Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Commun Biol ; 7(1): 933, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095612

ABSTRACT

Desensitisation of the mu-opioid receptor (MOR) is proposed to underlie the initiation of opioid analgesic tolerance and previous work has shown that agonist-induced phosphorylation of the MOR C-tail contributes to this desensitisation. Moreover, phosphorylation is important for ß-arrestin recruitment to the receptor, and ligands of different efficacies induce distinct phosphorylation barcodes. The C-tail 370TREHPSTANT379 motif harbours Ser/Thr residues important for these regulatory functions. 375Ser is the primary phosphorylation site of a ligand-dependent, hierarchical, and sequential process, whereby flanking 370Thr, 376Thr and 379Thr get subsequently and rapidly phosphorylated. Here we used GRK KO cells, phosphosite specific antibodies and site-directed mutagenesis to evaluate the contribution of the different GRK subfamilies to ligand-induced phosphorylation barcodes and ß-arrestin2 recruitment. We show that both GRK2/3 and GRK5/6 subfamilies promote phosphorylation of 370Thr and 375Ser. Importantly, only GRK2/3 induce phosphorylation of 376Thr and 379Thr, and we identify these residues as key sites to promote robust ß-arrestin recruitment to the MOR. These data provide insight into the mechanisms of MOR regulation and suggest that the cellular complement of GRK subfamilies plays an important role in determining the tissue responses of opioid agonists.


Subject(s)
Receptors, Opioid, mu , beta-Arrestin 2 , Phosphorylation , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Humans , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , HEK293 Cells , Protein Binding , Animals , G-Protein-Coupled Receptor Kinases/metabolism , G-Protein-Coupled Receptor Kinases/genetics
2.
Commun Biol ; 7(1): 802, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38956302

ABSTRACT

G protein-coupled receptors (GPCRs) are mainly regulated by GPCR kinase (GRK) phosphorylation and subsequent ß-arrestin recruitment. The ubiquitously expressed GRKs are classified into cytosolic GRK2/3 and membrane-tethered GRK5/6 subfamilies. GRK2/3 interact with activated G protein ßγ-subunits to translocate to the membrane. Yet, this need was not linked as a factor for bias, influencing the effectiveness of ß-arrestin-biased agonist creation. Using multiple approaches such as GRK2/3 mutants unable to interact with Gßγ, membrane-tethered GRKs and G protein inhibitors in GRK2/3/5/6 knockout cells, we show that G protein activation will precede GRK2/3-mediated ß-arrestin2 recruitment to activated receptors. This was independent of the source of free Gßγ and observable for Gs-, Gi- and Gq-coupled GPCRs. Thus, ß-arrestin interaction for GRK2/3-regulated receptors is inseparably connected with G protein activation. We outline a theoretical framework of how GRK dependence on free Gßγ can determine a GPCR's potential for biased agonism. Due to this inherent cellular mechanism for GRK2/3 recruitment and receptor phosphorylation, we anticipate generation of ß-arrestin-biased ligands to be mechanistically challenging for the subgroup of GPCRs exclusively regulated by GRK2/3, but achievable for GRK5/6-regulated receptors, that do not demand liberated Gßγ. Accordingly, GRK specificity of any GPCR is foundational for developing arrestin-biased ligands.


Subject(s)
G-Protein-Coupled Receptor Kinases , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Humans , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , HEK293 Cells , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/genetics , G-Protein-Coupled Receptor Kinases/metabolism , G-Protein-Coupled Receptor Kinases/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Phosphorylation , Animals , Signal Transduction
3.
Biochem Pharmacol ; 222: 116119, 2024 04.
Article in English | MEDLINE | ID: mdl-38461904

ABSTRACT

The glucagon-like peptide 1 receptor (GLP-1R) is a validated clinical target for the treatment of type 2 diabetes and obesity. Unlike most G protein-coupled receptors (GPCRs), the GLP-1R undergoes an atypical mode of internalisation that does not require ß-arrestins. While differences in GLP-1R trafficking and ß-arrestin recruitment have been observed between clinically used GLP-1R agonists, the role of G protein-coupled receptor kinases (GRKs) in affecting these pathways has not been comprehensively assessed. In this study, we quantified the contribution of GRKs to agonist-mediated GLP-1R internalisation and ß-arrestin recruitment profiles using cells where endogenous ß-arrestins, or non-visual GRKs were knocked out using CRISPR/Cas9 genome editing. Our results confirm the previously established atypical ß-arrestin-independent mode of GLP-1R internalisation and revealed that GLP-1R internalisation is dependent on the expression of GRKs. Interestingly, agonist-mediated GLP-1R ß-arrestin 1 and ß-arrestin 2 recruitment were differentially affected by endogenous GRK knockout with ß-arrestin 1 recruitment more sensitive to GRK knockout than ß-arrestin 2 recruitment. Moreover, individual overexpression of GRK2, GRK3, GRK5 or GRK6 in a newly generated GRK2/3/4/5/6 HEK293 cells, rescued agonist-mediated ß-arrestin 1 recruitment and internalisation profiles to similar levels, suggesting that there is no specific GRK isoform that drives these pathways. This study advances mechanistic understanding of agonist-mediated GLP-1R internalisation and provides novel insights into how GRKs may fine-tune GLP-1R signalling.


Subject(s)
Diabetes Mellitus, Type 2 , G-Protein-Coupled Receptor Kinases , Humans , Arrestins/genetics , Arrestins/metabolism , beta-Arrestin 1/metabolism , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , beta-Arrestins/metabolism , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , HEK293 Cells , Phosphorylation , Receptors, G-Protein-Coupled/metabolism
4.
Sci Signal ; 17(823): eadd9139, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38349966

ABSTRACT

Some G protein-coupled receptors (GPCRs) demonstrate biased signaling such that ligands of the same receptor exclusively or preferentially activate certain downstream signaling pathways over others. This phenomenon may result from ligand-specific receptor phosphorylation by GPCR kinases (GRKs). GPCR signaling can also exhibit location bias because GPCRs traffic to and signal from subcellular compartments in addition to the plasma membrane. Here, we investigated whether GRKs contributed to location bias in GPCR signaling. GRKs translocated to endosomes after stimulation of the chemokine receptor CXCR3 or other GPCRs in cultured cells. GRK2, GRK3, GRK5, and GRK6 showed distinct patterns of recruitment to the plasma membrane and to endosomes depending on the identity of the biased ligand used to activate CXCR3. Analysis of engineered forms of GRKs that localized to either the plasma membrane or endosomes demonstrated that biased CXCR3 ligands elicited different signaling profiles that depended on the subcellular location of the GRK. Each GRK exerted a distinct effect on the regulation of CXCR3 engagement of ß-arrestin, internalization, and activation of the downstream effector kinase ERK. Our work highlights a role for GRKs in location-biased GPCR signaling and demonstrates the complex interactions between ligands, GRKs, and cellular location that contribute to biased signaling.


Subject(s)
G-Protein-Coupled Receptor Kinases , Signal Transduction , Ligands , Signal Transduction/physiology , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism
5.
Biochem Soc Trans ; 51(2): 715-724, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37013982

ABSTRACT

G protein-coupled receptors (GPCRs) are key modulators of cell signaling. Multiple GPCRs are present in the heart where they regulate cardiac homeostasis including processes such as myocyte contraction, heart rate and coronary blood flow. GPCRs are pharmacological targets for several cardiovascular disorders including heart failure (HF) such as beta-adrenergic receptor (ßAR) blockers and angiotensin II receptor (AT1R) antagonists. The activity of GPCRs are finely regulated by GPCR kinases (GRKs), which phosphorylate agonist-occupied receptors and start the process of desensitization. Among the seven members of the GRK family, GRK2 and GRK5 are predominantly expressed in the heart, where they exhibit both canonical and non-canonical functions. Both kinases are known to be increased in cardiac pathologies and contribute to pathogenesis through their roles in different cellular compartments. Lowering or inhibiting their actions mediate cardioprotective effects against pathological cardiac growth and failing heart. Therefore, given their importance in cardiac dysfunction, these kinases are drawing attention as promising targets for the treatment of HF, which needs improved therapies. Over the past three decades, broad knowledge on GRK inhibition in HF has been gained by studies using genetically engineered animal models or through gene therapy with peptide inhibitors or using small molecule inhibitors. In this mini review, we summarize the work focusing on GRK2 and GRK5 but also discuss a couple of the non-abundant cardiac subtypes and their multi-functional roles in the normal and diseased heart and the potential and therapeutic targets.


Subject(s)
G-Protein-Coupled Receptor Kinases , Heart Failure , Animals , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , G-Protein-Coupled Receptor Kinases/therapeutic use , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Signal Transduction , Receptors, G-Protein-Coupled
6.
Front Immunol ; 13: 1039803, 2022.
Article in English | MEDLINE | ID: mdl-36451830

ABSTRACT

G-protein coupled receptor kinases (GRKs) participate in the regulation of chemokine receptors by mediating receptor desensitization. They can be recruited to agonist-activated G-protein coupled receptors (GPCRs) and phosphorylate their intracellular parts, which eventually blocks signal propagation and often induces receptor internalization. However, there is growing evidence that GRKs can also control cellular functions beyond GPCR regulation. Immune cells commonly express two to four members of the GRK family (GRK2, GRK3, GRK5, GRK6) simultaneously, but we have very limited knowledge about their interplay in primary immune cells. In particular, we are missing comprehensive studies comparing the role of this GRK interplay for (a) multiple GPCRs within one leukocyte type, and (b) one specific GPCR between several immune cell subsets. To address this issue, we generated mouse models of single, combinatorial and complete GRK knockouts in four primary immune cell types (neutrophils, T cells, B cells and dendritic cells) and systematically addressed the functional consequences on GPCR-controlled cell migration and tissue localization. Our study shows that combinatorial depletions of GRKs have pleiotropic and cell-type specific effects in leukocytes, many of which could not be predicted. Neutrophils lacking all four GRK family members show increased chemotactic migration responses to a wide range of GPCR ligands, whereas combinatorial GRK depletions in other immune cell types lead to pro- and anti-migratory responses. Combined depletion of GRK2 and GRK6 in T cells and B cells shows distinct functional outcomes for (a) one GPCR type in different cell types, and (b) different GPCRs in one cell type. These GPCR-type and cell-type specific effects reflect in altered lymphocyte chemotaxis in vitro and localization in vivo. Lastly, we provide evidence that complete GRK deficiency impairs dendritic cell homeostasis, which unexpectedly results from defective dendritic cell differentiation and maturation in vitro and in vivo. Together, our findings demonstrate the complexity of GRK functions in immune cells, which go beyond GPCR desensitization in specific leukocyte types. Furthermore, they highlight the need for studying GRK functions in primary immune cells to address their specific roles in each leukocyte subset.


Subject(s)
G-Protein-Coupled Receptor Kinases , Receptors, G-Protein-Coupled , Animals , Mice , G-Protein-Coupled Receptor Kinases/genetics , Ligands , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Leukocytes
7.
J Biol Chem ; 298(12): 102636, 2022 12.
Article in English | MEDLINE | ID: mdl-36273582

ABSTRACT

In the vertebrate retina, phosphorylation of photoactivated visual pigments in rods and cones by G protein-coupled receptor kinases (GRKs) is essential for sustained visual function. Previous in vitro analysis demonstrated that GRK1 and GRK7 are phosphorylated by PKA, resulting in a reduced capacity to phosphorylate rhodopsin. In vivo observations revealed that GRK phosphorylation occurs in the dark and is cAMP dependent. In many vertebrates, including humans and zebrafish, GRK1 is expressed in both rods and cones while GRK7 is expressed only in cones. However, mice express only GRK1 in both rods and cones and lack GRK7. We recently generated a mutation in Grk1 that deletes the phosphorylation site, Ser21. This mutant demonstrated delayed dark adaptation in mouse rods but not in cones in vivo, suggesting GRK1 may serve a different role depending upon the photoreceptor cell type in which it is expressed. Here, zebrafish were selected to evaluate the role of cAMP-dependent GRK phosphorylation in cone photoreceptor recovery. Electroretinogram analyses of larvae treated with forskolin show that elevated intracellular cAMP significantly decreases recovery of the cone photoresponse, which is mediated by Grk7a rather than Grk1b. Using a cone-specific dominant negative PKA transgene, we show for the first time that PKA is required for Grk7a phosphorylation in vivo. Lastly, immunoblot analyses of rod grk1a-/- and cone grk1b-/- zebrafish and Nrl-/- mouse show that cone-expressed Grk1 does not undergo cAMP-dependent phosphorylation in vivo. These results provide a better understanding of the function of Grk phosphorylation relative to cone adaptation and recovery.


Subject(s)
G-Protein-Coupled Receptor Kinases , Retinal Cone Photoreceptor Cells , Zebrafish Proteins , Zebrafish , Animals , G-Protein-Coupled Receptor Kinase 1/genetics , G-Protein-Coupled Receptor Kinase 1/metabolism , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Phosphorylation , Retinal Cone Photoreceptor Cells/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
J Biol Chem ; 298(9): 102351, 2022 09.
Article in English | MEDLINE | ID: mdl-35940305

ABSTRACT

G protein-coupled receptor (GPCR) kinases (GRKs) and arrestins mediate GPCR desensitization, internalization, and signaling. The spatial pattern of GPCR phosphorylation is predicted to trigger these discrete GRK and arrestin-mediated functions. Here, we provide evidence that distal carboxyl-terminal tail (C-tail), but not proximal, phosphorylation of the chemokine receptor CXCR4 specifies ßarrestin1 (ßarr1)-dependent signaling. We demonstrate by pharmacologic inhibition of GRK2/3-mediated phosphorylation of the chemokine receptor CXCR4 coupled with site-directed mutagenesis and bioluminescence resonance energy transfer approaches that distal, not proximal, C-tail phosphorylation sites are required for recruitment of the adaptor protein STAM1 (signal-transducing adaptor molecule) to ßarr1 and focal adhesion kinase phosphorylation but not extracellular signal-regulated kinase 1/2 phosphorylation. In addition, we show that GPCRs that have similarly positioned C-tail phosphoresidues are also able to recruit STAM1 to ßarr1. However, although necessary for some GPCRs, we found that distal C-tail sites might not be sufficient to specify recruitment of STAM1 to ßarr1 for other GPCRs. In conclusion, this study provides evidence that distal C-tail phosphorylation sites specify GRK-ßarrestin-mediated signaling by CXCR4 and other GPCRs.


Subject(s)
Arrestin , G-Protein-Coupled Receptor Kinases , Arrestin/metabolism , Arrestins/metabolism , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Phosphorylation/physiology , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , beta-Arrestins/genetics , beta-Arrestins/metabolism
9.
J Biol Chem ; 298(10): 102421, 2022 10.
Article in English | MEDLINE | ID: mdl-36030052

ABSTRACT

Recent studies identified a missense mutation in the gene coding for G protein-coupled receptor kinase 6 (GRK6) that segregates with type 2 diabetes (T2D). To better understand how GRK6 might be involved in T2D, we used pharmacological inhibition and genetic knockdown in the mouse ß-cell line, MIN6, to determine whether GRK6 regulates insulin dynamics. We show inhibition of GRK5 and GRK6 increased insulin secretion but reduced insulin processing while GRK6 knockdown revealed these same processing defects with reduced levels of cellular insulin. GRK6 knockdown cells also had attenuated insulin secretion but enhanced proinsulin secretion consistent with decreased processing. In support of these findings, we demonstrate GRK6 rescue experiments in knockdown cells restored insulin secretion after glucose treatment. The altered insulin profile appears to be caused by changes in the proprotein convertases, the enzymes responsible for proinsulin to insulin conversion, as GRK6 knockdown resulted in significantly reduced convertase expression and activity. To identify how the GRK6-P384S mutation found in T2D patients might affect insulin processing, we performed biochemical and cell biological assays to study the properties of the mutant. We found that while GRK6-P384S was more active than WT GRK6, it displayed a cytosolic distribution in cells compared to the normal plasma membrane localization of GRK6. Additionally, GRK6 overexpression in MIN6 cells enhanced proinsulin processing, while GRK6-P384S expression had little effect. Taken together, our data show that GRK6 regulates insulin processing and secretion in a glucose-dependent manner and provide a foundation for understanding the contribution of GRK6 to T2D.


Subject(s)
Diabetes Mellitus, Type 2 , G-Protein-Coupled Receptor Kinases , Insulin , Proinsulin , Animals , Mice , Diabetes Mellitus, Type 2/genetics , Glucose/pharmacology , Insulin/metabolism , Proinsulin/genetics , Proinsulin/metabolism , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Cell Line
10.
Protein Expr Purif ; 185: 105890, 2021 09.
Article in English | MEDLINE | ID: mdl-33971243

ABSTRACT

Human G-protein coupled receptor kinase 6 (GRK6) belongs to the GRK4 kinase subfamily of the G protein-coupled receptor kinase family which comprises of GRK1, GRK2, and GRK4. These kinases phosphorylate ligand-activated G-protein coupled receptors (GPCRs), driving heterotrimeric G protein coupling, desensitization of GPCR, and ß-arrestin recruitment. This reaction series mediates cellular signal pathways for cell survival, proliferation, migration and chemotaxis. GRK6 is a kinase target in multiple myeloma since it is highly expressed in myeloma cells compared to epithelial cells and has a significant role in mediating the chemotactic responses of T and B-lymphocytes. To support structure-based drug design, we describe three human GRK6 constructs, GRK6, GRK6His/EK, and GRK6His/TEV, designed for protein expression in Spodoptera frugiperda Sf9 insect cells. The first construct did not contain any purification tag whereas the other two constructs contained the His10 affinity tag, which increased purification yields. We report here that all three constructs of GRK6 were overexpressed in Sf9 insect cells and purified to homogeneity at levels that were suitable for co-crystallization of GRK6 with potential inhibitors. The yields of purified GRK6, GRK6His/EK, and GRK6His/TEV were 0.3 mg, 0.8 mg and 0.7 mg per liter of cell culture, respectively. In addition, we have shown that GRK6His/TEV with the His10 tag removed was highly homogeneous and monodisperse as observed by dynamic light scattering measurement and actively folded as exhibited by circular dichroism spectroscopy. The described methods will support the structure-based development of additional therapeutics against multiple myeloma.


Subject(s)
G-Protein-Coupled Receptor Kinases/isolation & purification , Neoplasm Proteins/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Animals , Antineoplastic Agents/chemical synthesis , Baculoviridae/genetics , Baculoviridae/metabolism , Chromatography/methods , Cloning, Molecular , Drug Design , G-Protein-Coupled Receptor Kinases/chemistry , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sf9 Cells , Spodoptera
11.
Epilepsia ; 62(3): 659-670, 2021 03.
Article in English | MEDLINE | ID: mdl-33570167

ABSTRACT

OBJECTIVE: One third of epilepsy patients do not become seizure-free using conventional medication. Therefore, there is a need for alternative treatments. Preclinical research using designer receptors exclusively activated by designer drugs (DREADDs) has demonstrated initial success in suppressing epileptic activity. Here, we evaluated whether long-term chemogenetic seizure suppression could be obtained in the intraperitoneal kainic acid rat model of temporal lobe epilepsy, when DREADDs were selectively expressed in excitatory hippocampal neurons. METHODS: Epileptic male Sprague Dawley rats received unilateral hippocampal injections of adeno-associated viral vector encoding the inhibitory DREADD hM4D(Gi), preceded by a cell-specific promotor targeting excitatory neurons. The effect of clozapine-mediated DREADD activation on dentate gyrus evoked potentials and spontaneous electrographic seizures was evaluated. Animals were systemically treated with single (.1 mg/kg/24 h) or repeated (.1 mg/kg/6 h) injections of clozapine. In addition, long-term continuous release of clozapine and olanzapine (2.8 mg/kg/7 days) using implantable minipumps was evaluated. All treatments were administered during the chronic epileptic phase and between 1.5 and 13.5 months after viral transduction. RESULTS: In the DREADD group, dentate gyrus evoked potentials were inhibited after clozapine treatment. Only in DREADD-expressing animals, clozapine reduced seizure frequency during the first 6 h postinjection. When administered repeatedly, seizures were suppressed during the entire day. Long-term treatment with clozapine and olanzapine both resulted in significant seizure-suppressing effects for multiple days. Histological analysis revealed DREADD expression in both hippocampi and some cortical regions. However, lesions were also detected at the site of vector injection. SIGNIFICANCE: This study shows that inhibition of the hippocampus using chemogenetics results in potent seizure-suppressing effects in the intraperitoneal kainic acid rat model, even 1 year after viral transduction. Despite a need for further optimization, chemogenetic neuromodulation represents a promising treatment prospect for temporal lobe epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Clozapine/therapeutic use , Epilepsy, Temporal Lobe/drug therapy , Olanzapine/therapeutic use , Receptors, Neurotransmitter/genetics , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/physiopathology , Disease Models, Animal , Evoked Potentials/physiology , G-Protein-Coupled Receptor Kinases/drug effects , G-Protein-Coupled Receptor Kinases/genetics , Gene Editing/methods , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Rats , Rats, Sprague-Dawley , Receptors, Neurotransmitter/drug effects , Seizures/prevention & control
12.
J Biol Chem ; 296: 100473, 2021.
Article in English | MEDLINE | ID: mdl-33639163

ABSTRACT

The type II sodium-dependent phosphate cotransporter (NPT2A) mediates renal phosphate uptake. The NPT2A is regulated by parathyroid hormone (PTH) and fibroblast growth factor 23, which requires Na+/H+ exchange regulatory factor-1 (NHERF1), a multidomain PDZ-containing phosphoprotein. Phosphocycling controls the association between NHERF1 and the NPT2A. Here, we characterize the critical involvement of G protein-coupled receptor kinase 6A (GRK6A) in mediating PTH-sensitive phosphate transport by targeted phosphorylation coupled with NHERF1 conformational rearrangement, which in turn allows phosphorylation at a secondary site. GRK6A, through its carboxy-terminal PDZ recognition motif, binds NHERF1 PDZ1 with greater affinity than PDZ2. However, the association between NHERF1 PDZ2 and GRK6A is necessary for PTH action. Ser162, a PKCα phosphorylation site in PDZ2, regulates the binding affinity between PDZ2 and GRK6A. Substitution of Ser162 with alanine (S162A) blocks the PTH action but does not disrupt the interaction between NHERF1 and the NPT2A. Replacement of Ser162 with aspartic acid (S162D) abrogates the interaction between NHERF1 and the NPT2A and concurrently PTH action. We used amber codon suppression to generate a phosphorylated Ser162(pSer162)-PDZ2 variant. KD values determined by fluorescence anisotropy indicate that incorporation of pSer162 increased the binding affinity to the carboxy terminus of GRK6A 2-fold compared with WT PDZ2. Molecular dynamics simulations predict formation of an electrostatic network between pSer162 and Asp183 of PDZ2 and Arg at position -1 of the GRK6A PDZ-binding motif. Our results suggest that PDZ2 plays a regulatory role in PTH-sensitive NPT2A-mediated phosphate transport and phosphorylation of Ser162 in PDZ2 modulates the interaction with GRK6A.


Subject(s)
G-Protein-Coupled Receptor Kinases/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Biological Transport , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , G-Protein-Coupled Receptor Kinases/genetics , Humans , Ion Transport , Molecular Dynamics Simulation , PDZ Domains/genetics , Parathyroid Hormone/metabolism , Phosphates/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Protein Conformation , Sodium-Hydrogen Exchangers/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
13.
PLoS One ; 16(2): e0247087, 2021.
Article in English | MEDLINE | ID: mdl-33600497

ABSTRACT

BACKGROUND: G protein-coupled receptor kinase 6 (GRK6) is part of the G protein-coupled receptor kinase family, whose members act as key regulators of seven-transmembrane receptor signalling. GRK6 seems to play a role in regulation of inflammatory processes, but mechanisms of transcriptional regulation of GRK6 expression in inflammatory cell lines have not been characterized. Protein kinase C (PKC) signalling is also involved in inflammatory regulation and an impact of PKC activation on GRK6 protein expression was described previously. Thus, the aim of this study was to 1) characterize the GRK6 promoter, and 2) investigate a potential influence of PKC on GRK6 expression. METHODS: Five deletion constructs of the GRK6 promoter were cloned. After transient transfection into a human T cell line, promoter activity was assessed using luciferase reporter gene assays. Putative transcription factor binding sites were identified, mutated, and binding was investigated using electrophoretic mobility shift assays (EMSA). Following stimulation with a PKC activator, GRK6 expression on mRNA and protein levels was assessed by reverse transcriptase qPCR and Western blots. RESULTS: Investigation of the GRK6 promoter revealed a putative cAMP responsive element (CRE), whose mutation led to decreased promoter activity (p = 0.0006). Functionality of the CRE binding protein (CREB) binding site was verified in EMSA blots. Stimulation with a PKC activator resulted in decreased GRK6 promoter activity (p = 0.0027), mRNA (p = 0.04) and protein expression. CONCLUSION: We characterized the human GRK6 promoter and identified promoter activity to be influenced by a CREB binding site. PKC might be one determinant contributing to altered GRK6 expression.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , G-Protein-Coupled Receptor Kinases/genetics , Response Elements/genetics , Base Sequence , Binding Sites , Cyclic AMP Response Element-Binding Protein/chemistry , Electrophoretic Mobility Shift Assay , G-Protein-Coupled Receptor Kinases/chemistry , G-Protein-Coupled Receptor Kinases/metabolism , Humans , Jurkat Cells , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Protein Binding , Protein Kinase C/metabolism , RNA, Messenger/metabolism
14.
Biochim Biophys Acta Mol Cell Res ; 1868(4): 118946, 2021 04.
Article in English | MEDLINE | ID: mdl-33385424

ABSTRACT

The zebrafish retina expresses four recoverin genes (rcv1a, rcv1b, rcv2a and rcv2b) and four opsin kinase genes (grk1a, grk1b, grk7a and grk7b) coding for recoverin and G protein-coupled receptor kinase (opsin kinase) paralogs, respectively. Both protein groups are suggested to form regulatory complexes in rod and cone outer segments, but at present, we lack information about co-localization of recoverin and opsin kinases in zebrafish retinae and which protein-protein interacting pairs form. We analyzed the distribution and co-localization of recoverin and opsin kinase expression in the zebrafish retina. For this purpose, we used custom-tailored monospecific antibodies revealing that the amount of recoverin paralogs in a zebrafish retina can differ by more than one order of magnitude with the highest amount for recoverin 1a and 2b. Further, immunohistochemical labelling showed presence of recoverin 1a in all rod cell compartments, but it only co-localized with opsin kinase 1a in rod outer segments. In contrast, recoverin 2b was only detected in double cones and co-localized with opsin kinases 1b, 7a and 7b. Further, we investigated the interaction between recoverin and opsin kinase variants by surface plasmon resonance spectroscopy indicating interaction of recoverin 1a and recoverin 2b with all opsin kinases. However, binding kinetics for recoverin 1a differed from those observed with recoverin 2b that showed slower association and dissociation processes. Our results indicate diverse recoverin and opsin kinase properties due to differential expression and interaction profiles.


Subject(s)
G-Protein-Coupled Receptor Kinases/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Recoverin/metabolism , Zebrafish/metabolism , Animals , Cloning, Molecular , G-Protein-Coupled Receptor Kinases/genetics , Gene Expression Regulation , Protein Interaction Maps , Recoverin/genetics , Surface Plasmon Resonance , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Cancer Res ; 81(2): 501-514, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33158816

ABSTRACT

The ability of a receptor to preferentially activate only a subset of available downstream signal cascades is termed biased signaling. Although comprehensively recognized for the G protein-coupled receptors (GPCR), this process is scarcely explored downstream of receptor tyrosine kinases (RTK), including the cancer-relevant insulin-like growth factor-1 receptor (IGF1R). Successful IGF1R targeting requires receptor downregulation, yet therapy-mediated removal from the cell surface activates cancer-protective ß-arrestin-biased signaling (ß-arr-BS). As these overlapping processes are initiated by the ß-arr/IGF1R interaction and controlled by GPCR-kinases (GRK), we explored GRKs as potential anticancer therapeutic targets to disconnect IGF1R downregulation and ß-arr-BS. Transgenic modulation demonstrated that GRK2 inhibition or GRK6 overexpression enhanced degradation of IGF1R, but both scenarios sustained IGF1-induced ß-arr-BS. Pharmacologic inhibition of GRK2 by the clinically approved antidepressant, serotonin reuptake inhibitor paroxetine (PX), recapitulated the effects of GRK2 silencing with dose- and time-dependent IGF1R downregulation without associated ß-arr-BS. In vivo, PX treatment caused substantial downregulation of IGF1R, suppressing the growth of Ewing's sarcoma xenografts. Functional studies reveal that PX exploits the antagonism between ß-arrestin isoforms; in low ligand conditions, PX favored ß-arrestin1/Mdm2-mediated ubiquitination/degradation of IGF1R, a scenario usually exclusive to ligand abundancy, making PX more effective than antibody-mediated IGF1R downregulation. This study provides the rationale, molecular mechanism, and validation of a clinically feasible concept for "system bias" targeting of the IGF1R to uncouple downregulation from signaling. Demonstrating system bias as an effective anticancer approach, our study reveals a novel strategy for the rational design or repurposing of therapeutics to selectively cross-target the IGF1R or other RTK. SIGNIFICANCE: This work provides insight into the molecular and biological roles of biased signaling downstream RTK and provides a novel "system bias" strategy to increase the efficacy of anti-IGF1R-targeted therapy in cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/pathology , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , Gene Expression Regulation, Neoplastic , Receptor, IGF Type 1/metabolism , Sarcoma, Ewing/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Proliferation , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinases/genetics , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Nude , Phosphorylation , Receptor, IGF Type 1/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
16.
Biochem Biophys Res Commun ; 530(4): 692-698, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32768194

ABSTRACT

In this report, we explored if G protein coupled receptor kinases (GRKs) can help modulate the heat stress responses of Caenorhabditis (C.) elegans. Loss of function grk-2 C. elegans mutants were more tolerant to increases in heat and display an ability for heat stress-associated hormesis at a longer exposure time unlike the wild type N2 animals and the loss of function grk-1 C. elegans mutants. The loss of function grk-1 mutants recovered more from acute heat stress compared to the wild type N2 animals. Animals with low Ce-GRK2 protein expression showed increased DAF-16 nuclear localization during the early stages of heat stress exposure compared to the other RNAi-treated animals, demonstrating altered insulin/insulin-like growth factor signaling (IIS) pathway activity in response to the stress. pdk-1 and akt-1 may play key roles in conjunction with Ce-GRK2 in the heat stress response. Collectively, these findings demonstrate that GRKs influence C. elegans heat stress behaviors.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , G-Protein-Coupled Receptor Kinases/metabolism , Heat-Shock Response , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Forkhead Transcription Factors/metabolism , G-Protein-Coupled Receptor Kinases/genetics , Longevity , Loss of Function Mutation , Thermotolerance
17.
Mol Pain ; 16: 1744806920930858, 2020.
Article in English | MEDLINE | ID: mdl-32484026

ABSTRACT

AIMS: The arcuate nucleus is a vital brain region for coursing of pain command. G protein-coupled kinase 6 (GRK6) accommodates signaling through G protein-coupled receptors. Studies have demonstrated that GRK6 is involved in inflammatory pain and neuropathic pain. The present study was designed to explore the role and the underlying mechanism of GRK6 in arcuate nucleus of chronic visceral pain. METHODS: Chronic visceral pain of rats was induced by neonatal maternal deprivation and evaluated by monitoring the threshold of colorectal distension. Western blotting, immunofluorescence, real-time quantitative polymerase chain reaction techniques, and Nissl staining were employed to determine the expression and mutual effect of GRK6 with nuclear factor κB (NF-κB). RESULTS: Expression of GRK6 in arcuate nucleus was significantly reduced in neonatal maternal deprivation rats when compared with control rats. GRK6 was mainly expressed in arcuate nucleus neurons, but not in astrocytes, and a little in microglial cells. Neonatal maternal deprivation reduced the percentage of GRK6-positive neurons of arcuate nucleus. Overexpression of GRK6 by Lentiviral injection into arcuate nucleus reversed chronic visceral pain in neonatal maternal deprivation rats. Furthermore, the expression of NF-κB in arcuate nucleus was markedly upregulated in neonatal maternal deprivation rats. NF-κB selective inhibitor pyrrolidine dithiocarbamate suppressed chronic visceral pain in neonatal maternal deprivation rats. GRK6 and NF-κB were expressed in the arcuate nucleus neurons. Importantly, overexpression of GRK6 reversed NF-κB expression at the protein level. In contrast, injection of pyrrolidine dithiocarbamate once daily for seven consecutive days did not alter GRK6 expression in arcuate nucleus of neonatal maternal deprivation rats. CONCLUSIONS: Present data suggest that GRK6 might be a pivotal molecule participated in the central mechanisms of chronic visceral pain, which might be mediated by inhibiting NF-κB signal pathway. Overexpression of GRK6 possibly represents a potential strategy for therapy of chronic visceral pain.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Chronic Pain/metabolism , Down-Regulation , G-Protein-Coupled Receptor Kinases/genetics , Maternal Deprivation , NF-kappa B/metabolism , Up-Regulation/genetics , Visceral Pain/metabolism , Animals , Animals, Newborn , Chronic Pain/complications , Down-Regulation/drug effects , G-Protein-Coupled Receptor Kinases/metabolism , Male , NF-kappa B/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Pyrrolidines/pharmacology , Rats, Sprague-Dawley , Thiocarbamates/pharmacology , Up-Regulation/drug effects , Visceral Pain/complications
18.
J Biol Chem ; 294(20): 8023-8036, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30936203

ABSTRACT

G protein-coupled receptor (GPCR) signaling is regulated by members of the protein kinase C (PKC) and GPCR kinase (GRK) families, although the relative contribution of each to GPCR function varies among specific GPCRs. The CXC motif receptor 4 (CXCR4) is a member of the GPCR superfamily that binds the CXC motif chemokine ligand 12 (CXCL12), initiating signaling that is subsequently terminated in part by internalization and lysosomal degradation of CXCR4. The purpose of this study is to define the relative contribution of PKC and GRK to CXCR4 signaling attenuation by studying their effects on CXCR4 lysosomal trafficking and degradation. Our results demonstrate that direct activation of PKC via the phorbol ester phorbol 12-myristate 13-acetate (PMA) mimics CXCL12-mediated desensitization, internalization, ubiquitination, and lysosomal trafficking of CXCR4. In agreement, heterologous activation of PKC by stimulating the chemokine receptor CXCR5 with its ligand, CXCL13, also mimics CXCL12-mediated desensitization, internalization, ubiquitination, and lysosomal degradation of CXCR4. Similar to CXCL12, PMA promotes PKC-dependent phosphorylation of serine residues within CXCR4 C-tail that are required for binding and ubiquitination by the E3 ubiquitin ligase AIP4 (atrophin-interacting protein 4). However, inhibition of PKC activity does not alter CXCL12-mediated ubiquitination and degradation of CXCR4, suggesting that other kinases are also required. Accordingly, siRNA-mediated depletion of GRK6 results in decreased degradation and ubiquitination of CXCR4. Overall, these results suggest that PKC and GRK6 contribute to unique aspects of CXCR4 phosphorylation and lysosomal degradation to ensure proper signal propagation and termination.


Subject(s)
Lysosomes/metabolism , Proteolysis , Receptors, CXCR4/metabolism , Signal Transduction , Ubiquitination , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , HEK293 Cells , HeLa Cells , Humans , Lysosomes/genetics , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Receptors, CXCR4/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
19.
FEBS Open Bio ; 9(4): 605-617, 2019 04.
Article in English | MEDLINE | ID: mdl-30984536

ABSTRACT

We previously reported that the expression of G protein-coupled receptor kinase 6 (GRK6) is significantly downregulated in lung adenocarcinoma (LADC) tissues, and low expression levels of GRK6 are correlated with poor survival prognosis. However, the specific regulatory mechanisms and functions of GRK6 in LADC remain unknown. Here, we report that GRK6 mRNA expression levels are downregulated in LADC tissues compared to those in matched adjacent non-tumor tissues (P < 0.001). The promoter of the GRK6 gene was found to be hypermethylated in LADC tissues, and its methylation was correlated with both GRK6 expression and pathology grade. GRK6 promoter hypermethylation may predict shorter overall survival. Treatment with 5-aza-2'-deoxycytidine significantly enhanced GRK6 gene expression. Four binding sites of CCAAT/enhancer-binding protein-α (C/EBPα) in the CpG island of the GRK6 gene promoter were predicted in silico, of which three sites were further confirmed by ChIP. Decreased binding of C/EBPα to binding sites 1, 3 and 4 of the GRK6 gene promoter was observed in LADC tissues. Inhibition of C/EBPα significantly inhibited GRK6 expression, while overexpression of C/EBPα significantly promoted GRK6 expression. In addition, overexpression of GRK6 significantly suppressed, while GRK6 knockdown promoted cell migration and invasion. Overexpression of GRK6 enhanced E-cadherin expression and suppressed vimentin expression, and silencing of GRK6 had the opposite effects. Furthermore, ectopic expression of GRK6 significantly decreased matrix metalloproteinase (MMP) 2 and MMP7 protein expression levels. Our findings suggest that hypermethylation of the GRK6 gene promoter suppressed binding of C/EBPα, thereby contributing to the promotion of cell migration and invasion. The methylation status of the GRK6 promoter might be suitable for use as an epigenetic biomarker, and the C/EBPα-GRK6 signaling pathway may be a potential target for LADC.


Subject(s)
Adenocarcinoma of Lung/genetics , CCAAT-Enhancer-Binding Proteins/genetics , G-Protein-Coupled Receptor Kinases/genetics , Gene Expression Regulation, Neoplastic/genetics , Lung Neoplasms/genetics , Promoter Regions, Genetic , Adenocarcinoma of Lung/physiopathology , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Movement , DNA Methylation , Female , G-Protein-Coupled Receptor Kinases/chemistry , G-Protein-Coupled Receptor Kinases/metabolism , Gene Knockdown Techniques , Humans , Lung Neoplasms/physiopathology , Male , Middle Aged , Neoplastic Processes , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Mol Immunol ; 106: 12-21, 2019 02.
Article in English | MEDLINE | ID: mdl-30576947

ABSTRACT

Chemerin receptor (CMKLR1) is a G protein-coupled receptor (GPCR) implicated in macrophage-mediated inflammation and in several forms of human arthritis. Analogous to other GPCR, CMKLR1 is likely regulated by G protein-coupled receptor kinase (GRK) phosphorylation of intracellular domains in an activation-dependent manner, which leads to recruitment and termination of intracellular signaling via desensitization and internalization of the receptor. The ubiquitously expressed GRK family members include GRK2, GRK3, GRK5, and GRK6, but it is unknown which GRK regulates CMKLR1 cellular and signaling functions. Our data show that activation of CMKLR1 by chemerin in primary macrophages leads to signaling and functional outcomes that are regulated by GRK6 and ß-arrestin 2. We show that arrestin recruitment to CMKLR1 following chemerin stimulation is enhanced with co-expression of GRK6. Further, internalization of endogenous CMKLR1, following the addition of chemerin, is decreased in inflammatory macrophages from GRK6- and ß-arrestin 2-deficient mice. These GRK6- and ß-arrestin 2-deficient macrophages display increased migration toward chemerin and altered AKT and Extracellular-signal Related Kinase (ERK) signaling. Our findings show that chemerin-activated CMKLR1 regulation in inflammatory macrophages is largely GRK6 and ß-arrestin mediated, which may impact innate immunity and have therapeutic implications in rheumatic disease.


Subject(s)
Chemokines/immunology , G-Protein-Coupled Receptor Kinases/immunology , Immunity, Innate , Intercellular Signaling Peptides and Proteins/immunology , Macrophages/immunology , Receptors, G-Protein-Coupled/immunology , beta-Arrestin 2/immunology , Animals , Cell Line , Chemokines/genetics , G-Protein-Coupled Receptor Kinases/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/genetics , Macrophages/pathology , Mice , Mice, Knockout , Receptors, Chemokine , Receptors, G-Protein-Coupled/genetics , Rheumatic Diseases/genetics , Rheumatic Diseases/immunology , Rheumatic Diseases/pathology , beta-Arrestin 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL