Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 24.
Article in English | MEDLINE | ID: mdl-38077016

ABSTRACT

Mutational patterns caused by APOBEC3 cytidine deaminase activity are evident throughout human cancer genomes. In particular, the APOBEC3A family member is a potent genotoxin that causes substantial DNA damage in experimental systems and human tumors. However, the mechanisms that ensure genome stability in cells with active APOBEC3A are unknown. Through an unbiased genome-wide screen, we define the Structural Maintenance of Chromosomes 5/6 (SMC5/6) complex as essential for cell viability when APOBEC3A is active. We observe an absence of APOBEC3A mutagenesis in human tumors with SMC5/6 dysfunction, consistent with synthetic lethality. Cancer cells depleted of SMC5/6 incur substantial genome damage from APOBEC3A activity during DNA replication. Further, APOBEC3A activity results in replication tract lengthening which is dependent on PrimPol, consistent with re-initiation of DNA synthesis downstream of APOBEC3A-induced lesions. Loss of SMC5/6 abrogates elongated replication tracts and increases DNA breaks upon APOBEC3A activity. Our findings indicate that replication fork lengthening reflects a DNA damage response to APOBEC3A activity that promotes genome stability in an SMC5/6-dependent manner. Therefore, SMC5/6 presents a potential therapeutic vulnerability in tumors with active APOBEC3A.

2.
J Biol Chem ; 299(11): 105269, 2023 11.
Article in English | MEDLINE | ID: mdl-37739036

ABSTRACT

Prenylation is an irreversible post-translational modification that supports membrane interactions of proteins involved in various cellular processes, including migration, proliferation, and survival. Dysregulation of prenylation contributes to multiple disorders, including cancers and vascular and neurodegenerative diseases. Prenyltransferases tether isoprenoid lipids to proteins via a thioether linkage during prenylation. Pharmacological inhibition of the lipid synthesis pathway by statins is a therapeutic approach to control hyperlipidemia. Building on our previous finding that statins inhibit membrane association of G protein γ (Gγ) in a subtype-dependent manner, we investigated the molecular reasoning for this differential inhibition. We examined the prenylation of carboxy-terminus (Ct) mutated Gγ in cells exposed to Fluvastatin and prenyl transferase inhibitors and monitored the subcellular localization of fluorescently tagged Gγ subunits and their mutants using live-cell confocal imaging. Reversible optogenetic unmasking-masking of Ct residues was used to probe their contribution to prenylation and membrane interactions of the prenylated proteins. Our findings suggest that specific Ct residues regulate membrane interactions of the Gγ polypeptide, statin sensitivity, and extent of prenylation. Our results also show a few hydrophobic and charged residues at the Ct are crucial determinants of a protein's prenylation ability, especially under suboptimal conditions. Given the cell and tissue-specific expression of different Gγ subtypes, our findings indicate a plausible mechanism allowing for statins to differentially perturb heterotrimeric G protein signaling in cells depending on their Gγ-subtype composition. Our results may also provide molecular reasoning for repurposing statins as Ras oncogene inhibitors and the failure of using prenyltransferase inhibitors in cancer treatment.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Protein Prenylation , Humans , Amino Acid Motifs , Drug Resistance/genetics , HeLa Cells , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Models, Molecular , Mutation , Protein Prenylation/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Signal Transduction/drug effects
3.
bioRxiv ; 2023 Jul 04.
Article in English | MEDLINE | ID: mdl-37461501

ABSTRACT

Prenylation is a universal and irreversible post-translational modification that supports membrane interactions of proteins involved in various cellular processes, including migration, proliferation, and survival. Thus, dysregulation of prenylation contributes to multiple disorders, including cancers, vascular diseases, and neurodegenerative diseases. During prenylation, prenyltransferase enzymes tether metabolically produced isoprenoid lipids to proteins via a thioether linkage. Pharmacological inhibition of the lipid synthesis pathway by statins has long been a therapeutic approach to control hyperlipidemia. Building on our previous finding that statins inhibit membrane association of G protein γ (Gγ) in a subtype-dependent manner, we investigated the molecular reasoning for this differential. We examined the prenylation efficacy of carboxy terminus (Ct) mutated Gγ in cells exposed to Fluvastatin and prenyl transferase inhibitors and monitored the subcellular localization of fluorescently tagged Gγ subunits and their mutants using live-cell confocal imaging. Reversible optogenetic unmasking-masking of Ct residues was used to probe their contribution to the prenylation process and membrane interactions of the prenylated proteins. Our findings suggest that specific Ct residues regulate membrane interactions of the Gγ polypeptide statin sensitivity, and prenylation efficacy. Our results also show that a few hydrophobic and charged residues at the Ct are crucial determinants of a protein's prenylation ability, especially under suboptimal conditions. Given the cell and tissue-specific expression of different Gγ subtypes, our findings explain how and why statins differentially perturb heterotrimeric G protein signaling in specific cells and tissues. Our results may provide molecular reasoning for repurposing statins as Ras oncogene inhibitors and the failure of using prenyltransferase inhibitors in cancer treatment.

4.
Sci Rep ; 13(1): 2771, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36797332

ABSTRACT

Phosphatidylinositol (3,4,5) trisphosphate (PIP3) is a plasma membrane-bound signaling phospholipid involved in many cellular signaling pathways that control crucial cellular processes and behaviors, including cytoskeleton remodeling, metabolism, chemotaxis, and apoptosis. Therefore, defective PIP3 signaling is implicated in various diseases, including cancer, diabetes, obesity, and cardiovascular diseases. Upon activation by G protein-coupled receptors (GPCRs) or receptor tyrosine kinases (RTKs), phosphoinositide-3-kinases (PI3Ks) phosphorylate phosphatidylinositol (4,5) bisphosphate (PIP2), generating PIP3. Though the mechanisms are unclear, PIP3 produced upon GPCR activation attenuates within minutes, indicating a tight temporal regulation. Our data show that subcellular redistributions of G proteins govern this PIP3 attenuation when GPCRs are activated globally, while localized GPCR activation induces sustained subcellular PIP3. Interestingly the observed PIP3 attenuation was Gγ subtype-dependent. Considering distinct cell-tissue-specific Gγ expression profiles, our findings not only demonstrate how the GPCR-induced PIP3 response is regulated depending on the GPCR activity gradient across a cell, but also show how diversely cells respond to spatial and temporal variability of external stimuli.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Signal Transduction/physiology , Receptors, G-Protein-Coupled/metabolism , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , Phosphatidylinositols/metabolism
5.
J Biol Chem ; 298(12): 102618, 2022 12.
Article in English | MEDLINE | ID: mdl-36272647

ABSTRACT

Heterotrimeric G proteins (αßγ subunits) that are activated by G protein-coupled receptors (GPCRs) mediate the biological responses of eukaryotic cells to extracellular signals. The α subunits and the tightly bound ßγ subunit complex of G proteins have been extensively studied and shown to control the activity of effector molecules. In contrast, the potential roles of the large family of γ subunits have been less studied. In this review, we focus on present knowledge about these proteins. Induced loss of individual γ subunit types in animal and plant models result in strikingly distinct phenotypes indicating that γ subtypes play important and specific roles. Consistent with these findings, downregulation or upregulation of particular γ subunit types result in various types of cancers. Clues about the mechanistic basis of γ subunit function have emerged from imaging the dynamic behavior of G protein subunits in living cells. This shows that in the basal state, G proteins are not constrained to the plasma membrane but shuttle between membranes and on receptor activation ßγ complexes translocate reversibly to internal membranes. The translocation kinetics of ßγ complexes varies widely and is determined by the membrane affinity of the associated γ subtype. On translocating, some ßγ complexes act on effectors in internal membranes. The variation in translocation kinetics determines differential sensitivity and adaptation of cells to external signals. Membrane affinity of γ subunits is thus a parsimonious and elegant mechanism that controls information flow to internal cell membranes while modulating signaling responses.


Subject(s)
GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Receptors, G-Protein-Coupled , Animals , Cell Membrane/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , Signal Transduction , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Humans
6.
Mol Biol Cell ; 32(16): 1446-1458, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34106735

ABSTRACT

G protein beta-gamma (Gßγ) subunits anchor to the plasma membrane (PM) through the carboxy-terminal (CT) prenyl group in Gγ. This interaction is crucial for the PM localization and functioning of Gßγ, allowing GPCR-G protein signaling to proceed. The diverse Gγ family has 12 members, and we have recently shown that the signaling efficacies of major Gßγ effectors are Gγ-type dependent. This dependency is due to the distinct series of membrane-interacting abilities of Gγ. However, the molecular process allowing for Gßγ subunits to exhibit a discrete and diverse range of Gγ-type-dependent membrane affinities is unclear and cannot be explained using only the type of prenylation. The present work explores the unique designs of membrane-interacting CT residues in Gγ as a major source for this Gγ-type-dependent Gßγ signaling. Despite the type of prenylation, the results show signaling efficacy at the PM, and associated cell behaviors of Gßγ are governed by crucially located specific amino acids in the five to six residue preprenylation region of Gγ. The provided molecular picture of Gγ-membrane interactions may explain how cells gain Gγ-type-dependent G protein-GPCR signaling as well as how Gßγ elicits selective signaling at various subcellular compartments.


Subject(s)
Cell Membrane/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Protein Prenylation , Signal Transduction , GTP-Binding Protein beta Subunits/metabolism , HeLa Cells , Humans
7.
J Biol Chem ; 296: 100702, 2021.
Article in English | MEDLINE | ID: mdl-33901492

ABSTRACT

Phospholipase C ß (PLCß), which is activated by the Gq family of heterotrimeric G proteins, hydrolyzes the inner membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2), generating diacylglycerol and inositol 1,4,5-triphosphate (IP3). Because Gq and PLCß regulate many crucial cellular processes and have been identified as major disease drivers, activation and termination of PLCß signaling by the Gαq subunit have been extensively studied. Gq-coupled receptor activation induces intense and transient PIP2 hydrolysis, which subsequently recovers to a low-intensity steady-state equilibrium. However, the molecular underpinnings of this equilibrium remain unclear. Here, we explored the influence of signaling crosstalk between Gq and Gi/o pathways on PIP2 metabolism in living cells using single-cell and optogenetic approaches to spatially and temporally constrain signaling. Our data suggest that the Gßγ complex is a component of the highly efficient lipase GαqGTP-PLCß-Gßγ. We found that over time, Gßγ dissociates from this lipase complex, leaving the less-efficient GαqGTP-PLCß lipase complex and allowing the significant partial recovery of PIP2 levels. Our findings also indicate that the subtype of the Gγ subunit in Gßγ fine-tunes the lipase activity of Gq-PLCß, in which cells expressing Gγ with higher plasma membrane interaction show lower PIP2 recovery. Given that Gγ shows cell- and tissue-specific subtype expression, our findings suggest the existence of tissue-specific distinct Gq-PLCß signaling paradigms. Furthermore, these results also outline a molecular process that likely safeguards cells from excessive Gq signaling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/metabolism , Cell Membrane/metabolism , HeLa Cells , Humans , Hydrolysis , Models, Molecular , Phospholipase C beta/chemistry , Protein Binding , Protein Conformation , Signal Transduction
8.
Cell Signal ; 82: 109947, 2021 06.
Article in English | MEDLINE | ID: mdl-33582184

ABSTRACT

G protein-coupled receptors (GPCRs) transmit information to the cell interior by transducing external signals to heterotrimeric G protein subunits, Gα and Gßγ subunits, localized on the inner leaflet of the plasma membrane. Though the initial focus was mainly on Gα-mediated events, Gßγ subunits were later identified as major contributors to GPCR-G protein signalling. A broad functional array of Gßγ signalling has recently been attributed to Gß and Gγ subtype diversity, comprising 5 Gß and 12 Gγ subtypes, respectively. In addition to displaying selectivity towards each other to form the Gßγ dimer, numerous studies have identified preferences of distinct Gßγ combinations for specific GPCRs, Gα subtypes and effector molecules. Importantly, Gß and Gγ subtype-dependent regulation of downstream effectors, representing a diverse range of signalling pathways and physiological functions have been found. Here, we review the literature on the repercussions of Gß and Gγ subtype diversity on direct and indirect regulation of GPCR/G protein signalling events and their physiological outcomes. Our discussion additionally provides perspective in understanding the intricacies underlying molecular regulation of subtype-specific roles of Gßγ signalling and associated diseases.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Animals , Humans
9.
Anal Bioanal Chem ; 411(19): 4481-4508, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30927013

ABSTRACT

G protein-coupled receptors (GPCRs), G proteins, and their signaling associates are major signal transducers that control the majority of cellular signaling and regulate key biological functions including immune, neurological, cardiovascular, and metabolic processes. These pathways are targeted by over one-third of drugs on the market; however, the current understanding of their function is limited and primarily derived from cell-destructive approaches providing an ensemble of static, multi-cell information about the status and composition of molecules. Spatiotemporal behavior of molecules involved is crucial to understanding in vivo cell behaviors both in health and disease, and the advent of genetically encoded fluorescence proteins and small fluorophore-based biosensors has facilitated the mapping of dynamic signaling in cells with subcellular acuity. Since we and others have developed optogenetic methods to regulate GPCR-G protein signaling in single cells and subcellular regions using dedicated wavelengths, the desire to develop and adopt optogenetically amenable assays to measure signaling has motivated us to take a broader look at the available optical tools and approaches compatible with measuring single-cell and subcellular GPCR-G protein signaling. Here we review such key optical approaches enabling the examination of GPCR, G protein, secondary messenger, and downstream molecules such as kinase and lipid signaling in living cells. The methods reviewed employ both fluorescence and bioluminescence detection. We not only further elaborate the underlying principles of these sensors but also discuss the experimental criteria and limitations to be considered during their use in single-cell and subcellular signal mapping.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Single-Cell Analysis , Subcellular Fractions/metabolism , Fluorescence , Gene Expression/physiology , Humans , Protein Binding , Receptors, G-Protein-Coupled/physiology
10.
Cell Signal ; 58: 34-43, 2019 06.
Article in English | MEDLINE | ID: mdl-30849518

ABSTRACT

G protein αq-coupled receptors (Gq-GPCRs) primarily signal through GαqGTP mediated phospholipase Cß (PLCß) stimulation and the subsequent hydrolysis of phosphatidylinositol 4, 5 bisphosphate (PIP2). Though Gq-heterotrimer activation results in both GαqGTP and Gßγ, unlike Gi/o-receptors, it is unclear if Gq-coupled receptors employ Gßγ as a major signal transducer. Compared to Gi/o- and Gs-coupled receptors, we observed that most cell types exhibit a limited free Gßγ generation upon Gq-pathway and Gαq/11 heterotrimer activation. We show that cells transfected with Gαq or endogenously expressing more than average-levels of Gαq/11 compared to Gαs and Gαi exhibit a distinct signaling regime primarily characterized by recovery-resistant PIP2 hydrolysis. Interestingly, the elevated Gq-expression is also associated with enhanced free Gßγ generation and signaling. Furthermore, the gene GNAQ, which encodes for Gαq, has recently been identified as a cancer driver gene. We also show that GNAQ is overexpressed in tumor samples of patients with Kidney Chromophobe (KICH) and Kidney renal papillary (KIRP) cell carcinomas in a matched tumor-normal sample analysis, which demonstrates the clinical significance of Gαq expression. Overall, our data indicates that cells usually express low Gαq levels, likely safeguarding cells from excessive calcium as wells as from Gßγ signaling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Signal Transduction , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Expression , HeLa Cells , Humans , Hydrolysis , Phospholipase C beta/metabolism , Transfection
11.
Mol Pharmacol ; 95(4): 361-375, 2019 04.
Article in English | MEDLINE | ID: mdl-30765461

ABSTRACT

Guanine nucleotide-binding proteins (G proteins) facilitate the transduction of external signals to the cell interior, regulate most eukaryotic signaling, and thus have become crucial disease drivers. G proteins largely function at the inner leaflet of the plasma membrane (PM) using covalently attached lipid anchors. Both small monomeric and heterotrimeric G proteins are primarily prenylated, either with a 15-carbon farnesyl or a 20-carbon geranylgeranyl polyunsaturated lipid. The mevalonate [3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase] pathway synthesizes lipids for G-protein prenylation. It is also the source of the precursor lipids for many biomolecules, including cholesterol. Consequently, the rate-limiting enzymes of the mevalonate pathway are major targets for cholesterol-lowering medications and anticancer drug development. Although prenylated G protein γ (Gγ) is essential for G protein-coupled receptor (GPCR)-mediated signaling, how mevalonate pathway inhibitors, statins, influence subcellular distribution of Gßγ dimer and Gαßγ heterotrimer, as well as their signaling upon GPCR activation, is poorly understood. The present study shows that clinically used statins not only significantly disrupt PM localization of Gßγ but also perturb GPCR-G protein signaling and associated cell behaviors. The results also demonstrate that the efficiency of prenylation inhibition by statins is Gγ subtype-dependent and is more effective toward farnesylated Gγ types. Since Gγ is required for Gßγ signaling and shows a cell- and tissue-specific subtype distribution, the present study can help understand the mechanisms underlying clinical outcomes of statin use in patients. This work also reveals the potential of statins as clinically usable drugs to control selected GPCR-G protein signaling.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Proteins/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholesterol/metabolism , HeLa Cells , Humans , Mevalonic Acid/pharmacology , Mice , Protein Prenylation/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects
12.
Int Rev Cell Mol Biol ; 339: 133-191, 2018.
Article in English | MEDLINE | ID: mdl-29776603

ABSTRACT

Heterotrimeric guanine nucleotide-binding proteins (G proteins) deliver external signals to the cell interior, upon activation by the external signal stimulated G protein-coupled receptors (GPCRs).While the activated GPCRs control several pathways independently, activated G proteins control the vast majority of cellular and physiological functions, ranging from vision to cardiovascular homeostasis. Activated GPCRs dissociate GαGDPßγ heterotrimer into GαGTP and free Gßγ. Earlier, GαGTP was recognized as the primary signal transducer of the pathway and Gßγ as a passive signaling modality that facilitates the activity of Gα. However, Gßγ later found to regulate more number of pathways than GαGTP does. Once liberated from the heterotrimer, free Gßγ interacts and activates a diverse range of signaling regulators including kinases, lipases, GTPases, and ion channels, and it does not require any posttranslation modifications. Gßγ family consists of 48 members, which show cell- and tissue-specific expressions, and recent reports show that cells employ the subtype diversity in Gßγ to achieve desired signaling outcomes. In addition to activated GPCRs, which induce free Gßγ generation and the rate of GTP hydrolysis in Gα, which sequester Gßγ in the heterotrimer, terminating Gßγ signaling, additional regulatory mechanisms exist to regulate Gßγ activity. In this chapter, we discuss structure and function, subtype diversity and its significance in signaling regulation, effector activation, regulatory mechanisms as well as the disease relevance of Gßγ in eukaryotes.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Binding Sites , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , Humans , Models, Biological
13.
J Biol Chem ; 293(8): 2974-2989, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29317505

ABSTRACT

G protein ßγ subunit (Gßγ) is a major signal transducer and controls processes ranging from cell migration to gene transcription. Despite having significant subtype heterogeneity and exhibiting diverse cell- and tissue-specific expression levels, Gßγ is often considered a unified signaling entity with a defined functionality. However, the molecular and mechanistic basis of Gßγ's signaling specificity is unknown. Here, we demonstrate that Gγ subunits, bearing the sole plasma membrane (PM)-anchoring motif, control the PM affinity of Gßγ and thereby differentially modulate Gßγ effector signaling in a Gγ-specific manner. Both Gßγ signaling activity and the migration rate of macrophages are strongly dependent on the PM affinity of Gγ. We also found that the type of C-terminal prenylation and five to six pre-CaaX motif residues at the PM-interacting region of Gγ control the PM affinity of Gßγ. We further show that the overall PM affinity of the Gßγ pool of a cell type is a strong predictor of its Gßγ signaling-activation efficacy. A kinetic model encompassing multiple Gγ types and parameterized for empirical Gßγ behaviors not only recapitulated experimentally observed signaling of Gßγ, but also suggested a Gγ-dependent, active-inactive conformational switch for the PM-bound Gßγ, regulating effector signaling. Overall, our results unveil crucial aspects of signaling and cell migration regulation by Gγ type-specific PM affinities of Gßγ.


Subject(s)
Calcium Signaling , Cell Membrane/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Macrophages/metabolism , Models, Biological , Animals , Cell Membrane/chemistry , Cell Movement , Computational Biology , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/genetics , Half-Life , HeLa Cells , Humans , Kinetics , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macrophage Activation , Macrophages/cytology , Macrophages/immunology , Mice , Protein Interaction Domains and Motifs , Protein Prenylation , Protein Transport , RAW 264.7 Cells , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
14.
Methods Cell Biol ; 142: 1-25, 2017.
Article in English | MEDLINE | ID: mdl-28964328

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors in eukaryotic genomes. They control a variety of cellular and physiological processes such as hormone secretion and heart rate, and therefore are associated with a majority of pathological conditions including cancer and heart diseases. Currently established assays to measure ligand-induced activation of GPCRs and G proteins possess limitations such as being time consuming, indirect, and expensive. Thus, an efficient method to measure GPCR-G protein activation is required to identify novel pharmacological modulators to control them and gain insights about molecular underpinnings of the associated pathways. Activation of GPCRs induces dissociation of G protein heterotrimers to form GαGTP and free Gßγ. Free Gßγ subunits have been shown to translocate reversibly from the plasma membrane to internal membranes. Gßγ translocation therefore represents the GPCR-G protein activation, and thus, imaging of this process can be used to quantify the kinetics and magnitude of the pathway activation-deactivation in real time in living cells. The objective of this chapter is to elaborate the protocols of (i) generation and optimization of the required sensor constructs; (ii) development of cell culture, transient transfection, imaging, and optogenetic procedures; (iii) imaging and data analysis methods; and (iv) stable cell line generation, pertaining to this assay to measure GPCR-G protein activation.


Subject(s)
Biological Assay/methods , Heterotrimeric GTP-Binding Proteins/metabolism , Optical Imaging/methods , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Biological Assay/instrumentation , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Cell Membrane/metabolism , HeLa Cells , Humans , Kinetics , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Molecular Imaging/instrumentation , Molecular Imaging/methods , Optical Imaging/instrumentation , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...