Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.200
Filter
1.
Nat Commun ; 15(1): 7684, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227390

ABSTRACT

A long-held tenet in inositol-lipid signaling is that cleavage of membrane phosphoinositides by phospholipase Cß (PLCß) isozymes to increase cytosolic Ca2+ in living cells is exclusive to Gq- and Gi-sensitive G protein-coupled receptors (GPCRs). Here we extend this central tenet and show that Gs-GPCRs also partake in inositol-lipid signaling and thereby increase cytosolic Ca2+. By combining CRISPR/Cas9 genome editing to delete Gαs, the adenylyl cyclase isoforms 3 and 6, or the PLCß1-4 isozymes, with pharmacological and genetic inhibition of Gq and G11, we pin down Gs-derived Gßγ as driver of a PLCß2/3-mediated cytosolic Ca2+ release module. This module does not require but crosstalks with Gαs-dependent cAMP, demands Gαq to release PLCß3 autoinhibition, but becomes Gq-independent with mutational disruption of the PLCß3 autoinhibited state. Our findings uncover the key steps of a previously unappreciated mechanism utilized by mammalian cells to finetune their calcium signaling regulation through Gs-GPCRs.


Subject(s)
Calcium Signaling , Calcium , Phospholipase C beta , Receptors, G-Protein-Coupled , Humans , Phospholipase C beta/metabolism , Phospholipase C beta/genetics , HEK293 Cells , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , CRISPR-Cas Systems , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Cyclic AMP/metabolism , Animals , Gene Editing , Cytosol/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/genetics , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/genetics
2.
Biochem J ; 481(18): 1187-1202, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39258799

ABSTRACT

Phosphatidylinositol is a precursor of various phosphoinositides, which play crucial roles in intracellular signaling and membrane dynamics and have impact on diverse aspects of cell physiology. Phosphoinositide synthesis and turnover occur in the cytoplasmic leaflet of the organellar and plasma membranes. P4-ATPases (lipid flippases) are responsible for translocating membrane lipids from the exoplasmic (luminal) to the cytoplasmic leaflet, thereby regulating membrane asymmetry. However, the mechanism underlying phosphatidylinositol translocation across cellular membranes remains elusive. Here, we discovered that the phosphatidylcholine flippases ATP8B1, ATP8B2, and ATP10A can also translocate phosphatidylinositol at the plasma membrane. To explore the function of these phosphatidylinositol flippases, we used cells depleted of CDC50A, a protein necessary for P4-ATPase function and ATP8B1 and ATP8B2, which express in HeLa cells. Upon activation of the Gq-coupled receptor, depletion of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] was accelerated in CDC50A knockout (KO) and ATP8B1/8B2 double KO cells compared with control cells, suggesting a decrease in PtdIns(4,5)P2 levels within the plasma membrane of the KO cells upon stimulation. These findings highlight the important role of P4-ATPases in maintaining phosphoinositide homeostasis and suggest a mechanism for asymmetry of phosphatidylinositol in the cytoplasmic leaflet of the plasma membrane.


Subject(s)
Adenosine Triphosphatases , Cell Membrane , Homeostasis , Phosphatidylinositols , Humans , Cell Membrane/metabolism , HeLa Cells , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Phosphatidylinositols/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics
3.
BMC Genomics ; 25(1): 800, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182029

