Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.894
Filter
1.
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
2.
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
4.
PLoS Biol ; 21(12): e3002188, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38055679

ABSTRACT

Chemokine-like receptor 1 (CMKLR1), also known as chemerin receptor 23 (ChemR23) or chemerin receptor 1, is a chemoattractant G protein-coupled receptor (GPCR) that responds to the adipokine chemerin and is highly expressed in innate immune cells, including macrophages and neutrophils. The signaling pathways of CMKLR1 can lead to both pro- and anti-inflammatory effects depending on the ligands and physiological contexts. To understand the molecular mechanisms of CMKLR1 signaling, we determined a high-resolution cryo-electron microscopy (cryo-EM) structure of the CMKLR1-Gi signaling complex with chemerin9, a nanopeptide agonist derived from chemerin, which induced complex phenotypic changes of macrophages in our assays. The cryo-EM structure, together with molecular dynamics simulations and mutagenesis studies, revealed the molecular basis of CMKLR1 signaling by elucidating the interactions at the ligand-binding pocket and the agonist-induced conformational changes. Our results are expected to facilitate the development of small molecule CMKLR1 agonists that mimic the action of chemerin9 to promote the resolution of inflammation.


Subject(s)
Intercellular Signaling Peptides and Proteins , Signal Transduction , Cryoelectron Microscopy , Receptors, G-Protein-Coupled/physiology , Chemokines/physiology
5.
Proc Natl Acad Sci U S A ; 120(43): e2311131120, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37844228

ABSTRACT

Many neurons in the central nervous system produce a single primary cilium that serves as a specialized signaling organelle. Several neuromodulatory G-protein-coupled receptors (GPCRs) localize to primary cilia in neurons, although it is not understood how GPCR signaling from the cilium impacts circuit function and behavior. We find that the vertebrate ancient long opsin A (VALopA), a Gi-coupled GPCR extraretinal opsin, targets to cilia of zebrafish spinal neurons. In the developing 1-d-old zebrafish, brief light activation of VALopA in neurons of the central pattern generator circuit for locomotion leads to sustained inhibition of coiling, the earliest form of locomotion. We find that a related extraretinal opsin, VALopB, is also Gi-coupled, but is not targeted to cilia. Light-induced activation of VALopB also suppresses coiling, but with faster kinetics. We identify the ciliary targeting domains of VALopA. Retargeting of both opsins shows that the locomotory response is prolonged and amplified when signaling occurs in the cilium. We propose that ciliary localization provides a mechanism for enhancing GPCR signaling in central neurons.


Subject(s)
Receptors, G-Protein-Coupled , Zebrafish , Animals , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Opsins , Rod Opsins , Neurons , Cilia/physiology
6.
Gut Microbes ; 15(2): 2257269, 2023 12.
Article in English | MEDLINE | ID: mdl-37749885

ABSTRACT

Intestinal epithelial cell (IEC) regulation of barrier function and mucosal homeostasis enables the establishment of a harmonious gut microenvironment. However, host-derived regulatory networks that modulate intestinal antimicrobial defenses have not been fully defined. Herein we generated mice with IEC-specific deletion of Gpr65 (Gpr65ΔIEC) and investigated the role of epithelial GPR65 using DSS- and C. rodentium-induced murine colitis models. RNA sequencing analysis was conducted on colonic IECs from Gpr65fl/fl and Gpr65ΔIEC mice, and colonoids and colonic epithelial cell lines were used to evaluate the pH-sensing effect of GPR65. The expression of GPR65 was determined in IECs from patients with inflammatory bowel disease (IBD) and DSS colitis mice by qRT-PCR, Western blot, and immunohistochemistry, respectively. We observed that the absence of GPR65 in IECs abrogated homeostatic antimicrobial programs, including the production of antimicrobial peptides (AMPs) and defense response-associated proteins. Gpr65ΔIEC mice displayed dysbiosis of the gut microbiota and were prone to DSS- and C. rodentium-induced colitis, as characterized by significantly disrupted epithelial antimicrobial responses, pathogen invasion, and increased inflammatory infiltrates in the inflamed colon. RNA sequencing analysis revealed that deletion of GPR65 in IECs provoked dramatic transcriptome changes with respect to the downregulation of immune and defense responses to bacteria. Forced AMP induction assays conducted in vivo or in ex vivo colonoids revealed that IEC-intrinsic GPR65 signaling drove antimicrobial defense. Mechanistically, GPR65 signaling promoted STAT3 phosphorylation to optimize mucosal defense responses. Epithelial cell line and colonoid assays further confirmed that epithelial GPR65 sensing pH synergized with IL-22 to facilitate antimicrobial responses. Finally, the expression of GPR65 was markedly decreased in the inflamed epithelia of IBD patients and DSS colitis mice. Our findings define an important role of epithelial GPR65 in regulating intestinal homeostasis and mucosal inflammation and point toward a potential therapeutic approach by targeting GPR65 in the treatment of IBD.


