Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Cancer Lett ; 571: 216333, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37543278

ABSTRACT

The mechanisms underlying the progression of prostate cancer (PCa) to neuroendocrine prostate cancer (NEPC), an aggressive PCa variant, are largely unclear. Two prominent NEPC phenotypes are elevated NE marker expression and heightened angiogenesis. Identifying the still elusive direct molecular links connecting angiogenesis and neuroendocrine differentiation (NED) is crucial for our understanding and targeting of NEPC. Here we found that histone deacetylase 2 (HDAC2), whose role in NEPC has not been reported, is one of the most upregulated epigenetic regulators in NEPC. HDAC2 promotes both NED and angiogenesis. G protein-coupled receptor kinase 3 (GRK3), also upregulated in NEPC, is a critical promoter for both phenotypes too. Of note, GRK3 phosphorylates HDAC2 at S394, which enhances HDAC2's epigenetic repression of potent anti-angiogenic factor Thrombospondin 1 (TSP1) and master NE-repressor RE1 Silencing Transcription Factor (REST). Intriguingly, REST suppresses angiogenesis while TSP1 suppresses NE marker expression in PCa cells, indicative of their novel functions and their synergy in cross-repressing the two phenotypes. Furthermore, the GRK3-HDAC2 pathway is activated by androgen deprivation therapy and hypoxia, both known to promote NED and angiogenesis in PCa. These results indicate that NED and angiogenesis converge on GRK3-enhanced HDAC2 suppression of REST and TSP1, which constitutes a key missing link between two prominent phenotypes of NEPC.


Subject(s)
G-Protein-Coupled Receptor Kinase 3 , Histone Deacetylase 2 , Prostatic Neoplasms , Humans , Male , Androgen Antagonists , Cell Differentiation , Cell Line, Tumor , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Signal Transduction/genetics , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism
2.
Cells ; 12(7)2023 03 23.
Article in English | MEDLINE | ID: mdl-37048054

ABSTRACT

Paget's Disease of Bone (PDB) is a metabolic bone disease that is characterized by dysregulated osteoclast function leading to focal abnormalities of bone remodeling. It can lead to pain, fracture, and bone deformity. G protein-coupled receptor kinase 3 (GRK3) is an important negative regulator of G protein-coupled receptor (GPCR) signaling. GRK3 is known to regulate GPCR function in osteoblasts and preosteoblasts, but its regulatory function in osteoclasts is not well defined. Here, we report that Grk3 expression increases during osteoclast differentiation in both human and mouse primary cells and established cell lines. We also show that aged mice deficient in Grk3 develop bone lesions similar to those seen in human PDB and other Paget's Disease mouse models. We show that a deficiency in Grk3 expression enhances osteoclastogenesis in vitro and proliferation of hematopoietic osteoclast precursors in vivo but does not affect the osteoclast-mediated bone resorption function or cellular senescence pathway. Notably, we also observe decreased Grk3 expression in peripheral blood mononuclear cells of patients with PDB compared with age- and gender-matched healthy controls. Our data suggest that GRK3 has relevance to the regulation of osteoclast differentiation and that it may have relevance to the pathogenesis of PDB and other metabolic bone diseases associated with osteoclast activation.


Subject(s)
Bone Diseases, Metabolic , Bone Resorption , G-Protein-Coupled Receptor Kinase 3 , Osteitis Deformans , Animals , Humans , Mice , Bone Diseases, Metabolic/pathology , Bone Resorption/metabolism , Leukocytes, Mononuclear/metabolism , Osteitis Deformans/genetics , Osteitis Deformans/metabolism , Osteoclasts/metabolism , Osteogenesis , G-Protein-Coupled Receptor Kinase 3/genetics
3.
Neurosci Lett ; 760: 136092, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34197905

ABSTRACT

Though it is well known that G protein-coupled receptor kinase 2 [GRK2] is involved in regulation of mu opioid receptor [MOR] desensitization and morphine-related behaviors, the potential role of GRK2 in regulation of kappa opioid receptor [KOR] functions in vivo has not been established yet. A couple of recent studies have found that GRK2 activity desensitizes KOR functions via decreasing G protein-coupled signaling with sensitizing arrestin-coupled signaling. Nalfurafine, a G protein-biased KOR full agonist, produces an inhibitory effect on alcohol intake in mice, with fewer side effects (sedation, aversion, or anxiety/depression-like behaviors). Using RNA sequencing (RNA-seq) analysis, we first identified that nuclear transcript level of grk2 [adrbk1] (but not other grks) was significantly up-regulated in mouse nucleus accumbens shell (NAcs) after chronic excessive alcohol drinking, suggesting alcohol specifically increased NAcs grk2 expression. We then tested whether selective GRK2/3 inhibitor CMPD101 could alter alcohol intake and found that CMPD101 alone had no effect on alcohol drinking. Therefore, we hypothesized that the grk2 increase in the NAcs could modulate the nalfurafine effect on alcohol intake via interacting with the G protein-mediated KOR signaling. Nalfurafine decreased alcohol drinking in a dose-related manner, and pretreatment with CMPD101 enhanced the reduction in alcohol intake induced by nalfurafine, indicating an involvement of GRK2/3 blockade in modulating G protein-biased KOR agonism of nalfurafine. Together, our study provides initial evidence relevant to the transcriptional change of grk2 gene in the NAc shell after excessive alcohol drinking. Pharmacological GRK2/3 blockade enhanced nalfurafine's efficacy, suggesting a GRK2/3-mediated mechanism, probably through the G protein-mediated KOR signaling.


Subject(s)
Alcoholism/drug therapy , G-Protein-Coupled Receptor Kinase 2/metabolism , Morphinans/pharmacology , Nucleus Accumbens/drug effects , Receptors, Opioid, kappa/agonists , Spiro Compounds/pharmacology , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Ethanol/administration & dosage , Ethanol/adverse effects , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 3/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 3/metabolism , Humans , Male , Mice , Morphinans/therapeutic use , Nucleus Accumbens/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, Opioid, kappa/metabolism , Spiro Compounds/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use
4.
Cells ; 10(3)2021 03 11.
Article in English | MEDLINE | ID: mdl-33799570

ABSTRACT

Background: The atypical chemokine receptor 3 (ACKR3) belongs to the superfamily of G protein-coupled receptors (GPCRs). Unlike classical GPCRs, this receptor does not activate G proteins in most cell types but recruits ß-arrestins upon activation. ACKR3 plays an important role in cancer and vascular diseases. As recruitment of ß-arrestins is triggered by phosphorylation of the C-terminal tail of GPCRs, we studied the role of different potential phosphorylation sites within the ACKR3 C-tail to further delineate the molecular mechanism of internalization and trafficking of this GPCR. Methods: We used various bioluminescence and fluorescence resonance energy transfer-based sensors and techniques in Human Embryonic Kidney (HEK) 293T cells expressing WT or phosphorylation site mutants of ACKR3 to measure CXCL12-induced recruitment of ß-arrestins and G-protein-coupled receptor kinases (GRKs), receptor internalization and trafficking. Results: Upon CXCL12 stimulation, ACKR3 recruits both ß-arrestin 1 and 2 with equivalent kinetic profiles. We identified interactions with GRK2, 3 and 5, with GRK2 and 3 being important for ß-arrestin recruitment. Upon activation, ACKR3 internalizes and recycles back to the cell membrane. We demonstrate that ß-arrestin recruitment to the receptor is mainly determined by a single cluster of phosphorylated residues on the C-tail of ACKR3, and that residue T352 and in part S355 are important residues for ß-arrestin1 recruitment. Phosphorylation of the C-tail appears essential for ligand-induced internalization and important for differential ß-arrestin recruitment. GRK2 and 3 play a key role in receptor internalization. Moreover, ACKR3 can still internalize when ß-arrestin recruitment is impaired or in the absence of ß-arrestins, using alternative internalization pathways. Our data indicate that distinct residues within the C-tail of ACKR3 differentially regulate CXCL12-induced ß-arrestin recruitment, ACKR3 trafficking and internalization.


Subject(s)
Endocytosis , Receptors, CXCR/metabolism , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , Biosensing Techniques , Chemokine CXCL12/pharmacology , Fluorescence Resonance Energy Transfer , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , HEK293 Cells , Humans , Kinetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Receptors, CXCR/agonists , Receptors, CXCR/genetics , beta-Arrestin 1/genetics , beta-Arrestin 2/genetics
5.
Mol Cell Biochem ; 476(3): 1505-1516, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33392923

ABSTRACT

G protein-coupled receptor kinases (GRKs), in addition to their role in modulating signal transduction mechanisms associated with activated G protein-coupled receptors (GPCRs), can also interact with many non-GPCR proteins to mediate cellular responses to chemotherapeutics. The rationale for this study is based on the presumption that GRK2 modulates the responses of cancer cells to the chemotherapeutic cisplatin. In this report, we show that GRK2 modulates the responses of cancer cells to cisplatin. Cervical cancer HeLa cells stably transfected with GRK2 shRNA, to decrease GRK2 protein expression, show increased sensitivity to cisplatin. Of interest, these cells also show increased accumulation of NADPH, associating with decreased NADP buildup, at low concentrations of cisplatin tested. These changes in NADPH and NADP levels are also observed in the breast cancer MDA MB 231 cells, which has lower endogenous GRK2 protein expression levels, but not BT549, a breast cancer cell line with higher GRK2 protein expression. This effect of NADPH accumulation may be associated with a decrease in NADPH oxidase 4 (NOX4) protein expression, which is found to correlate with GRK2 protein expression in cancer cells-a relationship which mimics that observed in cardiomyocytes. Furthermore, like in cardiomyocytes, GRK2 and NOX4 interact to form complexes in cancer cells. Collectively, these results suggest that GRK2 interacts with NOX4 to modify cisplatin sensitivity in cancer cells and may also factor into the success of cisplatin-based regimens.


