked by reward-predictive cues is accompanied by a rapid suppression of glutamate release. By enabling multiplexed imaging of dopamine with other circuit components in vivo, RdLight1 opens avenues for understanding many aspects of dopamine biology.


Subject(s)
Behavior, Animal/physiology , Biosensing Techniques/methods , Brain/metabolism , Dopamine/metabolism , Neurons/metabolism , Animals , Cues , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Reward
13.
Nat Chem Biol ; 17(5): 558-566, 2021 05.
Article in English | MEDLINE | ID: mdl-33649598

ABSTRACT

G-protein-coupled receptor-regulated cAMP production from endosomes can specify signaling to the nucleus by moving the source of cAMP without changing its overall amount. How this is possible remains unknown because cAMP gradients dissipate over the nanoscale, whereas endosomes typically localize micrometers from the nucleus. We show that the key location-dependent step for endosome-encoded transcriptional control is nuclear entry of cAMP-dependent protein kinase (PKA) catalytic subunits. These are sourced from punctate accumulations of PKA holoenzyme that are densely distributed in the cytoplasm and titrated by global cAMP into a discrete metastable state, in which catalytic subunits are bound but dynamically exchange. Mobile endosomes containing activated receptors collide with the metastable PKA puncta and pause in close contact. We propose that these properties enable cytoplasmic PKA to act collectively like a semiconductor, converting nanoscale cAMP gradients generated from endosomes into microscale elevations of free catalytic subunits to direct downstream signaling.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Cytoplasm/metabolism , Endosomes/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/genetics , Animals , Catalytic Domain , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Clathrin Heavy Chains/antagonists & inhibitors , Clathrin Heavy Chains/genetics , Clathrin Heavy Chains/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cytoplasm/ultrastructure , Dynamin I/genetics , Dynamin I/metabolism , Endosomes/ultrastructure , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Receptors, Adrenergic, beta-2/genetics
14.
PLoS Genet ; 16(10): e1009103, 2020 10.
Article in English | MEDLINE | ID: mdl-33052901

ABSTRACT

G protein-coupled receptors (GPCRs) allow cells to respond to chemical and sensory stimuli through generation of second messengers, such as cyclic AMP (cAMP), which in turn mediate a myriad of processes, including cell survival, proliferation, and differentiation. In order to gain deeper insights into the complex biology and physiology of these key cellular pathways, it is critical to be able to globally map the molecular factors that shape cascade function. Yet, to this date, efforts to systematically identify regulators of GPCR/cAMP signaling have been lacking. Here, we combined genome-wide screening based on CRISPR interference with a novel sortable transcriptional reporter that provides robust readout for cAMP signaling, and carried out a functional screen for regulators of the pathway. Due to the sortable nature of the platform, we were able to assay regulators with strong and moderate phenotypes by analyzing sgRNA distribution among three fractions with distinct reporter expression. We identified 45 regulators with strong and 50 regulators with moderate phenotypes not previously known to be involved in cAMP signaling. In follow-up experiments, we validated the functional effects of seven newly discovered mediators (NUP93, PRIM1, RUVBL1, PKMYT1, TP53, SF3A2, and HRAS), and showed that they control distinct steps of the pathway. Thus, our study provides proof of principle that the screening platform can be applied successfully to identify bona fide regulators of GPCR/second messenger cascades in an unbiased and high-throughput manner, and illuminates the remarkable functional diversity among GPCR regulators.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Proliferation/genetics , Cyclic AMP/genetics , Receptors, G-Protein-Coupled/genetics , ATPases Associated with Diverse Cellular Activities/genetics , Carrier Proteins/genetics , Cell Differentiation/genetics , Cells, Cultured , DNA Helicases/genetics , DNA Primase/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA Splicing Factors/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics
15.
J Biol Chem ; 297(1): 100907, 2021 07.
Article in English | MEDLINE | ID: mdl-34166681

ABSTRACT

Endosomal signaling downstream of G-protein-coupled receptors (GPCRs) has emerged as a novel paradigm with important pharmacological and physiological implications. However, our knowledge of the functional consequences of intracellular signaling is incomplete. To begin to address this gap, we combined an optogenetic approach for site-specific generation of the prototypical second messenger generated by active GPCRs, cyclic AMP (cAMP), with unbiased mass-spectrometry-based analysis of the phosphoproteome. We identified 218 unique, high-confidence sites whose phosphorylation is either increased or decreased in response to cAMP elevation. We next determined that the same amount of cAMP produced from the endosomal membrane led to more robust changes in phosphorylation than the plasma membrane. Remarkably, this was true for the entire repertoire of 218 identified targets and irrespective of their annotated subcellular localizations (endosome, cell surface, nucleus, cytosol). Furthermore, we identified a particularly strong endosome bias for a subset of proteins that are dephosphorylated in response to cAMP. Through bioinformatics analysis, we established these targets as putative substrates for protein phosphatase 2A (PP2A), and we propose compartmentalized activation of PP2A by cAMP-responsive kinases as the likely underlying mechanism. Altogether, our study extends the concept that endosomal signaling is a significant functional contributor to cellular responsiveness to cAMP by establishing a unique role for localized cAMP production in defining categorically distinct phosphoresponses.


Subject(s)
Cyclic AMP/metabolism , Endosomes/metabolism , Phosphoproteins/metabolism , Proteome/metabolism , Animals , HEK293 Cells , Humans , Phosphoproteins/chemistry , Phosphorylation , Protein Domains , Protein Phosphatase 2/metabolism , Proteome/chemistry
16.
Traffic ; 20(2): 130-136, 2019 02.
Article in English | MEDLINE | ID: mdl-30578610

ABSTRACT

G protein-coupled receptors (GPCRs) physically connect extracellular information with intracellular signal propagation. Membrane trafficking plays a supportive role by "bookending" signaling events: movement through the secretory pathway delivers GPCRs to the cell surface where receptors can sample the extracellular environment, while endocytosis and endolysosomal membrane trafficking provide a versatile system to titrate cellular signaling potential and maintain homeostatic control. Recent evidence suggests that, in addition to these important effects, GPCR trafficking actively shapes the cellular signaling response by altering the location and timing of specific receptor-mediated signaling reactions. Here, we review key experimental evidence underlying this expanding view, focused on GPCR signaling mediated through activation of heterotrimeric G proteins located in the cytoplasm. We then discuss lingering and emerging questions regarding the interface between GPCR signaling and trafficking.


Subject(s)
Endosomes/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Humans , Protein Multimerization , Protein Transport , Receptors, G-Protein-Coupled/chemistry
17.
Mol Pharmacol ; 99(5): 392-398, 2021 05.
Article in English | MEDLINE | ID: mdl-33361190

ABSTRACT

Advances in proteomic methodologies based on quantitative mass spectrometry are now transforming pharmacology and experimental biology more broadly. The present review will discuss several examples based on work in the author's laboratory, which focuses on delineating relationships between G protein-coupled receptor signaling and trafficking in the endocytic network. The examples highlighted correspond to those discussed in a talk presented at the 2019 EB/ASPET meeting, which was organized by Professor Joe Beavo to commemorate his receipt of the Julius Axelrod Award. SIGNIFICANCE STATEMENT: GPCRs are allosteric machines that signal by interacting with other cellular proteins, and this, in turn, is determined by a complex interplay between the biochemical, subcellular localization, and membrane trafficking properties of receptors relative to transducer and regulatory proteins. The present minireview highlights recent advances and challenges in elucidating this dynamic cell biology and toward delineating the cellular basis of drug action at the level of defined GPCR interaction networks using proteomic approaches enabled by quantitative mass spectrometry.


Subject(s)
Protein Transport/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Humans , Proteomics/methods
18.
Nat Rev Mol Cell Biol ; 10(9): 609-22, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19696798

ABSTRACT

Cell signalling and endocytic membrane trafficking have traditionally been viewed as distinct processes. Although our present understanding is incomplete and there are still great controversies, it is now recognized that these processes are intimately and bidirectionally linked in animal cells. Indeed, many recent examples illustrate how endocytosis regulates receptor signalling (including signalling from receptor tyrosine kinases and G protein-coupled receptors) and, conversely, how signalling regulates the endocytic pathway. The mechanistic and functional principles that underlie the relationship between signalling and endocytosis in cell biology are becoming increasingly evident across many systems.


Subject(s)
Cell Membrane/physiology , Endocytosis/physiology , Signal Transduction , Animals , Humans
20.
Nature ; 2020 May 19.
Article in English | MEDLINE | ID: mdl-32433633
SELECTION OF CITATIONS
SEARCH DETAIL