Subject(s)
Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Colitis/chemically induced , Colitis/genetics , Hydrogen-Ion Concentration , Inflammation , Inflammatory Bowel Diseases/genetics , Receptors, G-Protein-Coupled/physiology
7.
Aging Cell ; 22(5): e13815, 2023 05.
Article in English | MEDLINE | ID: mdl-36895142

ABSTRACT

Studies in diverse species have associated higher temperatures with shorter lifespan and lower temperatures with longer lifespan. These inverse effects of temperature on longevity are traditionally explained using the rate of living theory, which posits that higher temperatures increase chemical reaction rates, thus speeding up the aging process. Recent studies have identified specific molecules and cells that affect the longevity response to temperature, indicating that this response is regulated, not simply thermodynamic. Here, we demonstrate that in Caenorhabditis elegans, functional loss of NPR-8, a G protein-coupled receptor related to mammalian neuropeptide Y receptors, increases worm lifespan at 25°C but not at 20°C or 15°C, and that the lifespan extension at 25°C is regulated by the NPR-8-expressing AWB and AWC chemosensory neurons as well as AFD thermosensory neurons. Integrative transcriptomic analyses revealed that both warm temperature and old age profoundly alter gene expression and that genes involved in the metabolic and biosynthetic processes increase expression at 25°C relative to 20°C, indicating elevated metabolism at warm temperature. These data demonstrate that the temperature-induced longevity response is neurally regulated and also provide a partial molecular basis for the rate of living theory, suggesting that these two views are not mutually exclusive. Genetic manipulation and functional assays further uncovered that the NPR-8-dependent longevity response to warm temperature is achieved by regulating the expression of a subset of collagen genes. As increased collagen expression is a common feature of many lifespan-extending interventions and enhanced stress resistance, collagen expression could be critical for healthy aging.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Collagen , Gene Expression Regulation , Hot Temperature , Longevity , Receptors, G-Protein-Coupled , Longevity/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Gene Expression Profiling , Collagen/genetics , Sensory Receptor Cells/metabolism , Gene Deletion
8.
Cells ; 12(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36831293

ABSTRACT

G protein-coupled receptors (GPCRs) are transmembrane signal transducers that regulate a plethora of physiological and pathological processes [...].


Subject(s)
Neoplasms , Receptors, G-Protein-Coupled , Humans , Receptors, G-Protein-Coupled/physiology , Stromal Cells
9.
Biochem Soc Trans ; 51(1): 13-20, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36688421

ABSTRACT

G protein-coupled receptor (GPCR) family members can sense an extraordinary variety of biomolecules to activate intracellular signalling cascades that modulate key aspects of cell physiology. Apart from their crucial role in maintaining cell homeostasis, these critical sensory and modulatory properties have made GPCRs the most successful drug target class to date. However, establishing direct links between receptor activation of specific intracellular partners and individual physiological outcomes is still an ongoing challenge. By studying this receptor signalling complexity at increasing resolution through the development of novel biosensors and high-throughput techniques, a growing number of studies are revealing how receptor function can be diversified in a spatial, temporal or cell-specific manner. This mini-review will introduce recent examples of this context-dependent receptor signalling and discuss how it can impact our understanding of receptor function in health and disease, and contribute to the search of more selective, efficacious and safer GPCR drug candidates.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Receptors, G-Protein-Coupled/physiology , Cell Membrane , Drug Delivery Systems , Ligands
10.
J Neurochem ; 164(4): 454-467, 2023 02.
Article in English | MEDLINE | ID: mdl-36409000

ABSTRACT

Bile acids, which are synthesized in liver and colon, facilitate the digestion of dietary lipids. In addition to this metabolic function, they also act as molecular signals with activities in the nervous system. These are mediated primarily by a G-protein-coupled bile acid receptor (known as TGR5). Preceded by a long tradition in Chinese medicine, bile acids are now being investigated as therapeutic options in several neuropathologies. Specifically, one bile acid, tauroursodeoxycholic acid (TUDCA), which passes the blood-brain barrier and shows anti-inflammatory and anti-apoptotic effects, has been tested in animal models of spinal cord injury (SCI). In this review, we discuss the evidence for a therapeutic benefit in these preclinical experiments. At the time of writing, 12 studies with TGR5 agonists have been published that report functional outcomes with rodent models of SCI. Most investigations found cytoprotective effects and benefits regarding the recovery of sensorimotor function in the subacute phase. When TUDCA was applied in a hydrogel into the lesion site, a significant improvement was obtained at 2 weeks after SCI. However, no lasting improvements with TUDCA treatment were found, when animals were assessed in later, chronic stages. A combination of TUDCA with stem cell injection failed to improve the effect of the cellular treatment. We conclude that the evidence does not support the use of TUDCA as a treatment of SCI. Nevertheless, cytoprotective effects suggest that different modes of application or combinatorial therapies might still be explored.


Subject(s)
Spinal Cord Injuries , Taurochenodeoxycholic Acid , Animals , Taurochenodeoxycholic Acid/pharmacology , Taurochenodeoxycholic Acid/therapeutic use , Spinal Cord Injuries/pathology , Models, Animal , Receptors, G-Protein-Coupled/physiology
11.
Crit Rev Food Sci Nutr ; 63(22): 5594-5607, 2023.
Article in English | MEDLINE | ID: mdl-34978220

ABSTRACT

Nutrients sensing is crucial for fundamental metabolism and physiological functions, and it is also an essential component for maintaining body homeostasis. Traditionally, basic taste receptors exist in oral cavity to sense sour, sweet, bitter, umami, salty and et al. Recent studies indicate that gut can sense the composition of nutrients by activating relevant taste receptors, thereby exerting specific direct or indirect effects. Gut taste receptors, also named as intestinal nutrition receptors, including at least bitter, sweet and umami receptors, have been considered to be activated by certain nutrients and participate in important intestinal physiological activities such as eating behavior, intestinal motility, nutrient absorption and metabolism. Additionally, gut taste receptors can regulate appetite and body weight, as well as maintain homeostasis via targeting hormone secretion or regulating the gut microbiota. On the other hand, malfunction of gut taste receptors may lead to digestive disorders, and then result in obesity, type 2 diabetes and gastrointestinal diseases. At present, researchers have confirmed that the brain-gut axis may play indispensable roles in these diseases via the secretion of brain-gut peptides, but the mechanism is still not clear. In this review, we summarize the current observation of knowledge in gut taste systems in order to shed light on revealing their important nutritional functions and promoting clinical implications.


Subject(s)
Diabetes Mellitus, Type 2 , Taste Buds , Humans , Taste/physiology , Diabetes Mellitus, Type 2/metabolism , Nutrients , Diet , Receptors, G-Protein-Coupled/physiology , Taste Perception/physiology
12.
Commun Biol ; 5(1): 1183, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333465

ABSTRACT

The relaxin/insulin-like family peptide receptor 2 (RXFP2) belongs to the family of class A G-protein coupled receptors (GPCRs) and it is the only known target for the insulin-like factor 3 peptide (INSL3). The importance of this ligand-receptor pair in the development of the gubernacular ligament during the transabdominal phase of testicular descent is well established. More recently, RXFP2 has been implicated in maintaining healthy bone formation. In this report, we describe the discovery of a small molecule series of RXFP2 agonists. These compounds are highly potent, efficacious, and selective RXFP2 allosteric agonists that induce gubernacular invagination in mouse embryos, increase mineralization activity in human osteoblasts in vitro, and improve bone trabecular parameters in adult mice. The described RXFP2 agonists are orally bioavailable and display favorable pharmacokinetic properties, which allow for future evaluation of the therapeutic benefits of modulating RXFP2 activation in disease models.


Subject(s)
Relaxin , Male , Adult , Humans , Mice , Animals , Relaxin/pharmacology , Insulin/pharmacology , Receptors, G-Protein-Coupled/physiology , Testis , Gonadal Steroid Hormones , Receptors, Peptide
13.
FASEB J ; 36(10): e22560, 2022 10.
Article in English | MEDLINE | ID: mdl-36165236

ABSTRACT

Angiogenesis inhibitor drugs targeting vascular endothelial growth factor (VEGF) signaling to the endothelial cell (EC) are used to treat various cancer types. However, primary or secondary resistance to therapy is common. Clinical and pre-clinical studies suggest that alternative pro-angiogenic factors are upregulated after VEGF pathway inhibition. Therefore, identification of alternative pro-angiogenic pathway(s) is critical for the development of more effective anti-angiogenic therapy. Here we study the role of apelin as a pro-angiogenic G-protein-coupled receptor ligand in tumor growth and angiogenesis. We found that loss of apelin in mice delayed the primary tumor growth of Lewis lung carcinoma 1 and B16F10 melanoma when combined with the VEGF receptor tyrosine kinase inhibitor, sunitinib. Targeting apelin in combination with sunitinib markedly reduced the tumor vessel density, and decreased microvessel remodeling. Apelin loss reduced angiogenic sprouting and tip cell marker gene expression in comparison to the sunitinib-alone-treated mice. Single-cell RNA sequencing of tumor EC demonstrated that the loss of apelin prevented EC tip cell differentiation. Thus, apelin is a potent pro-angiogenic cue that supports initiation of tumor neovascularization. Together, our data suggest that targeting apelin may be useful as adjuvant therapy in combination with VEGF signaling inhibition to inhibit the growth of advanced tumors.


Subject(s)
Neoplasms, Experimental , Neoplasms , Angiogenesis Inhibitors/pharmacology , Animals , Apelin , Ligands , Mice , Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptors, G-Protein-Coupled/physiology , Receptors, Vascular Endothelial Growth Factor , Sunitinib/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factors/therapeutic use
14.
Front Immunol ; 13: 907733, 2022.
Article in English | MEDLINE | ID: mdl-35874704

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to joint damage and even disability. Although there are various clinical therapies for RA, some patients still have poor or no response. Thus, the development of new drug targets remains a high priority. In this review, we discuss the role of G-protein-coupled receptors (GPCRs), including chemokine receptors, melanocortin receptors, lipid metabolism-related receptors, adenosine receptors, and other inflammation-related receptors, on mechanisms of RA, such as inflammation, lipid metabolism, angiogenesis, and bone destruction. Additionally, we summarize the latest clinical trials on GPCR targeting to provide a theoretical basis and guidance for the development of innovative GPCR-based clinical drugs for RA.


Subject(s)
Arthritis, Rheumatoid , Arthritis, Rheumatoid/drug therapy , Humans , Inflammation , Receptors, Chemokine , Receptors, G-Protein-Coupled/physiology , Receptors, Purinergic P1
15.
Biochem Pharmacol ; 202: 115115, 2022 08.
Article in English | MEDLINE | ID: mdl-35671790

ABSTRACT

Type 2 diabetes and obesity have reached pandemic proportions throughout the world, so much so that the World Health Organisation coined the term "Globesity" to help encapsulate the magnitude of the problem. G protein-coupled receptors (GPCRs) are highly tractable drug targets due to their wide involvement in all aspects of physiology and pathophysiology, indeed, GPCRs are the targets of approximately 30% of the currently approved drugs. GPCRs are also broadly involved in key physiologies that underlie type 2 diabetes and obesity including feeding reward, appetite and satiety, regulation of blood glucose levels, energy homeostasis and adipose function. Despite this, only two GPCRs are the target of approved pharmaceuticals for treatment of type 2 diabetes and obesity. In this review we discuss the role of these, and select other candidate GPCRs, involved in various facets of type 2 diabetic or obese pathophysiology, how they might be targeted and the potential reasons why pharmaceuticals against these targets have not progressed to clinical use. Finally, we provide a perspective on the current development pipeline of anti-obesity drugs that target GPCRs.


Subject(s)
Diabetes Mellitus, Type 2 , Appetite , Diabetes Mellitus, Type 2/drug therapy , Humans , Obesity/drug therapy , Receptors, G-Protein-Coupled/physiology
16.
J Pharmacol Sci ; 148(3): 307-314, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35177210

ABSTRACT

G protein-coupled receptor 3 (GPR3) constitutively activates Gαs proteins without any ligands and is predominantly expressed in neurons. Since the expression and physiological role of GPR3 in immune cells is still unknown, we examined the possible role of GPR3 in T lymphocytes. The expression of GPR3 was upregulated 2 h after phorbol 12-myristate 13-acetate (PMA)/ionomycin stimulation and was sustained in Jurkat cells, a human T lymphocyte cell line. In addition, the expression of nuclear receptor 4 group A member 2 (NR4A2) was highly modulated by GPR3 expression. Additionally, GPR3 expression was linked with the transcriptional promoter activity of NR4A in Jurkat cells. In mouse CD4+ T cells, transient GPR3 expression was induced immediately after the antigen receptor stimulation. However, the expression of NR4A2 was not modulated in CD4+ T cells from GPR3-knockout mice after stimulation, and the population of Treg cells in thymocytes and splenocytes was not affected by GPR3 knockout. By contrast, spontaneous effector activation in both CD4+ T cells and CD8+ T cells was observed in GPR3-knockout mice. In summary, GPR3 is immediately induced by T cell stimulation and play an important role in the suppression of effector T cell activation.


Subject(s)
Lymphocyte Activation/genetics , Receptors, G-Protein-Coupled/physiology , T-Lymphocytes/immunology , Animals , Cell Line , Chromogranins/metabolism , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , T-Lymphocytes/metabolism
17.
PLoS One ; 17(1): e0262389, 2022.
Article in English | MEDLINE | ID: mdl-35061800

ABSTRACT

G protein-coupled estrogen receptor (GPER) was reported to be a potential target in the breast cancer therapy. This study aimed to illuminate the function of GPER and its mediated PI3K/AKT pathway in cryptotanshinone (CPT) inducing cell apoptosis and antiproliferation effect on GPER positive breast cancer MCF-7 cells. Cell proliferation was tested by MTT assay. Apoptosis rates were tested by Annexin V-FITC/PI double staining and the cell cycle was researched by flow cytometry. Autodock vina was applied to make molecular docking between CPT or estradiol and GPER. siRNA technique and GPER specific agonist G-1 or antagonist G-15 were applied to verify the mediated function of GPER. Apoptosis and cell cycle related proteins, as well as the key proteins on PI3K/AKT signaling pathway were detected by western blot. The results indicated that CPT could exert antiproliferation effects by arresting cell cycle in G2/M phase and downregulating the expression of cyclin D, cyclin B and cyclin A. Besides, apoptosis induced by CPT was observed. CPT might be a novel GPER binding compounds. Significantly, suppression of PI3K/AKT signal transduction by CPT was further increased by G-1 and decreased by G-15. The study revealed that the effect of antiproliferation and apoptosis treating with CPT on MCF-7 cells might be through the downregulation of PI3K/AKT pathway mediated by activated GPER.


Subject(s)
Breast Neoplasms/drug therapy , Phenanthrenes/pharmacology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , China , Estrogens/pharmacology , Female , Humans , MCF-7 Cells , Phenanthrenes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/drug effects
18.
J Pharmacol Sci ; 148(2): 214-220, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35063136

ABSTRACT

Pulmonary hypertension (PH) is a severe and progressive disease that causes elevated right ventricular systolic pressure, right ventricular hypertrophy and ultimately right heart failure. However, the underlying pathophysiologic mechanisms are poorly understood. We previously showed that 3,4-l-dihydroxylphenyalanine (DOPA) sensitizes vasomotor response to sympathetic tone via coupling between the adrenergic receptor alpha1 (ADRA1) and a G protein-coupled receptor 143 (GPR143), a DOPA receptor. We investigated whether DOPA similarly enhances ADRA1-mediated contraction in pulmonary arteries isolated from rats, and whether GPR143 is involved in the PH pathogenesis. Pretreating the isolated pulmonary arteries with DOPA 1 µM enhanced vasoconstriction in response to phenylephrine, an ADRA1 agonist, but not to U-46619, a thromboxane A2 agonist or endothelin-1. We generated Gpr143 gene-deficient (Gpr143-/y) rats, and confirmed that DOPA did not augment phenylephrine-induced contractile response in Gpr143-/y rat pulmonary arteries. We utilized a rat model of monocrotaline (MCT)-induced PH. In the MCT model, the right ventricular systolic pressure was attenuated in the Gpr143-/y rats than in WT rats. Phenylephrine-induced cell migration and proliferation were also suppressed in Gpr143-/y pulmonary artery smooth muscle cells than in WT cells. Our result suggests that GPR143 is involved in the PH pathogenesis in the rat models of PH.


Subject(s)
Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Monocrotaline/adverse effects , Receptors, G-Protein-Coupled/physiology , Receptors, Neurotransmitter/genetics , Systole , Ventricular Function, Right/genetics , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Disease Models, Animal , Heart Failure/etiology , Hypertrophy, Right Ventricular/etiology , In Vitro Techniques , Male , Pulmonary Artery/physiology , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/physiology , Vasoconstriction/drug effects , Vasoconstriction/genetics , Ventricular Dysfunction, Right/etiology
19.
Life Sci ; 291: 120307, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35016881

ABSTRACT

AIM: Glioblastoma multiforme (GBM) is the most common and aggressive primary adult brain tumor. GBM is characterized by a heterogeneous population of cells that are resistant to chemotherapy. Recently, we have synthesized CHBC, a novel indole derivative targeted to GBM biomarker G-protein-coupled receptor 17 and inhibitor of GBM cells. In this study, CHBC was further investigated to characterize the efficiency of this agonist at the molecular level and its underlying mechanism in GBM cell death induction. MATERIALS AND METHODS: The effect of CHBC and TMZ was determined using time dependent inhibitor assay in glioblastoma cells, LN229 and SNB19. Drug induced cell cycle arrest was measured using PI staining followed by image analysis. The induction of apoptosis and mechanism of action of CHBC was studied using apoptosis, caspase 3/7 and mitochondrial membrane permeability assays. Modulation of the key genes involved in MAPK signaling pathway was also measured using immunoblotting array. KEY FINDINGS: The inhibitory kinetic study has revealed that CHBC inhibited SNB19 and LN229 cell growth in a time-dependent manner. Furthermore, CHBC with the IC50 of 85 µM, mediated cell death through an apoptosis mechanism in both studied cell lines. The study also has revealed that CHBC targets GPR17 leading to the induction of apoptosis via the activation of Caspase 3/7 and dysfunction of mitochondrial membrane potential. In addition, CHBC treatment led to marked G2/M cell cycle arrest. The protein array has confirmed the anticancer effect of CHBC by the disruption of the mitogen-activated protein kinase pathway (MAPK). SIGNIFICANCE: Taken together, these results demonstrated that CHBC induced G2/M cell cycle arrest and apoptosis by disrupting MAPK signaling in human glioblastoma cells. This study concludes that CHBC represent a class of compounds for treating glioblastoma.


Subject(s)
Glioblastoma , Indoles , Receptors, G-Protein-Coupled , Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Indoles/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/drug effects , Temozolomide/pharmacology
20.
Crit Rev Food Sci Nutr ; 62(25): 7015-7024, 2022.
Article in English | MEDLINE | ID: mdl-33998842

ABSTRACT

Due to unique characteristics, umami substances have gained much attention in the food industry during the past decade as potential replacers to sodium or fat to increase food palatability. Umami is not only known to increase appetite, but also to increase satiety, and hence could be used to control food intake. Therefore, it is important to understand the mechanism(s) involved in umami taste perception. This review discusses current knowledge of the mechanism(s) of umami perception from receptor level to human brain imaging. New findings regarding the molecular mechanisms for detecting umami tastes and their pathway(s), and the peripheral and central coding to umami taste are reviewed. The representation of umami in the human brain and the individual variation in detecting umami taste and associations with genotype are discussed. The presence of umami taste receptors in the gastrointestinal tract, and the interactions between the brain and gut are highlighted. The review concludes that more research is required into umami taste perception to include not only oral umami taste perception, but also the wider "whole body" signaling mechanisms, to explore the interaction between the brain and gut in response to umami perception and ingestion.


Subject(s)
Taste Perception , Taste , Brain/diagnostic imaging , Brain/metabolism , Humans , Neuroimaging , Receptors, G-Protein-Coupled/physiology , Taste/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...