Subject(s)
Cisplatin/pharmacology , G-Protein-Coupled Receptor Kinase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Profiling , HeLa Cells , Humans , Neoplasms/metabolism , Phosphorylation , Protein Binding , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors
6.
Sci Rep ; 10(1): 17395, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060647

ABSTRACT

Most G protein-coupled receptors (GPCRs) recruit ß-arrestins and internalize upon agonist stimulation. For the µ-opioid receptor (µ-OR), this process has been linked to development of opioid tolerance. GPCR kinases (GRKs), particularly GRK2 and GRK3, have been shown to be important for µ-OR recruitment of ß-arrestin and internalization. However, the contribution of GRK2 and GRK3 to ß-arrestin recruitment and receptor internalization, remain to be determined in their complete absence. Using CRISPR/Cas9-mediated genome editing we established HEK293 cells with knockout of GRK2, GRK3 or both to dissect their individual contributions in ß-arrestin2 recruitment and µ-OR internalization upon stimulation with four different agonists. We showed that GRK2/3 removal reduced agonist-induced µ-OR internalization and ß-arrestin2 recruitment substantially and we found GRK2 to be more important for these processes than GRK3. Furthermore, we observed a sustained and GRK2/3 independent component of ß-arrestin2 recruitment to the plasma membrane upon µ-OR activation. Rescue expression experiments restored GRK2/3 functions. Inhibition of GRK2/3 using the small molecule inhibitor CMPD101 showed a high similarity between the genetic and pharmacological approaches, cross-validating the specificity of both. However, off-target effects were observed at high CMPD101 concentrations. These GRK2/3 KO cell lines should prove useful for a wide range of studies on GPCR function.


Subject(s)
CRISPR-Cas Systems , Endocytosis/physiology , G-Protein-Coupled Receptor Kinase 2/physiology , G-Protein-Coupled Receptor Kinase 3/physiology , Receptors, Opioid, mu/metabolism , beta-Arrestin 2/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/genetics , Gene Editing , Gene Knockdown Techniques , HEK293 Cells , Humans , Phosphorylation , Reproducibility of Results
7.
Biochem Biophys Res Commun ; 528(3): 432-439, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32505358

ABSTRACT

GRK-mediated receptor phosphorylation followed by association with ß-arrestins has been proposed to be the molecular mechanism involved in the desensitization of G protein-coupled receptors (GPCRs). However, this mechanism does not explain the desensitization of some GPCRs, such as dopamine D3 receptor (D3R), which does not undergo GRK-mediated phosphorylation. Loss-of-function approaches and mutants of dopamine D2 receptor and D3R, which exhibit different desensitization properties, were used to identify the cellular components and processes responsible for desensitization. D3R mediated the recruitment of Mdm2 to the cytosol, which resulted in the constitutive ubiquitination of ß-arrestin2 in the resting state. Under desensitization conditions, cytosolic Mdm2 returned to the nucleus, resulting in the deubiquitination of cytosolic ß-arrestins. Deubiquitinated ß-arrestins formed a tight complex with Gßγ, thereby sequestering it, causing interference in D3R signaling. In conclusion, this study shows that ß-arrestins, depending on their ubiquitination status, control the G protein cycling by regulating their interactions with Gßγ. This is a novel mechanism proposed to explain how certain GPCRs can undergo desensitization without receptor phosphorylation.


Subject(s)
Receptors, Dopamine D3/metabolism , Receptors, G-Protein-Coupled/metabolism , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Gene Knockdown Techniques , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Mutation , Phosphorylation , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Ubiquitination , beta-Arrestins/metabolism
8.
Front Immunol ; 11: 720, 2020.
Article in English | MEDLINE | ID: mdl-32391018

ABSTRACT

Chemokines are essential for guiding cell migration. Atypical chemokine receptors (ACKRs) contribute to the cell migration process by binding, internalizing and degrading local chemokines, which enables the formation of confined gradients. ACKRs are heptahelical membrane spanning molecules structurally related to G-protein coupled receptors (GPCRs), but seem to be unable to signal through G-proteins upon ligand binding. ACKR4 internalizes the chemokines CCL19, CCL21, and CCL25 and is best known for shaping functional CCL21 gradients. Ligand binding to ACKR4 has been shown to recruit ß-arrestins that has led to the assumption that chemokine scavenging relies on ß-arrestin-mediated ACKR4 trafficking, a common internalization route taken by class A GPCRs. Here, we show that CCL19, CCL21, and CCL25 readily recruited ß-arrestin1 and ß-arrestin2 to human ACKR4, but found no evidence for ß-arrestin-dependent or independent ACKR4-mediated activation of the kinases Erk1/2, Akt, or Src. However, we demonstrate that ß-arrestins interacted with ACKR4 in the steady-state and contributed to the spontaneous trafficking of the receptor in the absence of chemokines. Deleting the C-terminus of ACKR4 not only interfered with the interaction of ß-arrestins, but also with the uptake of fluorescently labeled cognate chemokines. We identify the GPCR kinase GRK3, and to a lesser extent GRK2, but not GRK4, GRK5, and GRK6, to be recruited to chemokine-stimulated ACKR4. We show that GRK3 recruitment proceded the recruitment of ß-arrestins upon ACKR4 engagement and that GRK2/3 inhibition partially interfered with steady-state interaction and chemokine-driven recruitment of ß-arrestins to ACKR4. Overexpressing ß-arrestin2 accelerated the uptake of fluorescently labeled CCL19, indicating that ß-arrestins contribute to the chemokine scavenging activity of ACKR4. By contrast, cells lacking ß-arrestins were still capable to take up fluorescently labeled CCL19 demonstrating that ß-arrestins are dispensable for chemokine scavenging by ACKR4.


Subject(s)
Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Chemokines, CC/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Receptors, CCR/metabolism , Signal Transduction/genetics , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , HeLa Cells , Humans , Plasmids/genetics , Plasmids/metabolism , Protein Binding/genetics , Receptors, CCR/genetics , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Transfection , beta-Arrestin 2/genetics
9.
Med Sci Monit ; 25: 7407-7417, 2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31609302

ABSTRACT

BACKGROUND The initiation of atherosclerosis (AS) is attributed to the dysfunction of endothelial cells (ECs) via the inhibition of g protein-coupled estrogen receptor (GPER). In the current study, we assessed the potential of Ginsenoside Rb1 (Rb1) to attenuate the dysfunction of ECs via GPER-mediated PI3K/Akt pathway. MATERIAL AND METHODS AS was induced in rabbits and then the AS rabbits were treated with Rb1. Thereafter, the ECs were isolated from AS and healthy rabbits, and treated with Rb1. The effect of Rb1 on blood lipid levels in AS rabbits and on apoptosis, inflammatory response, and GPER/PI3K/Akt axis activity in ECs was detected. Furthermore, the activities of GPER and PI3K were modulated to verify the key role of the axis in the anti-AS effect of Rb1. RESULTS The levels of total cholesterol, low-density lipoprotein (LDL), and triglyceride in AS rabbits were suppressed by Rb1 while the high-density lipoprotein (HDL) level was increased. In in vitro assays, Rb1 administration inhibited apoptosis process and the production of pro-inflammation cytokines in AS ECs. The expression levels of GPER, p-PI3K, and p-Akt were upregulated by Rb1, associated with the increased level of Bcl-2 and reduced level of Bax. When the activity of GPER was inhibited by GP-15 in AS ECs, the treatment effect of Rb1 was blocked. However, the activation of PI3K could restore the protective effect of Rb1 after the inhibition of GPER. CONCLUSIONS The anti-AS potential of Rb1 was exerted by restoring the regular function of ECs via the activation of GPER-mediated PI3K/Akt signaling.


Subject(s)
Atherosclerosis/physiopathology , Endothelial Cells/drug effects , Ginsenosides/pharmacology , Animals , Apoptosis/drug effects , Atherosclerosis/drug therapy , China , Diet, High-Fat , Disease Models, Animal , Endothelial Cells/metabolism , G-Protein-Coupled Receptor Kinase 3 , Ginsenosides/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols , Proto-Oncogene Proteins c-akt/metabolism , Rabbits , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
10.
Biochem Biophys Res Commun ; 520(2): 327-332, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31604529

ABSTRACT

The Raf kinase inhibitor protein (RKIP) activates ß-adrenoceptors (ß-AR) and thereby induces a well-tolerated cardiac contractility and prevents heart failure in mice. Different to RKIP-mediated ß-AR activation, chronic activation of ß-AR by catecholamines was shown to be detrimental for the heart. RKIP is an endogenous inhibitor of G protein coupled receptor kinase 2 (GRK2); it binds GRK2 and thereby inhibits GRK2 mediated ß-AR phosphorylation and desensitization. Here, we evaluate RKIP-mediated effects on ß-AR to explore new strategies for ß-AR modulation. Co-immunoprecipitation assays and pull-down assays revealed subtype specificity of RKIP for the cardiac GRK isoforms GRK2 and GRK3 - not GRK5 - as well as several RKIP binding sites within their N-termini (GRK21-185 and GRK31-185). Overexpression of these N-termini prevented ß2-AR phosphorylation and internalization, subsequently increased receptor signaling in HEK293 cells and cardiomyocyte contractility. Co-immunoprecipitation assays of ß2-AR with these N-terminal GRK fragments revealed a direct interaction suggesting a steric interference of the fragments with the functional GRK-receptor interaction. Altogether, N-termini of GRK2 and GRK3 efficiently simulate RKIP effects on ß-AR signaling in HEK293 cells and in cardiomyocytes by their binding to ß2-AR and, thus, provide important insights for the development of new strategies to modulate ß2-AR signaling.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Receptors, Adrenergic, beta-2/metabolism , Animals , Binding Sites , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , HEK293 Cells , Humans , Mice, Inbred Strains , Myocytes, Cardiac , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Phosphorylation , Receptors, Adrenergic, beta-2/genetics
11.
Mol Cell Biochem ; 461(1-2): 103-118, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31363957

ABSTRACT

G protein-coupled receptor kinases (GRKs) phosphorylate the activated forms of G protein-coupled receptors (GPCRs), leading to receptor desensitization and internalization. In addition, GRKs can modify the activity of many non-GPCR-signaling pathways as well, controlling other cellular functions beyond that directly associated with a GPCR. In this report, we show that cervical cancer HeLa cells and breast cancer MDA MB 231 cells with reduced GRK5 expression display increased sensitivity to the apoptotic effects of paclitaxel (Taxol). This effect in cancer cells with low GRK5 levels could be because of blunted histone deacetylase 6 (HDAC6) activity that leads to an increase in α-tubulin acetylation levels, which augments paclitaxel sensitivity. We demonstrate that GRK5 and HDAC6 form a signaling complex in cells and in vitro. GRK5 phosphorylates HDAC6 at Ser-21 to promote its deacetylase activity. Therefore, the GRK5-HDAC6 interaction may contribute to paclitaxel resistance in cancer cells.


Subject(s)
Drug Resistance, Neoplasm/drug effects , G-Protein-Coupled Receptor Kinase 5/metabolism , Paclitaxel/pharmacology , Acetylation , Apoptosis/drug effects , Biocatalysis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Docetaxel/pharmacology , Female , G-Protein-Coupled Receptor Kinase 3/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , HeLa Cells , Histone Deacetylase 6/metabolism , Histones/metabolism , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Tubulin/metabolism
12.
Int J Mol Sci ; 20(15)2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31366084

ABSTRACT

Duration of receptor antagonism, measured as the recovery of agonist responsiveness, is gaining attention as a method to evaluate the 'effective' target-residence for antagonists. These functional assays might be a good alternative for kinetic binding assays in competition with radiolabeled or fluorescent ligands, as they are performed on intact cells and better reflect consequences of dynamic cellular processes on duration of receptor antagonism. Here, we used a bioluminescence resonance energy transfer (BRET)-based assay that monitors heterotrimeric G protein activation via scavenging of released Venus-Gß1γ2 by NanoLuc (Nluc)-tagged membrane-associated-C-terminal fragment of G protein-coupled receptor kinase 3 (masGRK3ct-Nluc) as a tool to probe duration of G protein-coupled receptor (GPCR) antagonism. The Gαi-coupled histamine H3 receptor (H3R) was used in this study as prolonged antagonism is associated with adverse events (e.g., insomnia) and consequently, short-residence time ligands might be preferred. Due to its fast and prolonged response, this assay can be used to determine the duration of functional antagonism by measuring the recovery of agonist responsiveness upon washout of pre-bound antagonist, and to assess antagonist re-equilibration time via Schild-plot analysis. Re-equilibration of pre-incubated antagonist with agonist and receptor could be followed in time to monitor the transition from insurmountable to surmountable antagonism. The BRET-based G protein activation assay can detect differences in the recovery of H3R responsiveness and re-equilibration of pre-bound antagonists between the tested H3R antagonists. Fast dissociation kinetics were observed for marketed drug pitolisant (Wakix®) in this assay, which suggests that short residence time might be beneficial for therapeutic targeting of the H3R.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Histamine H3 Antagonists/pharmacology , Receptors, Histamine H3/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Protein Binding
13.
Neurobiol Aging ; 81: 177-189, 2019 09.
Article in English | MEDLINE | ID: mdl-31306812

ABSTRACT

Parkinson's disease (PD) is often managed with L-3,4-dihydroxyphenylalanine (L-DOPA), which is still the gold standard to relieve the clinical motor symptoms of PD. However, chronic use of L-DOPA leads to significant motor complications, especially L-DOPA-induced dyskinesia (LID), which limit the therapeutic benefit. Few options are available for the pharmacological management of LID partly due to the inadequacy of our mechanistic understanding of the syndrome. We focused on the role of the histamine (HA) H2 receptor (H2R) in the striatum, which others have shown to be involved in the development of LID. We generated LID in a hemiparkinsonian mouse model and tested the signaling effects of ranitidine, an H2R antagonist. We used histidine decarboxylase deficient mice (Hdc-Ko) which lacks HA to study the role of G-protein-coupled receptor kinases (GRKs) in HA deficiency. Loss of HA in Hdc-Ko mice did not result in the downregulation of GRKs, especially GRK3 and GRK6, which were previously found to be reduced in hemiparkinsonian animal models. Ranitidine, when given along with L-DOPA, normalized the expression of GRK3 in the dopamine-depleted striatum thereby inhibiting LID in mice. The extracellular signal regulated kinase and ΔFosB signaling pathways were attenuated in the lesioned striatum when ranitidine was combined with L-DOPA than L-DOPA alone. These results demonstrate that ranitidine inhibits LID by normalizing the levels of GRK3, extracellular signal regulated kinase activation, and FosB accumulation in the dopamine-depleted striatum via HA H2R antagonism.


Subject(s)
Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/prevention & control , G-Protein-Coupled Receptor Kinase 3/metabolism , Histamine H2 Antagonists/therapeutic use , Levodopa/adverse effects , Parkinson Disease/drug therapy , Proto-Oncogene Proteins c-fos/metabolism , Ranitidine/therapeutic use , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Levodopa/therapeutic use , MAP Kinase Signaling System , Mice, Inbred C57BL , Parkinson Disease/metabolism , Receptors, Histamine H2
14.
Sci Signal ; 12(574)2019 03 26.
Article in English | MEDLINE | ID: mdl-30914485

ABSTRACT

Agonists of the nociceptin/orphanin FQ opioid peptide (NOP) receptor, a member of the opioid receptor family, are under active investigation as novel analgesics, but their modes of signaling are less well characterized than those of other members of the opioid receptor family. Therefore, we investigated whether different NOP receptor ligands showed differential signaling or functional selectivity at the NOP receptor. Using newly developed phosphosite-specific antibodies to the NOP receptor, we found that agonist-induced NOP receptor phosphorylation occurred primarily at four carboxyl-terminal serine (Ser) and threonine (Thr) residues, namely, Ser346, Ser351, Thr362, and Ser363, and proceeded with a temporal hierarchy, with Ser346 as the first site of phosphorylation. G protein-coupled receptor kinases 2 and 3 (GRK2/3) cooperated during agonist-induced phosphorylation, which, in turn, facilitated NOP receptor desensitization and internalization. A comparison of structurally distinct NOP receptor agonists revealed dissociation in functional efficacies between G protein-dependent signaling and receptor phosphorylation. Furthermore, in NOP-eGFP and NOP-eYFP mice, NOP receptor agonists induced multisite phosphorylation and internalization in a dose-dependent and agonist-selective manner that could be blocked by specific antagonists. Our study provides new tools to study ligand-activated NOP receptor signaling in vitro and in vivo. Differential agonist-selective NOP receptor phosphorylation by chemically diverse NOP receptor agonists suggests that differential signaling by NOP receptor agonists may play a role in NOP receptor ligand pharmacology.


Subject(s)
Receptors, Opioid/agonists , Amino Acid Sequence , Animals , Antibody Specificity , Dose-Response Relationship, Drug , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Genes, Reporter , HEK293 Cells , Humans , Ligands , Mice , Models, Molecular , Phosphorylation , Phosphoserine/analysis , Phosphothreonine/analysis , Protein Processing, Post-Translational , Receptors, Opioid/immunology , Receptors, Opioid/metabolism , Recombinant Proteins/metabolism , Structure-Activity Relationship , Nociceptin Receptor
15.
Toxicol Sci ; 166(2): 288-305, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30379318

ABSTRACT

We have shown that acute ozone inhalation activates sympathetic-adrenal-medullary and hypothalamus-pituitary-adrenal stress axes, and adrenalectomy (AD) inhibits ozone-induced lung injury and inflammation. Therefore, we hypothesized that stress hormone receptor agonists (ß2 adrenergic-ß2AR and glucocorticoid-GR) will restore the ozone injury phenotype in AD, while exacerbating effects in sham-surgery (SH) rats. Male Wistar Kyoto rats that underwent SH or AD were treated with vehicles (saline + corn oil) or ß2AR agonist clenbuterol (CLEN, 0.2 mg/kg, i.p.) + GR agonist dexamethasone (DEX, 2 mg/kg, s.c.) for 1 day and immediately prior to each day of exposure to filtered air or ozone (0.8 ppm, 4 h/day for 1 or 2 days). Ozone-induced increases in PenH and peak-expiratory flow were exacerbated in CLEN+DEX-treated SH and AD rats. CLEN+DEX affected breath waveform in all rats. Ozone exposure in vehicle-treated SH rats increased bronchoalveolar lavage fluid (BALF) protein, N-acetyl glucosaminidase activity (macrophage activation), neutrophils, and lung cytokine expression while reducing circulating lymphocyte subpopulations. AD reduced these ozone effects in vehicle-treated rats. At the doses used herein, CLEN+DEX treatment reversed the protection offered by AD and exacerbated most ozone-induced lung effects while diminishing circulating lymphocytes. CLEN+DEX in air-exposed SH rats also induced marked protein leakage and reduced circulating lymphocytes but did not increase BALF neutrophils. In conclusion, circulating stress hormones and their receptors mediate ozone-induced vascular leakage and inflammatory cell trafficking to the lung. Those receiving ß2AR and GR agonists for chronic pulmonary diseases, or with increased circulating stress hormones due to psychosocial stresses, might have altered sensitivity to air pollution.


Subject(s)
Adrenalectomy , Adrenergic beta-2 Receptor Agonists/pharmacology , Lung Injury/drug therapy , Ozone/toxicity , Pneumonia/drug therapy , Receptors, Glucocorticoid/agonists , Animals , Bronchoalveolar Lavage Fluid/chemistry , Clenbuterol/pharmacokinetics , Corticosterone/blood , Cytokines/metabolism , Dexamethasone/pharmacology , Epinephrine/blood , G-Protein-Coupled Receptor Kinase 3/metabolism , Leukocytes/metabolism , Lung Injury/chemically induced , Lung Injury/metabolism , Lymphocytes/metabolism , Male , Pneumonia/chemically induced , Pneumonia/metabolism , Random Allocation , Rats , Rats, Inbred WKY , Respiratory Function Tests
16.
Methods Mol Biol ; 1820: 33-41, 2018.
Article in English | MEDLINE | ID: mdl-29884935

ABSTRACT

Like in other sensory systems, adaptation is an essential process in the olfactory system, required for its proper functioning. However, the precise molecular mechanism underlying the adaptation process has not been fully understood, especially at the receptor level. Here, we describe methods to evaluate the role of GRK3, one of the members of the GRK family responsible for the desensitization of non-olfactory G-protein-coupled receptor (GPCR), in desensitization of olfactory receptor (OR) using a heterologous expression system. As a parameter to characterize the degree of desensitization, we measure (1) the maximal response to an agonist by either cAMP or Ca2+ imaging assay and (2) the kinetic time course for recovery to basal levels by Ca2+ imaging assay. Differences in the degree of desensitization in the presence or absence of GRK3 can be examined by comparing these parameters, leading to evaluation of GRK3.


Subject(s)
Calcium Signaling , Cyclic AMP/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Receptors, Odorant/metabolism , Smell , G-Protein-Coupled Receptor Kinase 3/genetics , HEK293 Cells , Humans , Receptors, Odorant/genetics
17.
Biomed Res Int ; 2018: 1569701, 2018.
Article in English | MEDLINE | ID: mdl-29693002

ABSTRACT

Patients with obstructive sleep apnea syndrome (OSAS) showed higher prevalence in cardiovascular diseases due to aberrant hypoxia and oxidative stress. However, not all OSAS patients end up with cardiovascular disorders, and identification of novel biomarker will be invaluable for differentiating patients at risk. Here we tested the serum matrix metalloproteinase-9 (MMP-9) levels in 47 untreated OSAS patients and found that the MMP-9 level was positively correlated with severity of OSAS, which was consistent with hypoxia degree and duration. Besides, the MMP-9 level was higher in patients complicated with systematic hypertension (P < 0.001). Furthermore, we selected those OSAS patients without any cardiovascular dysfunction (n = 35) and followed up for up to five years. By the end of follow-up, 12 patients had hypertension onset and 3 patients had left ventricular hypertrophy. By analyzing the clinical outcomes with MMP-9 expression, we demonstrated that high serum MMP-9 in OSAS patients was a risk factor for occurrence of cardiovascular diseases. In addition, we cultured the vascular endothelial cells (VEC) from rat aorta in hypoxia condition to investigate whether MMP-9 was elevated due to hypoxia in OSAS patients. Cellular results revealed that the expression, secretion, and activity of MMP-9 were all upregulated by hypoxia and can cleave the beta2-adrenergic receptor (ß2AR) on VEC surface. Our results not only determined MMP-9 as a risk factor for cardiovascular diseases in OSAS patients, but also showed the possible involvement of hypoxia-MMP-9-ß2AR signaling axis.


Subject(s)
Cardiovascular Diseases/metabolism , Hypertension/metabolism , Matrix Metalloproteinase 9/metabolism , Sleep Apnea, Obstructive/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Female , G-Protein-Coupled Receptor Kinase 3/metabolism , Humans , Hypoxia/metabolism , Male , Middle Aged , Rats , Risk Factors
18.
Eur J Pharmacol ; 831: 9-19, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29698717

ABSTRACT

Alpha1-adrenoceptors induce prostate smooth muscle contraction, and hold a prominent role for pathophysiology and therapy of lower urinary tract symptoms in benign prostatic hyperplasia. G protein-coupled receptors are regulated by posttranslational regulation, including phosphorylation by G protein-coupled receptor kinases 2 and 3 (GRK2/3). Although posttranslational adrenoceptor regulation has been recently suggested to occur in the prostate, this is still marginally understood. With the newly developed CMPD101, a small molecule inhibitor with assumed specificity for GRK2/3 is now available. Here, we studied effects of CMPD101 on smooth muscle contraction of human prostate tissue. Electric field stimulation caused frequency-dependent contractions, which were inhibited concentration-dependently by CMPD101 (5 µM, 50 µM). 50 µM of CMPD101 did not affect myosin light chain (MCL) phosphorylation or Rho kinase activity, and did not alter contractions induced by highmolar KCl. Noradrenaline, the α1-adrenoceptor agonist phenylephrine, endothelin-1, and the thromboxane A2 analogue U46619 induced concentration-dependent contractions, which were inhibited by CMPD101 (50 µM). CMPD101 (50 µM) did not change phosphorylation of ß2-adrenoceptors or ß2-adrenergic relaxation of prostate strips. Molecular detection by Western blot and peroxidase staining suggested expression of GRK2 and GRK3 in human prostates. Double labeling in fluorescence staining confirmed that immunoreactivity for GRK2 and GRK3 was located to smooth muscle cells in the prostate stroma. In conclusion, CMPD101 inhibits adrenergic, neurogenic, and non-adrenergic smooth muscle contractions in the human prostate. Underlying mechanisms may be independent from GRK inhibition, and from inhibition of MLC kinase and Rho kinase. This may point to unknown properties of CMPD101.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Benzamides/pharmacology , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 3/antagonists & inhibitors , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Prostate/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptors, Adrenergic, beta-1/drug effects , Triazoles/pharmacology , Adrenergic alpha-1 Receptor Agonists/pharmacology , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Electric Stimulation , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Humans , In Vitro Techniques , Male , Middle Aged , Muscle, Smooth/enzymology , Phosphorylation , Potassium Chloride/pharmacology , Prostate/enzymology , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects
19.
J Biol Chem ; 293(16): 6161-6171, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29487132

ABSTRACT

The dopamine D2 receptor (D2R) is a G protein-coupled receptor (GPCR) that is critical for many central nervous system functions. The D2R carries out these functions by signaling through two transducers: G proteins and ß-arrestins (ßarrs). Selectively engaging either the G protein or ßarr pathway may be a way to improve drugs targeting GPCRs. The current model of GPCR signal transduction posits a chain of events where G protein activation ultimately leads to ßarr recruitment. GPCR kinases (GRKs), which are regulated by G proteins and whose kinase action facilitates ßarr recruitment, bridge these pathways. Therefore, ßarr recruitment appears to be intimately tied to G protein activation via GRKs. Here we sought to understand how GRK2 action at the D2R would be disrupted when G protein activation is eliminated and the effect of this on ßarr recruitment. We used two recently developed biased D2R mutants that can preferentially interact either with G proteins or ßarrs as well as a ßarr-biased D2R ligand, UNC9994. With these functionally selective tools, we investigated the mechanism whereby the ßarr-preferring D2R achieves ßarr pathway activation in the complete absence of G protein activation. We describe how direct, G protein-independent recruitment of GRK2 drives interactions at the ßarr-preferring D2R and also contributes to ßarr recruitment at the WT D2R. Additionally, we found an additive interaction between the ßarr-preferring D2R mutant and UNC9994. These results reveal that the D2R can directly recruit GRK2 without G protein activation and that this mechanism may have relevance to achieving ßarr-biased signaling.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Proteins/metabolism , Receptors, Dopamine D2/metabolism , Dopamine Agonists/pharmacology , Energy Transfer , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/antagonists & inhibitors , HEK293 Cells , Humans , Models, Theoretical , Mutation , Pertussis Toxin/metabolism , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Receptors, Dopamine D2/genetics , Signal Transduction , beta-Arrestins/metabolism
20.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R574-R583, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29212811

ABSTRACT

Increased ß-adrenergic receptor (ß-AR)-mediated activation of adenylyl cyclase (AC) in rat liver during aging has been linked to age-related increases in hepatic glucose output and hepatosteatosis. In this study, we investigated the expression of ß-ARs, individual receptor subtypes, and G protein-coupled receptor (GPCR) regulatory proteins in livers from aging rats. Radioligand-binding studies demonstrated that ß-AR density increased by greater than threefold in hepatocyte membranes from senescent (24-mo-old) compared with young adult (7-mo-old) rats and that this phenomenon was blocked by food restriction, which is known to retard aging processes in rodents. Competition-binding studies revealed a mixed population of ß1- and ß2-AR subtypes in liver membranes over the adult life span, with a trend for greater ß2-AR density with age. Expression of both ß-AR subtype mRNAs in rat liver increased with age, whereas ß2- but not ß1-AR protein levels declined in livers of old animals. Immunoreactive ß2- but not ß1-ARs were preferentially distributed in pericentral hepatic regions. Levels of GRK2/3 and ß-arrestin 2 proteins, which are involved in downregulation of agonist-activated GPCRs, including ß-ARs, increased during aging. Insofar as sympathetic tone increases with age, our findings suggest that, despite enhanced agonist-mediated downregulation of hepatic ß-ARs preferentially affecting the ß2-AR subtype, increased generation of both receptor subtypes during aging augments the pool of plasma membrane-bound ß-ARs coupled to AC in hepatocytes. This study thus identifies one or both ß-AR subtypes as possible therapeutic targets involved in aberrant hepatic processes of glucose and lipid metabolism during aging.


Subject(s)
Aging/metabolism , Cell Membrane/metabolism , Energy Metabolism , Hepatocytes/metabolism , Liver/metabolism , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Age Factors , Aging/genetics , Animals , Caloric Restriction , Energy Metabolism/genetics , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism , Gene Expression Regulation , Glucose/metabolism , Ligands , Lipid Metabolism , Liver/physiopathology , Male , Rats, Inbred F344 , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...