ABSTRACT

BACKGROUND: The Gα family plays a crucial role in the complex reproductive regulatory network of teleosts. However, the characterization and function of Gα family members, especially Gαq, remain poorly understood in teleosts. To analyze the characterization, expression, and function of grass carp (Ctenopharyngodon idella) Gαq, we identified the Gα family members in grass carp genome, and analyzed the expression, distribution, and signal transduction of Gαq/gnaq. We also explored the role of Gαq in the reproductive regulation of grass carp. RESULTS: Our results showed that the grass carp genome contains 27 Gα genes with 46 isoforms, which are divided into four subfamilies: Gαs, Gαi/o, Gαq/11, and Gα12/13. The expression level of Cignaq in the testis was the highest and significantly higher than in other tissues, followed by the hypothalamus and brain. The luteinizing hormone receptor (LHR) was mainly localized to the nucleus in grass carp oocytes, with signals also present in follicular cells. In contrast, Gαq signal was mainly found in the cytoplasm of oocytes, with no signal in follicular cells. In the testis, Gαq and LHR were co-localized in the cytoplasm. Furthermore, the grass carp Gαq recombinant protein significantly promoted Cipgr expression. CONCLUSIONS: These results provided preliminary evidence for understanding the role of Gαq in the reproductive regulation of teleosts.


Subject(s)
Carps , Reproduction , Animals , Carps/genetics , Carps/metabolism , Reproduction/genetics , Fish Proteins/genetics , Fish Proteins/metabolism , Male , Female , Signal Transduction , Phylogeny , Genome , Testis/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Oocytes/metabolism
4.
Ecotoxicol Environ Saf ; 283: 116969, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39216220

ABSTRACT

Novel Psychoactive Substances (NPS) derived from tryptamines has been detected in aquatic environments, leading to environmental toxicology concerns. However, the specific toxicological mechanism, underlying these NPS, remains unclear. In our previous work, we used 5-Methoxy-N-isopropyl-N-methyltryptamine (5-MeO-MiPT) as the representative drug for NPS, and found that, 5-MeO-MiPT led to obvious behavioral inhibition and oxidative stress responses in zebrafishes model. In this study, Zebrafish were injected with varying concentrations of 5-MeO-MiPT for 30 days. RNA-seq, qPCR, metabolomics, and histopathological analyses were conducted to assess gene expression and tissue integrity. This study confirms that 5-MeO-MiPT substantially influences the transcription and expression of 13 selected genes, including ucp1, pet100, grik3, and grik4, mediated by the Gαq/11-PLCß signaling pathway. We elucidate the molecular mechanism that 5-MeO-MiPT can inhibit DAG-Ca2+/Pkc/Erk, Pkc/Pla2/PLCs and Ca2+/Camk Ⅱ/NMDA, while enhance Ca2+/Creb. Those secondary signaling pathways may be the mechanisms mediating 5-MeO-MiPT inhibiting normal behavior in zebrafish. These findings offer novel insights into the toxicological effects and addiction mechanisms of 5-MeO-MiPT. Moreover, it presents promising avenues for investigating other tryptamine-based NPS and offers a new direction for diagnosing and treating liver-brain pathway-related diseases.


Subject(s)
Signal Transduction , Tryptamines , Zebrafish , Animals , Signal Transduction/drug effects , Tryptamines/toxicity , Phospholipase C beta/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Water Pollutants, Chemical/toxicity , Oxidative Stress/drug effects , Psychotropic Drugs/toxicity , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 121(36): e2411846121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190348

ABSTRACT

Optogenetic techniques provide genetically targeted, spatially and temporally precise approaches to correlate cellular activities and physiological outcomes. In the nervous system, G protein-coupled receptors (GPCRs) have essential neuromodulatory functions through binding extracellular ligands to induce intracellular signaling cascades. In this work, we develop and validate an optogenetic tool that disrupts Gαq signaling through membrane recruitment of a minimal regulator of G protein signaling (RGS) domain. This approach, Photo-induced Gα Modulator-Inhibition of Gαq (PiGM-Iq), exhibited potent and selective inhibition of Gαq signaling. Using PiGM-Iq we alter the behavior of Caenorhabditis elegans and Drosophila with outcomes consistent with GPCR-Gαq disruption. PiGM-Iq changes axon guidance in cultured dorsal root ganglia neurons in response to serotonin. PiGM-Iq activation leads to developmental deficits in zebrafish embryos and larvae resulting in altered neuronal wiring and behavior. Furthermore, by altering the minimal RGS domain, we show that this approach is amenable to Gαi signaling. Our unique and robust optogenetic Gα inhibiting approaches complement existing neurobiological tools and can be used to investigate the functional effects neuromodulators that signal through GPCR and trimeric G proteins.


Subject(s)
Caenorhabditis elegans , GTP-Binding Protein alpha Subunits, Gq-G11 , Optogenetics , RGS Proteins , Signal Transduction , Zebrafish , Animals , Optogenetics/methods , Caenorhabditis elegans/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , RGS Proteins/metabolism , RGS Proteins/genetics , Zebrafish/embryology , Neurons/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Protein Domains , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Drosophila/metabolism
6.
Invest Ophthalmol Vis Sci ; 65(8): 37, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39042403

ABSTRACT

Purpose: Cell lines are being used in preclinical uveal melanoma (UM) research. Because not all cell lines harbor typical GNAQ or GNA11 hotspot mutations, we aimed at better classifying them and determining whether we could find genetic causes to explain the protein and mRNA expression profiles of the cell lines. Methods: We studied protein and mRNA expression of 14 UM cell lines and determined the presence of single nucleotide variants and small insertions and deletions with next-generation sequencing and copy number alterations with a single nucleotide polymorphism array. The lists of differentially expressed proteins and genes were merged, and shared lists were created, keeping only terms with concordant mRNA and protein expression. Enrichment analyses were performed on the shared lists. Results: Cell lines Mel285 and Mel290 are separate from GNA-mutated cell lines and show downregulation of melanosome-related markers. Both lack typical UM mutations but each harbors four putatively deleterious variants in CTNNB1, PPP1R10, LIMCH1, and APC in Mel285 and ARID1A, PPP1R10, SPG11, and RNF43 in Mel290. The upregulated terms in Mel285 and Mel290 did not point to a convincing alternative origin. Mel285 shows loss of chromosomes 1p, 3p, partial 3q, 6, and partial 8p, whereas Mel290 shows loss of 1p and 6. Expression in the other 12 cell lines was related to BAP1 expression. Conclusions: Although Mel285 and Mel290 have copy number alterations that fit UM, multi-omics analyses show that they belong to a separate group compared to the other analyzed UM cell lines. Therefore, they may not be representative models to test potential therapeutic targets for UM.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , GTP-Binding Protein alpha Subunits , Gene Expression Regulation, Neoplastic , Melanoma , Mutation , RNA, Messenger , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Uveal Neoplasms , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Humans , Ubiquitin Thiolesterase/genetics , RNA, Messenger/genetics , GTP-Binding Protein alpha Subunits/genetics , Tumor Suppressor Proteins/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Cell Line, Tumor , DNA Copy Number Variations , Polymorphism, Single Nucleotide , DNA Mutational Analysis
7.
EMBO Mol Med ; 16(8): 1930-1956, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38977926

ABSTRACT

Pulmonary arterial hypertension (PAH) is a life-threatening disease with limited survival. Herein, we propose the pharmacological inhibition of Gq proteins as a novel concept to counteract pulmonary vasoconstriction and proliferation/migration of pulmonary artery smooth muscle cells (PASMCs) in PAH. We demonstrate that the specific pan-Gq inhibitor FR900359 (FR) induced a strong vasorelaxation in large and small pulmonary arteries in mouse, pig, and human subjects ex vivo. Vasorelaxation by FR proved at least as potent as the currently used triple therapy. We also provide in vivo evidence that local pulmonary application of FR prevented right ventricular systolic pressure increase in healthy mice as well as in mice suffering from hypoxia (Hx)-induced pulmonary hypertension (PH). In addition, we demonstrate that chronic application of FR prevented and also reversed Sugen (Su)Hx-induced PH in mice. We also demonstrate that Gq inhibition reduces proliferation and migration of PASMCs in vitro. Thus, our work illustrates a dominant role of Gq proteins for pulmonary vasoconstriction as well as remodeling and proposes direct Gq inhibition as a powerful pharmacological strategy in PH.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , Hypertension, Pulmonary , Pulmonary Artery , Animals , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Humans , Mice , Pulmonary Artery/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Swine , Vasodilation/drug effects , Male , Cell Proliferation/drug effects , Cell Movement/drug effects , Mice, Inbred C57BL , Depsipeptides
8.
Cell Rep ; 43(8): 114511, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39024101

ABSTRACT

Bombesin receptor subtype-3 (BRS3) is an important orphan G protein-coupled receptor that regulates energy homeostasis and insulin secretion. As a member of the bombesin receptor (BnR) family, the lack of known endogenous ligands and high-resolution structure has hindered the understanding of BRS3 signaling and function. We present two cryogenic electron microscopy (cryo-EM) structures of BRS3 in complex with the heterotrimeric Gq protein in its active states: one bound to the pan-BnR agonist BA1 and the other bound to the synthetic BRS3-specific agonist MK-5046. These structures reveal the architecture of the orthosteric ligand pocket underpinning molecular recognition and provide insights into the structural basis for BRS3's selectivity and low affinity for bombesin peptides. Examination of conserved micro-switches suggests a shared activation mechanism among BnRs. Our findings shed light on BRS3's ligand selectivity and signaling mechanisms, paving the way for exploring its therapeutic potential for diabetes, obesity, and related metabolic disorders.


Subject(s)
Receptors, Bombesin , Receptors, Bombesin/metabolism , Ligands , Humans , Cryoelectron Microscopy , Protein Binding , Animals , HEK293 Cells , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry
9.
Am J Hum Genet ; 111(7): 1370-1382, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38917801

ABSTRACT

Extra-axial cavernous hemangiomas (ECHs) are complex vascular lesions mainly found in the spine and cavernous sinus. Their removal poses significant risk due to their vascularity and diffuse nature, and their genetic underpinnings remain incompletely understood. Our approach involved genetic analyses on 31 tissue samples of ECHs employing whole-exome sequencing and targeted deep sequencing. We explored downstream signaling pathways, gene expression changes, and resultant phenotypic shifts induced by these mutations, both in vitro and in vivo. In our cohort, 77.4% of samples had somatic missense variants in GNA14, GNAQ, or GJA4. Transcriptomic analysis highlighted significant pathway upregulation, with the GNAQ c.626A>G (p.Gln209Arg) mutation elevating PI3K-AKT-mTOR and angiogenesis-related pathways, while GNA14 c.614A>T (p.Gln205Leu) mutation led to MAPK and angiogenesis-related pathway upregulation. Using a mouse xenograft model, we observed enlarged vessels from these mutations. Additionally, we initiated rapamycin treatment in a 14-year-old individual harboring the GNAQ c.626A>G (p.Gln209Arg) variant, resulting in gradual regression of cutaneous cavernous hemangiomas and improved motor strength, with minimal side effects. Understanding these mutations and their pathways provides a foundation for developing therapies for ECHs resistant to current therapies. Indeed, the administration of rapamycin in an individual within this study highlights the promise of targeted treatments in treating these complex lesions.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , GTP-Binding Protein alpha Subunits , Humans , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Animals , Mice , Female , Male , GTP-Binding Protein alpha Subunits/genetics , Mutation , Adult , Middle Aged , Signal Transduction , Hemangioma, Cavernous/genetics , Hemangioma, Cavernous/pathology , Adolescent , Exome Sequencing , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics
10.
Cell Rep ; 43(7): 114389, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38935498

ABSTRACT

Kisspeptin signaling through its G protein-coupled receptor, KISS1R, plays an indispensable role in regulating reproduction via the hypothalamic-pituitary-gonadal axis. Dysregulation of this pathway underlies severe disorders like infertility and precocious puberty. Here, we present cryo-EM structures of KISS1R bound to the endogenous agonist kisspeptin-10 and a synthetic analog TAK-448. These structures reveal pivotal interactions between peptide ligands and KISS1R extracellular loops for receptor activation. Both peptides exhibit a conserved binding mode, unveiling their common activation mechanism. Intriguingly, KISS1R displays a distinct 40° angular deviation in its intracellular TM6 region compared to other Gq-coupled receptors, enabling distinct interactions with Gq. This study reveals the molecular intricacies governing ligand binding and activation of KISS1R, while highlighting its exceptional ability to couple with Gq. Our findings pave the way for structure-guided design of therapeutics targeting this physiologically indispensable receptor.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , Kisspeptins , Receptors, Kisspeptin-1 , Humans , Receptors, Kisspeptin-1/metabolism , Kisspeptins/metabolism , Kisspeptins/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Protein Binding , HEK293 Cells , Cryoelectron Microscopy
11.
Molecules ; 29(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38893478

ABSTRACT

Transient receptor potential melastatin-8 (TRPM8) is a cation channel that is activated by cold and "cooling agents" such as menthol and icilin, which induce a cold sensation. The stimulation of TRPM8 activates an intracellular signaling cascade that ultimately leads to a change in the gene expression pattern of the cells. Here, we investigate the TRPM8-induced signaling pathway that links TRPM8 channel activation to gene transcription. Using a pharmacological approach, we show that the inhibition of phosphatidylinositol 4-phosphate 5 kinase α (PIP5K), an enzyme essential for the biosynthesis of phosphatidylinositol 4,5-bisphosphate, attenuates TRPM8-induced gene transcription. Analyzing the link between TRPM8 and Gq proteins, we show that the pharmacological inhibition of the ßγ subunits impairs TRPM8 signaling. In addition, genetic studies show that TRPM8 requires an activated Gα subunit for signaling. In the nucleus, the TRPM8-induced signaling cascade triggers the activation of the transcription factor AP-1, a complex consisting of a dimer of basic region leucine zipper (bZIP) transcription factors. Here, we identify the bZIP protein c-Jun as an essential component of AP-1 within the TRPM8-induced signaling cascade. In summary, with PIP5K, Gq subunits, and c-Jun, we identified key molecules in TRPM8-induced signaling from the plasma membrane to the nucleus.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , Phosphotransferases (Alcohol Group Acceptor) , Signal Transduction , TRPM Cation Channels , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Transcription Factor AP-1/metabolism , HEK293 Cells , Proto-Oncogene Proteins c-jun/metabolism , Animals
13.
FEBS Lett ; 598(14): 1783-1791, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38757247

ABSTRACT

Certain amino acid sites of 5-HT2AR play crucial roles in interacting with various G proteins. Hallucinogens and non-hallucinogens both act on 5-HT2AR but mediate different pharmacological effects, possibly due to the coupling of different G proteins. Therefore, this study identified the binding sites of hallucinogens and non-hallucinogens with 5-HT2AR through molecular docking. We conducted site mutation to examine the impact of these sites on G proteins, in order to find out the sites that can distinguish the pharmacological effects of hallucinogens and non-hallucinogens. Our results indicate that I4.60A and S3.39A did not affect the ability of hallucinogens to activate Gq signaling, but significantly reduced Gs signaling activation by hallucinogens. These results suggest that S3.39 and I4.60 are important for the activation of Gs signaling by hallucinogens.


Subject(s)
Receptor, Serotonin, 5-HT2A , Serine , Signal Transduction , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/chemistry , Humans , HEK293 Cells , Serine/metabolism , Serine/chemistry , Binding Sites , Isoleucine/metabolism , Isoleucine/chemistry , Molecular Docking Simulation , Hallucinogens/pharmacology , Hallucinogens/metabolism , Hallucinogens/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Protein Binding
14.
Vestn Oftalmol ; 140(2): 5-13, 2024.
Article in Russian | MEDLINE | ID: mdl-38742493

ABSTRACT

Circumscribed choroidal hemangioma (CCH) and early non-pigmented choroidal melanoma (CM) have similar clinical, ultrasound and morphometric features, which in some cases makes their differential diagnosis difficult. There are few studies in the literature devoted to a comparative analysis of the molecular genetic features of CCH and non-pigmented CM, and the results of those studies are contradictory. PURPOSE: This study attempts to develop a method of non-invasive molecular genetic differential diagnostics of CCH and non-pigmented CM. MATERIAL AND METHODS: Based on the results of clinical and instrumental examination methods, 60 patients (60 eyes) with CCH (n=30) and non-pigmented CM (n=30) were included in this prospective study. The control group consisted of 30 individuals without intraocular tumors. Mutations in the GNAQ/GNA11 genes were determined by real-time PCR using the analysis of genomic circulating tumor DNA isolated from peripheral blood plasma. The average follow-up period was 12.1±1.8 months. RESULTS: The study revealed a significant association of mutations in exons 4 and 5 of the GNAQ/GNA11 genes with the presence of non-pigmented CM (27/30; 90%). These mutations were not detected in the group of patients with CCH. Mutations in exons 4 and 5 of the GNAQ/GNA11 genes were also not detected in the control group of healthy individuals. CONCLUSION: This study proposes a method of non-invasive and low-cost differential diagnostics based on molecular genetic analysis and detection of mutations in exons 4 and 5 of the GNAQ and GNA11 genes, which are specific for CM (90%).


Subject(s)
Choroid Neoplasms , Hemangioma , Melanoma , Humans , Choroid Neoplasms/genetics , Choroid Neoplasms/diagnosis , Male , Female , Middle Aged , Diagnosis, Differential , Hemangioma/genetics , Hemangioma/diagnosis , Adult , Melanoma/genetics , Melanoma/diagnosis , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Mutation , Choroid/diagnostic imaging , Choroid/pathology , GTP-Binding Protein alpha Subunits/genetics , Prospective Studies
15.
Nat Commun ; 15(1): 4307, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811567

ABSTRACT

G protein-coupled receptors (GPCRs) are sophisticated signaling machines able to simultaneously elicit multiple intracellular signaling pathways upon activation. Complete (in)activation of all pathways can be counterproductive for specific therapeutic applications. This is the case for the serotonin 2 A receptor (5-HT2AR), a prominent target for the treatment of schizophrenia. In this study, we elucidate the complex 5-HT2AR coupling signature in response to different signaling probes, and its physiological consequences by combining computational modeling, in vitro and in vivo experiments with human postmortem brain studies. We show how chemical modification of the endogenous agonist serotonin dramatically impacts the G protein coupling profile of the 5-HT2AR and the associated behavioral responses. Importantly, among these responses, we demonstrate that memory deficits are regulated by Gαq protein activation, whereas psychosis-related behavior is modulated through Gαi1 stimulation. These findings emphasize the complexity of GPCR pharmacology and physiology and open the path to designing improved therapeutics for the treatment of stchizophrenia.


Subject(s)
Memory Disorders , Psychotic Disorders , Receptor, Serotonin, 5-HT2A , Serotonin , Animals , Female , Humans , Male , Mice , Brain/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , HEK293 Cells , Memory Disorders/metabolism , Psychotic Disorders/metabolism , Psychotic Disorders/drug therapy , Receptor, Serotonin, 5-HT2A/metabolism , Schizophrenia/metabolism , Serotonin/metabolism , Signal Transduction
16.
Toxicol Appl Pharmacol ; 487: 116976, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38777097

ABSTRACT

Staff and animals in livestock buildings are constantly exposed to fine particulate matter (PM2.5), which affects their respiratory health. However, its exact pathogenic mechanism remains unclear. Regulator of G-protein signaling 2 (RGS2) has been reported to play a regulatory role in pneumonia. The aim of this study was to explore the therapeutic potential of RGS2 in cowshed PM2.5-induced respiratory damage. PM2.5 was collected from a cattle farm, and the alveolar macrophages (NR8383) of the model animal rat were stimulated with different treatment conditions of cowshed PM2.5. The RGS2 overexpression vector was constructed and transfected it into cells. Compared with the control group, cowshed PM2.5 significantly induced a decrease in cell viability and increased the levels of apoptosis and proinflammatory factor expression. Overexpression of RGS2 ameliorated the above-mentioned cellular changes induced by cowshed PM2.5. In addition, PM2.5 has significantly induced intracellular Ca2+ dysregulation. Affinity inhibition of Gq/11 by RGS2 attenuated the cytosolic calcium signaling pathway mediated by PLCß/IP3R. To further investigate the causes and mechanisms of action of differential RGS2 expression, the possible effects of oxidative stress and TLR2/4 activation were investigated. The results have shown that RGS2 expression was not only regulated by oxidative stress-induced nitric oxide during cowshed PM2.5 cells stimulation but the activation of TLR2/4 had also an important inhibitory effect on its protein expression. The present study demonstrates the intracellular Ca2+ regulatory role of RGS2 during cellular injury, which could be a potential target for the prevention and treatment of PM2.5-induced respiratory injury.


Subject(s)
Macrophages, Alveolar , Particulate Matter , RGS Proteins , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Animals , RGS Proteins/genetics , RGS Proteins/metabolism , Particulate Matter/toxicity , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Rats , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Cattle , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Cell Line , Calcium Signaling/drug effects , Calcium/metabolism , Apoptosis/drug effects , Air Pollutants/toxicity
17.
Mol Pharmacol ; 105(6): 386-394, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38641412

ABSTRACT

The M3 muscarinic acetylcholine receptor (M3R) is a G protein-coupled receptor (GPCR) that regulates important physiologic processes, including vascular tone, bronchoconstriction, and insulin secretion. It is expressed on a wide variety of cell types, including pancreatic beta, smooth muscle, neuronal, and immune cells. Agonist binding to the M3R is thought to initiate intracellular signaling events primarily through the heterotrimeric G protein Gq. However, reports differ on the ability of M3R to couple to other G proteins beyond Gq. Using members from the four primary G protein families (Gq, Gi, Gs, and G13) in radioligand binding, GTP turnover experiments, and cellular signaling assays, including live cell G protein dissociation and second messenger assessment of cAMP and inositol trisphosphate, we show that other G protein families, particularly Gi and Gs, can also interact with the human M3R. We further show that these interactions are productive as assessed by amplification of classic second messenger signaling events. Our findings demonstrate that the M3R is more promiscuous with respect to G protein interactions than previously appreciated. SIGNIFICANCE STATEMENT: The study reveals that the human M3 muscarinic acetylcholine receptor (M3R), known for its pivotal roles in diverse physiological processes, not only activates intracellular signaling via Gq as previously known but also functionally interacts with other G protein families such as Gi and Gs, expanding our understanding of its versatility in mediating cellular responses. These findings signify a broader and more complex regulatory network governed by M3R and have implications for therapeutic targeting.


Subject(s)
GTP-Binding Proteins , Receptor, Muscarinic M3 , Signal Transduction , Animals , Humans , CHO Cells , Cricetulus , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Receptor, Muscarinic M3/metabolism , Signal Transduction/physiology
18.
Nature ; 629(8011): 481-488, 2024 May.
Article in English | MEDLINE | ID: mdl-38632411

