Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30.411
Filter
1.
Mol Biol Rep ; 51(1): 704, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824233

ABSTRACT

BACKGROUND: Tumor modeling using organoids holds potential in studies of cancer development, enlightening both the intracellular and extracellular molecular mechanisms behind different cancer types, biobanking, and drug screening. Intestinal organoids can be generated in vitro using a unique type of adult stem cells which are found at the base of crypts and are characterized by their high Lgr5 expression levels. METHODS AND RESULTS: In this study, we successfully established intestinal cancer organoid models by using both the BALB/c derived and mouse embryonic stem cells (mESCs)-derived intestinal organoids. In both cases, carcinogenesis-like model was developed by using azoxymethane (AOM) treatment. Carcinogenesis-like model was verified by H&E staining, immunostaining, relative mRNA expression analysis, and LC/MS analysis. The morphologic analysis demonstrated that the number of generated organoids, the number of crypts, and the intensity of the organoids were significantly augmented in AOM-treated intestinal organoids compared to non-AOM-treated ones. Relative mRNA expression data revealed that there was a significant increase in both Wnt signaling pathway-related genes and pluripotency transcription factors in the AOM-induced intestinal organoids. CONCLUSION: We successfully developed simple carcinogenesis-like models using mESC-based and Lgr5 + stem cell-based intestinal organoids. Intestinal organoid based carcinogenesi models might be used for personalized cancer therapy in the future.


Subject(s)
Azoxymethane , Carcinogenesis , Mouse Embryonic Stem Cells , Organoids , Wnt Signaling Pathway , Animals , Organoids/metabolism , Organoids/pathology , Mice , Azoxymethane/toxicity , Carcinogenesis/pathology , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Mouse Embryonic Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Mice, Inbred BALB C , Intestines/pathology , Intestinal Neoplasms/pathology , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Disease Models, Animal , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology
2.
Sci Signal ; 17(839): eade8041, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833528

ABSTRACT

A long-standing question in the field of signal transduction is how distinct signaling pathways interact with each other to control cell behavior. Growth factor receptors and G protein-coupled receptors (GPCRs) are the two major signaling hubs in eukaryotes. Given that the mechanisms by which they signal independently have been extensively characterized, we investigated how they may cross-talk with each other. Using linear ion trap mass spectrometry and cell-based biophysical, biochemical, and phenotypic assays, we found at least three distinct ways in which epidermal growth factor affected canonical G protein signaling by the Gi-coupled GPCR CXCR4 through the phosphorylation of Gαi. Phosphomimicking mutations in two residues in the αE helix of Gαi (tyrosine-154/tyrosine-155) suppressed agonist-induced Gαi activation while promoting constitutive Gßγ signaling. Phosphomimicking mutations in the P loop (serine-44, serine-47, and threonine-48) suppressed Gi activation entirely, thus completely segregating growth factor and GPCR pathways. As expected, most of the phosphorylation events appeared to affect intrinsic properties of Gαi proteins, including conformational stability, nucleotide binding, and the ability to associate with and to release Gßγ. However, one phosphomimicking mutation, targeting the carboxyl-terminal residue tyrosine-320, promoted mislocalization of Gαi from the plasma membrane, a previously uncharacterized mechanism of suppressing GPCR signaling through G protein subcellular compartmentalization. Together, these findings elucidate not only how growth factor and chemokine signals cross-talk through the phosphorylation-dependent modulation of Gαi but also how such cross-talk may generate signal diversity.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , Receptors, CXCR4 , Signal Transduction , Phosphorylation , Humans , HEK293 Cells , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Animals
3.
Biochemistry (Mosc) ; 89(4): 747-764, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38831510

ABSTRACT

G protein-coupled receptors (GPCRs) play a key role in the transduction of extracellular signals to cells and regulation of many biological processes, which makes these membrane proteins one of the most important targets for pharmacological agents. A significant increase in the number of resolved atomic structures of GPCRs has opened the possibility of developing pharmaceuticals targeting these receptors via structure-based drug design (SBDD). SBDD employs information on the structure of receptor-ligand complexes to search for selective ligands without the need for an extensive high-throughput experimental ligand screening and can significantly expand the chemical space for ligand search. In this review, we describe the process of deciphering GPCR structures using X-ray diffraction analysis and cryoelectron microscopy as an important stage in the rational design of drugs targeting this receptor class. Our main goal was to present modern developments and key features of experimental methods used in SBDD of GPCR-targeting agents to a wide range of specialists.


Subject(s)
Drug Design , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Humans , Ligands , Cryoelectron Microscopy , Animals , X-Ray Diffraction
4.
Nat Commun ; 15(1): 4687, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824166

ABSTRACT

Ligand-induced activation of G protein-coupled receptors (GPCRs) can initiate signaling through multiple distinct pathways with differing biological and physiological outcomes. There is intense interest in understanding how variation in GPCR ligand structure can be used to promote pathway selective signaling ("biased agonism") with the goal of promoting desirable responses and avoiding deleterious side effects. Here we present an approach in which a conventional peptide ligand for the type 1 parathyroid hormone receptor (PTHR1) is converted from an agonist which induces signaling through all relevant pathways to a compound that is highly selective for a single pathway. This is achieved not through variation in the core structure of the agonist, but rather by linking it to a nanobody tethering agent that binds with high affinity to a separate site on the receptor not involved in signal transduction. The resulting conjugate represents the most biased agonist of PTHR1 reported to date. This approach holds promise for facile generation of pathway selective ligands for other GPCRs.


Subject(s)
Receptor, Parathyroid Hormone, Type 1 , Receptors, G-Protein-Coupled , Signal Transduction , Single-Domain Antibodies , Ligands , Humans , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptor, Parathyroid Hormone, Type 1/agonists , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , HEK293 Cells , Signal Transduction/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Protein Binding , Animals , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism
5.
J Obstet Gynaecol ; 44(1): 2347430, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38835234

ABSTRACT

BACKGROUND: At present, the discovery of new biomarkers is of great significance for the early diagnosis, treatment and prognosis assessment of ovarian cancer. Previous findings indicated that aberrant G-protein-coupled receptor 176 (GPR176) expression might contribute to tumorigenesis and subsequent progression. However, the expression of GPR176 and the molecular mechanisms in ovarian cancer had not been investigated. METHODS: GPR176 expression was compared with clinicopathological features of ovarian cancer using immunohistochemical and bioinformatics analyses. GPR176-related genes and pathways were analysed using bioinformatics analysis. Additionally, the effects of GPR176 on ovarian cancer cell phenotypes were investigated. RESULTS: GPR176 expression positively correlated with elder age, clinicopathological staging, tumour residual status, and unfavourable survival of ovarian cancer, but negatively with purity loss, infiltration of B cells, and CD8+ T cells. Gene Set Enrichment Analysis showed that differential expression of GPR176 was involved in focal adhesion, ECM-receptor interaction, cell adhesion molecules and so on. STRING and Cytoscape were used to determine the top 10 nodes. Kyoto Encyclopaedia of Genes and Genomes analysis indicated that GPR176-related genes were involved in the ECM structural constituent and organisation and so on. GPR176 overexpression promoted the proliferation, anti-apoptosis, anti-pyroptosis, migration and invasion of ovarian cancer cells with overexpression of N-cadherin, Zeb1, Snail, Twist1, and under-expression of gasdermin D, caspase 1, and E-cadherin. CONCLUSION: GPR176 might be involved in the progression of ovarian cancer. It might be used as a biomarker to indicate the aggressive behaviour and poor prognosis of ovarian cancer and a target of genetic therapy.


Ovarian cancer is a gynecological cancer with high mortality. Due to the limited screening tests and treatments available, most ovarian cancer patients are diagnosed at a late stage and the prognosis is poor. The addition of new cancer diagnostic biomarkers and new intervention targets may improve quality of life and survival for patients with ovarian cancer. Previous studies have revealed the aberrant GPR176 expression might contribute to tumorigenesis and subsequent progression in many other tumours. In our study, GPR176 was found to promote the proliferation, anti-apoptosis, anti-pyroptosis, migration and invasion, EMT, and weakening the cellular adhesion of ovarian cancer cells, and involved in the Bcl-2/Bax or the PI3K/Akt/mTOR pathway. Therefore, abnormal expression of GPR176 might be served as a biomarker for aggressive behaviour and poor prognosis of ovarian cancer and a target for gene therapy.


Subject(s)
Ovarian Neoplasms , Receptors, G-Protein-Coupled , Humans , Female , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Middle Aged , Genetic Therapy/methods , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Computational Biology , Prognosis , Cell Proliferation/genetics , Carcinogenesis/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
6.
J Nanobiotechnology ; 22(1): 241, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735933

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) incidence is increasing in recent years due to intestinal flora imbalance, making oral probiotics a hotspot for research. However, numerous studies related to intestinal flora regulation ignore its internal mechanisms without in-depth research. RESULTS: Here, we developed a probiotic microgel delivery system (L.r@(SA-CS)2) through the layer-by-layer encapsulation technology of alginate (SA) and chitosan (CS) to improve gut microbiota dysbiosis and enhance anti-tumor therapeutic effect. Short chain fatty acids (SCFAs) produced by L.r have direct anti-tumor effects. Additionally, it reduces harmful bacteria such as Proteobacteria and Fusobacteriota, and through bacteria mutualophy increases beneficial bacteria such as Bacteroidota and Firmicutes which produce butyric acid. By binding to the G protein-coupled receptor 109A (GPR109A) on the surface of colonic epithelial cells, butyric acid can induce apoptosis in abnormal cells. Due to the low expression of GPR109A in colon cancer cells, MK-6892 (MK) can be used to stimulate GPR109A. With increased production of butyrate, activated GPR109A is able to bind more butyrate, which further promotes apoptosis of cancer cells and triggers an antitumor response. CONCLUSION: It appears that the oral administration of L.r@(SA-CS)2 microgels may provide a treatment option for CRC by modifying the gut microbiota.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Gastrointestinal Microbiome/drug effects , Probiotics/pharmacology , Humans , Fatty Acids, Volatile/metabolism , Animals , Limosilactobacillus reuteri/metabolism , Mice , Chitosan/chemistry , Alginates/chemistry , Alginates/pharmacology , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Administration, Oral , Colorectal Neoplasms/drug therapy , Cell Line, Tumor , Receptors, G-Protein-Coupled/metabolism , Microgels/chemistry , Mice, Inbred BALB C , Butyric Acid/pharmacology , Butyric Acid/metabolism
7.
J Zhejiang Univ Sci B ; 25(5): 389-409, 2024 May 15.
Article in English, Chinese | MEDLINE | ID: mdl-38725339

ABSTRACT

The short neuropeptide F (sNPF) family of peptides is a multifunctional group of neurohormones involved in the regulation of various physiological processes in insects. They have been found in a broad spectrum of species, but the number of isoforms in the precursor molecule varies from one to four. The receptor for sNPF (sNPFR), which belongs to the G protein-coupled receptor family, has been characterized in various insect orders and was shown to be an ortholog of the mammalian prolactin-releasing peptide receptor (PrPR). The sNPF signaling pathway interacts with other neurohormones such as insulin-like peptides, SIFamide, and pigment-dispersing factors (PDFs) to regulate various processes. The main physiological function of sNPF seems to be involved in the regulation of feeding, but the observed effects are species-specific. sNPF is also connected with the regulation of foraging behavior and the olfactory system. The influence of sNPF on feeding and thus energy metabolism may also indirectly affect other vital processes, such as reproduction and development. In addition, these neurohormones are involved in the regulation of locomotor activity and circadian rhythm in insects. This review summarizes the current state of knowledge about the sNPF system in insects.


Subject(s)
Insecta , Neuropeptides , Signal Transduction , Animals , Neuropeptides/metabolism , Neuropeptides/physiology , Insecta/physiology , Insecta/metabolism , Circadian Rhythm/physiology , Feeding Behavior , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Energy Metabolism
8.
Food Res Int ; 183: 114234, 2024 May.
Article in English | MEDLINE | ID: mdl-38760147

ABSTRACT

Bitterness is one of the five basic tastes generally considered undesirable. The widespread presence of bitter compounds can negatively affect the palatability of foods. The classification and sensory evaluation of bitter compounds have been the focus in recent research. However, the rigorous identification of bitter tastes and further studies to effectively mask or remove them have not been thoroughly evaluated. The present paper focuses on identification of bitter compounds in foods, structural-based activation of bitter receptors, and strategies to reduce bitter compounds in foods. It also discusses the roles of metabolomics and virtual screening analysis in bitter taste. The identification of bitter compounds has seen greater success through metabolomics with multivariate statistical analysis compared to conventional chromatography, HPLC, LC-MS, and NMR techniques. However, to avoid false positives, sensory recognition should be combined. Bitter perception involves the structural activation of bitter taste receptors (TAS2Rs). Only 25 human TAS2Rs have been identified as responsible for recognizing numerous bitter compounds, showcasing their high structural diversity to bitter agonists. Thus, reducing bitterness can be achieved through several methods. Traditionally, the removal or degradation of bitter substances has been used for debittering, while the masking of bitterness presents a new effective approach to improving food flavor. Future research in food bitterness should focus on identifying unknown bitter compounds in food, elucidating the mechanisms of activation of different receptors, and developing debittering techniques based on the entire food matrix.


Subject(s)
Receptors, G-Protein-Coupled , Taste Perception , Taste , Humans , Receptors, G-Protein-Coupled/metabolism , Flavoring Agents/analysis , Metabolomics/methods , Food Analysis/methods , Food Preferences
9.
PLoS Biol ; 22(5): e3002599, 2024 May.
Article in English | MEDLINE | ID: mdl-38713721

ABSTRACT

Synaptic adhesion molecules (SAMs) are evolutionarily conserved proteins that play an important role in the form and function of neuronal synapses. Teneurins (Tenms) and latrophilins (Lphns) are well-known cell adhesion molecules that form a transsynaptic complex. Recent studies suggest that Tenm3 and Lphn2 (gene symbol Adgrl2) are involved in hippocampal circuit assembly via their topographical expression. However, it is not known whether other teneurins and latrophilins display similar topographically restricted expression patterns during embryonic and postnatal development. Here, we reveal the cartography of all teneurin (Tenm1-4) and latrophilin (Lphn1-3 [Adgrl1-3]) paralog expression in the mouse hippocampus across prenatal and postnatal development as monitored by large-scale single-molecule RNA in situ hybridization mapping. Our results identify a striking heterogeneity in teneurin and latrophilin expression along the spatiotemporal axis of the hippocampus. Tenm2 and Tenm4 expression levels peak at the neonatal stage when compared to Tenm1 and Tenm3, while Tenm1 expression is restricted to the postnatal pyramidal cell layer. Tenm4 expression in the dentate gyrus (DG) exhibits an opposing topographical expression pattern in the embryonic and neonatal hippocampus. Our findings were validated by analyses of multiple RNA-seq datasets at bulk, single-cell, and spatial levels. Thus, our study presents a comprehensive spatiotemporal map of Tenm and Lphn expression in the hippocampus, showcasing their diverse expression patterns across developmental stages in distinct spatial axes.


Subject(s)
Gene Expression Regulation, Developmental , Hippocampus , Nerve Tissue Proteins , Receptors, Peptide , Animals , Hippocampus/metabolism , Hippocampus/embryology , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Receptors, Peptide/metabolism , Receptors, Peptide/genetics , Female , Mice, Inbred C57BL , Male , Tenascin , Receptors, G-Protein-Coupled
10.
Cell Genom ; 4(5): 100560, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38723606

ABSTRACT

GPCR signaling can contribute to establishing the tumor microenvironment and influence the progression and metabolism of tumors. Arora et al.1 describe a systems-level approach to investigate the patterns of co-expression of GPCR signaling pathway networks across diverse tumors and identify network components that correlate with patient-survival data across different cancer types.


Subject(s)
Neoplasms , Receptors, G-Protein-Coupled , Signal Transduction , Humans , Receptors, G-Protein-Coupled/metabolism , Neoplasms/metabolism , Neoplasms/drug therapy , Tumor Microenvironment , Molecular Targeted Therapy/methods
11.
Protein Sci ; 33(6): e5000, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38747401

ABSTRACT

G protein-coupled receptors (GPCRs) are one of the most important families of targets for drug discovery. One of the limiting steps in the study of GPCRs has been their stability, with significant and time-consuming protein engineering often used to stabilize GPCRs for structural characterization and drug screening. Unfortunately, computational methods developed using globular soluble proteins have translated poorly to the rational engineering of GPCRs. To fill this gap, we propose GPCR-tm, a novel and personalized structurally driven web-based machine learning tool to study the impacts of mutations on GPCR stability. We show that GPCR-tm performs as well as or better than alternative methods, and that it can accurately rank the stability changes of a wide range of mutations occurring in various types of class A GPCRs. GPCR-tm achieved Pearson's correlation coefficients of 0.74 and 0.46 on 10-fold cross-validation and blind test sets, respectively. We observed that the (structural) graph-based signatures were the most important set of features for predicting destabilizing mutations, which points out that these signatures properly describe the changes in the environment where the mutations occur. More specifically, GPCR-tm was able to accurately rank mutations based on their effect on protein stability, guiding their rational stabilization. GPCR-tm is available through a user-friendly web server at https://biosig.lab.uq.edu.au/gpcr_tm/.


Subject(s)
Protein Engineering , Protein Stability , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Protein Engineering/methods , Humans , Machine Learning , Mutation , Software , Models, Molecular
12.
J Transl Med ; 22(1): 450, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741146

ABSTRACT

BACKGROUND: Estetrol (E4) is a natural estrogen produced by the fetal liver during pregnancy. Due to its favorable safety profile, E4 was recently approved as estrogenic component of a new combined oral contraceptive. E4 is a selective ligand of estrogen receptor (ER)α and ERß, but its binding to the G Protein-Coupled Estrogen Receptor (GPER) has not been described to date. Therefore, we aimed to explore E4 action in GPER-positive Triple-Negative Breast Cancer (TNBC) cells. METHODS: The potential interaction between E4 and GPER was investigated by molecular modeling and binding assays. The whole transcriptomic modulation triggered by E4 in TNBC cells via GPER was explored through high-throughput RNA sequencing analyses. Gene and protein expression evaluations as well as migration and invasion assays allowed us to explore the involvement of the GPER-mediated induction of the plasminogen activator inhibitor type 2 (SERPINB2) in the biological responses triggered by E4 in TNBC cells. Furthermore, bioinformatics analysis was aimed at recognizing the biological significance of SERPINB2 in ER-negative breast cancer patients. RESULTS: After the molecular characterization of the E4 binding capacity to GPER, RNA-seq analysis revealed that the plasminogen activator inhibitor type 2 (SERPINB2) is one of the most up-regulated genes by E4 in a GPER-dependent manner. Worthy, we demonstrated that the GPER-mediated increase of SERPINB2 is engaged in the anti-migratory and anti-invasive effects elicited by E4 in TNBC cells. In accordance with these findings, a correlation between SERPINB2 levels and a good clinical outcome was found in ER-negative breast cancer patients. CONCLUSIONS: Overall, our results provide new insights into the mechanisms through which E4 can halt migratory and invasive features of TNBC cells.


Subject(s)
Cell Movement , Estetrol , Gene Expression Regulation, Neoplastic , Plasminogen Activator Inhibitor 2 , Receptors, Estrogen , Receptors, G-Protein-Coupled , Signal Transduction , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/genetics , Cell Movement/drug effects , Cell Line, Tumor , Receptors, G-Protein-Coupled/metabolism , Receptors, Estrogen/metabolism , Estetrol/pharmacology , Estetrol/metabolism , Female , Plasminogen Activator Inhibitor 2/metabolism , Protein Binding/drug effects , Neoplasm Invasiveness
13.
Front Immunol ; 15: 1374425, 2024.
Article in English | MEDLINE | ID: mdl-38745644

ABSTRACT

Various gut bacteria, including Lactobacillus plantarum, possess several enzymes that produce hydroxy fatty acids (FAs), oxo FAs, conjugated FAs, and partially saturated FAs from polyunsaturated FAs as secondary metabolites. Among these derivatives, we identified 10-oxo-cis-6,trans-11-octadecadienoic acid (γKetoC), a γ-linolenic acid (GLA)-derived enon FA, as the most effective immunomodulator, which inhibited the antigen-induced immunoactivation and LPS-induced production of inflammatory cytokines. The treatment with γKetoC significantly suppressed proliferation of CD4+ T cells, LPS-induced activation of bone marrow-derived dendritic cells (BMDCs), and LPS-induced IL-6 release from peritoneal cells, splenocytes, and CD11c+ cells isolated from the spleen. γKetoC also inhibited the release of inflammatory cytokines from BMDCs stimulated with poly-I:C, R-848, or CpG. Further in vitro experiments using an agonist of GPR40/120 suggested the involvement of these GPCRs in the effects of γKetoC on DCs. We also found that γKetoC stimulated the NRF2 pathway in DCs, and the suppressive effects of γKetoC and agonist of GPR40/120 on the release of IL-6 and IL-12 were reduced in Nrf2-/- BMDCs. We evaluated the role of NRF2 in the anti-inflammatory effects of γKetoC in a dextran sodium sulfate-induced colitis model. The oral administration of γKetoC significantly reduced body weight loss, improved stool scores, and attenuated atrophy of the colon, in wild-type C57BL/6 and Nrf2+/- mice with colitis. In contrast, the pathology of colitis was deteriorated in Nrf2-/- mice even with the administration of γKetoC. Collectively, the present results demonstrated the involvement of the NRF2 pathway and GPCRs in γKetoC-mediated anti-inflammatory responses.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Receptors, G-Protein-Coupled , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Mice , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Mice, Knockout , Cytokines/metabolism , Disease Models, Animal , Dextran Sulfate , Oleic Acids/pharmacology , Lactobacillus plantarum , Colitis/metabolism , Colitis/chemically induced , Colitis/drug therapy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/drug effects , Male
14.
Function (Oxf) ; 5(3): zqae010, 2024.
Article in English | MEDLINE | ID: mdl-38706960

ABSTRACT

The Olfr78 gene encodes a G-protein-coupled olfactory receptor that is expressed in several ectopic sites. Olfr78 is one of the most abundant mRNA species in carotid body (CB) glomus cells. These cells are the prototypical oxygen (O2) sensitive arterial chemoreceptors, which, in response to lowered O2 tension (hypoxia), activate the respiratory centers to induce hyperventilation. It has been proposed that Olfr78 is a lactate receptor and that glomus cell activation by the increase in blood lactate mediates the hypoxic ventilatory response (HVR). However, this proposal has been challenged by several groups showing that Olfr78 is not a physiologically relevant lactate receptor and that the O2-based regulation of breathing is not affected in constitutive Olfr78 knockout mice. In another study, constitutive Olfr78 knockout mice were reported to have altered systemic and CB responses to mild hypoxia. To further characterize the functional role of Olfr78 in CB glomus cells, we here generated a conditional Olfr78 knockout mouse strain and then restricted the knockout to glomus cells and other catecholaminergic cells by crossing with a tyrosine hydroxylase-specific Cre driver strain (TH-Olfr78 KO mice). We find that TH-Olfr78 KO mice have a normal HVR. Interestingly, glomus cells of TH-Olfr78 KO mice exhibit molecular and electrophysiological alterations as well as a reduced dopamine content in secretory vesicles and neurosecretory activity. These functional characteristics resemble those of CB neuroblasts in wild-type mice. We suggest that, although Olfr78 is not essential for CB O2 sensing, activation of Olfr78-dependent pathways is required for maturation of glomus cells.


Subject(s)
Carotid Body , Receptors, Odorant , Tyrosine 3-Monooxygenase , Animals , Male , Mice , Carotid Body/metabolism , Hypoxia/metabolism , Hypoxia/genetics , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/genetics
15.
Pharmacol Rev ; 76(3): 358-387, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697858

ABSTRACT

G-protein coupled receptors (GPCRs) transduce a wide range of extracellular signals. They are key players in the majority of biologic functions including vision, olfaction, chemotaxis, and immunity. However, as essential as most of them are to body function and homeostasis, overactivation of GPCRs has been implicated in many pathologic diseases such as cancer, asthma, and heart failure (HF). Therefore, an important feature of G protein signaling systems is the ability to control GPCR responsiveness, and one key process to control overstimulation involves initiating receptor desensitization. A number of steps are appreciated in the desensitization process, including cell surface receptor phosphorylation, internalization, and downregulation. Rapid or short-term desensitization occurs within minutes and involves receptor phosphorylation via the action of intracellular protein kinases, the binding of ß-arrestins, and the consequent uncoupling of GPCRs from their cognate heterotrimeric G proteins. On the other hand, long-term desensitization occurs over hours to days and involves receptor downregulation or a decrease in cell surface receptor protein level. Of the proteins involved in this biologic phenomenon, ß-arrestins play a particularly significant role in both short- and long-term desensitization mechanisms. In addition, ß-arrestins are involved in the phenomenon of biased agonism, where the biased ligand preferentially activates one of several downstream signaling pathways, leading to altered cellular responses. In this context, this review discusses the different patterns of desensitization of the α 1-, α 2- and the ß adrenoceptors and highlights the role of ß-arrestins in regulating physiologic responsiveness through desensitization and biased agonism. SIGNIFICANCE STATEMENT: A sophisticated network of proteins orchestrates the molecular regulation of GPCR activity. Adrenoceptors are GPCRs that play vast roles in many physiological processes. Without tightly controlled desensitization of these receptors, homeostatic imbalance may ensue, thus precipitating various diseases. Here, we critically appraise the mechanisms implicated in adrenoceptor desensitization. A better understanding of these mechanisms helps identify new druggable targets within the GPCR desensitization machinery and opens exciting therapeutic fronts in the treatment of several pathologies.


Subject(s)
Signal Transduction , Humans , Animals , Receptors, Adrenergic/metabolism , Receptors, Adrenergic/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , beta-Arrestins/metabolism
16.
Front Immunol ; 15: 1399459, 2024.
Article in English | MEDLINE | ID: mdl-38812508

ABSTRACT

Initiation of the bradykinin generation cascade is responsible for the occurrence of attacks in some types of angioedema without wheals. Hereditary angioedema due to C1 inhibitor deficiency (HAE-C1-INH) is one such clinical entity. In this paper, we explore the existing evidence that mast cells (MCs) degranulation may contribute to the activation of the kallikrein-kinin system cascade, followed by bradykinin formation and angioedema. We present the multidirectional effects of MC-derived heparin and other polyanions on the major components of the kinin-kallikrein system, particularly on the factor XII activation. Although, bradykinin- and histamine-mediated symptoms are distinct clinical phenomena, they share some common features, such as some similar triggers and a predilection to occur at sites where mast cells reside, namely the skin and mucous membranes. In addition, recent observations indicate a high incidence of hypersensitivity reactions associated with MC degranulation in the HAE-C1-INH patient population. However, not all of these can be explained by IgE-dependent mechanisms. Mast cell-related G protein-coupled receptor-X2 (MRGPRX2), which has recently attracted scientific interest, may be involved in the activation of MCs through a different pathway. Therefore, we reviewed MRGPRX2 ligands that HAE-C1-INH patients may be exposed to in their daily lives and that may affect MCs degranulation. We also discussed the known inter- and intra-individual variability in the course of HAE-C1-INH in relation to factors responsible for possible variability in the strength of the response to MRGPRX2 receptor stimulation. The above issues raise several questions for future research. It is not known to what extent a prophylactic or therapeutic intervention targeting the pathways of one mechanism (mast cell degranulation) may affect the other (bradykinin production), or whether the number of mast cells at a specific body site and their reactivity to triggers such as pressure, allergens or MRGPRX2 agonists may influence the occurrence of HAE-C1-INH attacks at that site.


Subject(s)
Bradykinin , Cell Degranulation , Mast Cells , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Humans , Mast Cells/immunology , Mast Cells/metabolism , Bradykinin/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Angioedema/metabolism , Angioedema/immunology , Angioedema/etiology , Nerve Tissue Proteins/metabolism , Kallikrein-Kinin System/physiology
17.
Structure ; 32(5): 517-519, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701749

ABSTRACT

G-protein-coupled receptor (GPCR) activation relies on conformational sampling, a nuanced but functionally key behavior well suited to elucidation by nuclear magnetic resonance (NMR) spectroscopy. In this issue of Structure, Thakur et al.1 demonstrate that judicious choice of experimental conditions for 19F NMR studies of a GPCR enables rationalization of functional and pharmacological behavior, leading to testable hypotheses correlating structure, dynamics, and function.


Subject(s)
Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/chemistry , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Magnetic Resonance Spectroscopy/methods
19.
PLoS One ; 19(5): e0298239, 2024.
Article in English | MEDLINE | ID: mdl-38691547

ABSTRACT

The sweet taste receptor, TAS1R2-TAS1R3, is expressed in taste bud cells, where it conveys sweetness, and also in intestinal enteroendocrine cells, where it may facilitate glucose absorption and assimilation. In the present study, our objective was to determine whether TAS1R2-TAS1R3 influences glucose metabolism bidirectionally via hyperactivation with 5 mM sucralose (n = 12) and inhibition with 2 mM sodium lactisole (n = 10) in mixture with 75 g glucose loads during oral glucose tolerance tests (OGTTs) in healthy humans. Plasma glucose, insulin, and glucagon were measured before, during, and after OGTTs up to 120 minutes post-prandially. We also assessed individual participants' sweet taste responses to sucralose and their sensitivities to lactisole sweetness inhibition. The addition of sucralose to glucose elevated plasma insulin responses to the OGTT (F(1, 11) = 4.55, p = 0.056). Sucralose sweetness ratings were correlated with early increases in plasma glucose (R2 = 0.41, p<0.05), as well as increases in plasma insulin (R2 = 0.38, p<0.05) when sucralose was added to the OGTT (15 minute AUC). Sensitivity to lactisole sweetness inhibition was correlated with decreased plasma glucose (R2 = 0.84, p<0.01) when lactisole was added to the OGTT over the whole test (120 minute AUC). In summary, stimulation and inhibition of the TAS1R2-TAS1R3 receptor demonstrates that TAS1R2-TAS1R3 helps regulate glucose metabolism in humans and may have translational implications for metabolic disease risk.


Subject(s)
Benzene Derivatives , Blood Glucose , Glucose Tolerance Test , Insulin , Receptors, G-Protein-Coupled , Sucrose , Sucrose/analogs & derivatives , Humans , Receptors, G-Protein-Coupled/metabolism , Male , Adult , Female , Sucrose/metabolism , Blood Glucose/metabolism , Insulin/metabolism , Insulin/blood , Taste/physiology , Young Adult , Thiazoles/pharmacology , Glucose/metabolism , Glucagon/metabolism , Glucagon/blood , Sweetening Agents/pharmacology
20.
Sci Signal ; 17(836): eadq3321, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38743810

ABSTRACT

Activation of GPR81 in white adipose tissue by lactate results in cancer-associated cachexia.


Subject(s)
Cachexia , Lactic Acid , Neoplasms , Receptors, G-Protein-Coupled , Humans , Lactic Acid/metabolism , Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Cachexia/metabolism , Animals , Adipose Tissue, White/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...