ABSTRACT

The human calcium-sensing receptor (CaSR) detects fluctuations in the extracellular Ca2+ concentration and maintains Ca2+ homeostasis1,2. It also mediates diverse cellular processes not associated with Ca2+ balance3-5. The functional pleiotropy of CaSR arises in part from its ability to signal through several G-protein subtypes6. We determined structures of CaSR in complex with G proteins from three different subfamilies: Gq, Gi and Gs. We found that the homodimeric CaSR of each complex couples to a single G protein through a common mode. This involves the C-terminal helix of each Gα subunit binding to a shallow pocket that is formed in one CaSR subunit by all three intracellular loops (ICL1-ICL3), an extended transmembrane helix 3 and an ordered C-terminal region. G-protein binding expands the transmembrane dimer interface, which is further stabilized by phospholipid. The restraint imposed by the receptor dimer, in combination with ICL2, enables G-protein activation by facilitating conformational transition of Gα. We identified a single Gα residue that determines Gq and Gs versus Gi selectivity. The length and flexibility of ICL2 allows CaSR to bind all three Gα subtypes, thereby conferring capacity for promiscuous G-protein coupling.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Receptors, Calcium-Sensing , Humans , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/chemistry , Models, Molecular , Protein Binding , Protein Multimerization , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/chemistry , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Binding Sites , Protein Structure, Secondary , Substrate Specificity
19.
Commun Biol ; 7(1): 362, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38521872

ABSTRACT

Muscarinic acetylcholine receptor M3 (M3) and its downstream effector Gq/11 are critical drug development targets due to their involvement in physiopathological processes. Although the structure of the M3-miniGq complex was recently published, the lack of information on the intracellular loop 3 (ICL3) of M3 and extensive modification of Gαq impedes the elucidation of the molecular mechanism of M3-Gq coupling under more physiological condition. Here, we describe the molecular mechanism underlying the dynamic interactions between full-length wild-type M3 and Gq using hydrogen-deuterium exchange mass spectrometry and NanoLuc Binary Technology-based cell systems. We propose a detailed analysis of M3-Gq coupling through examination of previously well-defined binding interfaces and neglected regions. Our findings suggest potential binding interfaces between M3 and Gq in pre-assembled and functionally active complexes. Furthermore, M3 ICL3 negatively affected M3-Gq coupling, and the Gαq AHD underwent unique conformational changes during M3-Gq coupling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , Receptors, Muscarinic , Receptors, Muscarinic/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry
20.
JCI Insight ; 9(9)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530370

ABSTRACT

Fibroblast growth factor 23 (FGF23) production has recently been shown to increase downstream of Gαq/11-PKC signaling in osteocytes. Inactivating mutations in the gene encoding Gα11 (GNA11) cause familial hypocalciuric hypercalcemia (FHH) due to impaired calcium-sensing receptor signaling. We explored the effect of Gα11 deficiency on FGF23 production in mice with heterozygous (Gna11+/-) or homozygous (Gna11-/-) ablation of Gna11. Both Gna11+/- and Gna11-/- mice demonstrated hypercalcemia and mildly raised parathyroid hormone levels, consistent with FHH. Strikingly, these mice also displayed increased serum levels of total and intact FGF23 and hypophosphatemia. Gna11-/- mice showed augmented Fgf23 mRNA levels in the liver and heart, but not in bone or bone marrow, and also showed evidence of systemic inflammation with elevated serum IL-1ß levels. Furin gene expression was significantly increased in the Gna11-/- liver, suggesting enhanced FGF23 cleavage despite the observed rise in circulating intact FGF23 levels. Gna11-/- mice had normal renal function and reduced serum levels of glycerol-3-phosphate, excluding kidney injury as the primary cause of elevated intact FGF23 levels. Thus, Gα11 ablation caused systemic inflammation and excess serum FGF23 in mice, suggesting that patients with FHH - at least those with GNA11 mutations - may be at risk for these complications.


Subject(s)
Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , GTP-Binding Protein alpha Subunits, Gq-G11 , Hypercalcemia , Mice, Knockout , Animals , Female , Male , Mice , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypercalcemia/genetics , Hypercalcemia/congenital , Hypercalcemia/blood , Hypercalcemia/metabolism , Hypophosphatemia/genetics , Hypophosphatemia/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/blood , Liver/metabolism , Parathyroid Hormone/blood , Parathyroid Hormone/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL