Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
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.
FASEB J ; 38(17): e70018, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39212304

ABSTRACT

Albuminuria is characterized by a disruption of the glomerular filtration barrier, which is composed of the fenestrated endothelium, the glomerular basement membrane, and the slit diaphragm. Nephrin is a major component of the slit diaphragm. Apart from hemodynamic effects, Ang II enhances albuminuria by ß-Arrestin2-mediated nephrin endocytosis. Blocking the AT1 receptor with candesartan and irbesartan reduces the Ang II-mediated nephrin-ß-Arrestin2 interaction. The inhibition of MAPK ERK 1/2 blocks Ang II-enhanced nephrin-ß-Arrestin2 binding. ERK 1/2 signaling, which follows AT1 receptor activation, is mediated by G-protein signaling, EGFR transactivation, and ß-Arrestin2 recruitment. A mutant AT1 receptor defective in EGFR transactivation and ß-Arrestin2 recruitment reduces the Ang II-mediated increase in nephrin ß-Arrestin2 binding. The mutation of ß-Arrestin2K11,K12, critical for AT1 receptor binding, completely abrogates the interaction with nephrin, independent of Ang II stimulation. ß-Arrestin2K11R,K12R does not influence nephrin cell surface expression. The data presented here deepen our molecular understanding of a blood-pressure-independent molecular mechanism of AT-1 receptor blockers (ARBs) in reducing albuminuria.


Subject(s)
Angiotensin II , Endocytosis , Membrane Proteins , Receptor, Angiotensin, Type 1 , Endocytosis/drug effects , Endocytosis/physiology , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Angiotensin II/pharmacology , Angiotensin II/metabolism , Humans , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , MAP Kinase Signaling System/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Mice , Albuminuria/metabolism , Podocytes/metabolism , Podocytes/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Biphenyl Compounds/pharmacology , Irbesartan/pharmacology , HEK293 Cells , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Benzimidazoles , Tetrazoles
3.
Biochem Pharmacol ; 226: 116362, 2024 08.
Article in English | MEDLINE | ID: mdl-38871335

ABSTRACT

AKT, also known as protein kinase B (PKB), serves as a crucial regulator of numerous biological functions, including cell growth, metabolism, and tumorigenesis. Increasing evidence suggests that the kinase activity of AKT is regulated via ubiquitination by various E3 ligase enzymes in response to different stimuli. However, the molecular mechanisms underlying insulin-induced AKT ubiquitination are not yet fully understood. Here, we show that activation of the insulin receptor (IR) leads to enhanced ubiquitination of AKT1 at K8 and K14 residues, facilitated by the cytosolic E3 ubiquitin ligase enzyme, TRAF6. Further investigation using AKT1 mutants with modified nucleocytoplasmic shuttling properties reveals that TRAF6-mediated AKT1 ubiquitination occurs within the nucleus in a ß-Arr2-dependent manner. The nuclear entry of TRAF6 depends on importin ß1, while ß-Arr2 regulates this process by facilitating the interaction between TRAF6 and importin ß1. Additionally, the ubiquitination of AKT1 is essential for its translocation to the activated IR on the plasma membrane, where it plays a functional role in recruiting Glut4 and facilitating glucose uptake. This study uncovers the cellular components and processes involved in insulin-induced ubiquitination and activation of AKT1, providing insights and detailed strategies for manipulating AKT1.


Subject(s)
Cell Nucleus , Insulin , Proto-Oncogene Proteins c-akt , TNF Receptor-Associated Factor 6 , Ubiquitination , beta-Arrestin 2 , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitination/physiology , Ubiquitination/drug effects , Insulin/metabolism , Insulin/pharmacology , Animals , TNF Receptor-Associated Factor 6/metabolism , Cell Nucleus/metabolism , Mice , Humans , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , HEK293 Cells
4.
BMC Musculoskelet Disord ; 25(1): 451, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844905

ABSTRACT

OBJECTIVE: Temporomandibular joint osteoarthritis (TMJOA) is a chronic degenerative joint disorder characterized by extracellular matrix degeneration and inflammatory response of condylar cartilage. ß-arrestin2 is an important regulator of inflammation response, while its role in TMJOA remains unknown. The objective of this study was to investigate the role of ß-arrestin2 in the development of TMJOA at the early stage and the underlying mechanism. METHODS: A unilateral anterior crossbite (UAC) model was established on eight-week-old wild-type (WT) and ß-arrestin2 deficiency mice to simulate the progression of TMJOA. Hematoxylin-eosin (HE) staining and microcomputed tomography (micro-CT) analysis were used for histological and radiographic assessment. Immunohistochemistry was performed to detect the expression of inflammatory and degradative cytokines, as well as autophagy related factors. Terminal-deoxynucleotidyl transferase mediated nick end labeling (TUNEL) assay was carried out to assess chondrocyte apoptosis. RESULTS: The loss of ß-arrestin2 aggravated cartilage degeneration and subchondral bone destruction in the model of TMJOA at the early stage. Furthermore, in UAC groups, the expressions of degradative (Col-X) and inflammatory (TNF-α and IL-1ß) factors in condylar cartilage were increased in ß-arrestin2 null mice compared with WT mice. Moreover, the loss of ß-arrestin2 promoted apoptosis and autophagic process of chondrocytes at the early stage of TMJOA. CONCLUSION: In conclusion, we demonstrated for the first time that ß-arrestin2 plays a protective role in the development of TMJOA at the early stage, probably by inhibiting apoptosis and autophagic process of chondrocytes. Therefore, ß-arrestin2 might be a potential therapeutic target for TMJOA, providing a new insight for the treatment of TMJOA at the early stage.


Subject(s)
Cartilage, Articular , Disease Models, Animal , Mandibular Condyle , Mice, Knockout , Osteoarthritis , Temporomandibular Joint Disorders , beta-Arrestin 2 , Animals , Osteoarthritis/metabolism , Osteoarthritis/pathology , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Mandibular Condyle/pathology , Mandibular Condyle/metabolism , Mandibular Condyle/diagnostic imaging , Mice , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/pathology , Temporomandibular Joint Disorders/diagnostic imaging , Temporomandibular Joint Disorders/etiology , Chondrocytes/metabolism , Chondrocytes/pathology , Mice, Inbred C57BL , Apoptosis , Temporomandibular Joint/pathology , Temporomandibular Joint/metabolism , Temporomandibular Joint/diagnostic imaging , Male , X-Ray Microtomography , Autophagy/physiology
5.
Nat Metab ; 6(7): 1268-1281, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38871982

ABSTRACT

Incretin-based therapies are highly successful in combatting obesity and type 2 diabetes1. Yet both activation and inhibition of the glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) in combination with glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) activation have resulted in similar clinical outcomes, as demonstrated by the GIPR-GLP-1R co-agonist tirzepatide2 and AMG-133 (ref. 3) combining GIPR antagonism with GLP-1R agonism. This underlines the importance of a better understanding of the GIP system. Here we show the necessity of ß-arrestin recruitment for GIPR function, by combining in vitro pharmacological characterization of 47 GIPR variants with burden testing of clinical phenotypes and in vivo studies. Burden testing of variants with distinct ligand-binding capacity, Gs activation (cyclic adenosine monophosphate production) and ß-arrestin 2 recruitment and internalization shows that unlike variants solely impaired in Gs signalling, variants impaired in both Gs and ß-arrestin 2 recruitment contribute to lower adiposity-related traits. Endosomal Gs-mediated signalling of the variants shows a ß-arrestin dependency and genetic ablation of ß-arrestin 2 impairs cyclic adenosine monophosphate production and decreases GIP efficacy on glucose control in male mice. This study highlights a crucial impact of ß-arrestins in regulating GIPR signalling and overall preservation of biological activity that may facilitate new developments in therapeutic targeting of the GIPR system.


Subject(s)
Phenotype , Receptors, Gastrointestinal Hormone , beta-Arrestins , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Animals , Mice , Humans , beta-Arrestins/metabolism , Genetic Variation , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Signal Transduction , Gastric Inhibitory Polypeptide/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Obesity/metabolism , Obesity/genetics , Male , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/genetics
6.
Biochem Pharmacol ; 224: 116190, 2024 06.
Article in English | MEDLINE | ID: mdl-38604257

ABSTRACT

Arrestins are key negative regulators of G Protein-Coupled Receptors (GPCRs) through mediation of G protein desensitisation and receptor internalisation. Arrestins can also contribute to signal transduction by scaffolding downstream signalling effectors for activation. GPCR kinase (GRK) enzymes phosphorylate the intracellular C-terminal domain, or intracellular loop regions of GPCRs to promote arrestin interaction. There are seven different GRK subtypes, which may uniquely phosphorylate the C-terminal tail in a type of 'phosphorylation barcode,' potentially differentially contributing to arrestin translocation and arrestin-dependent signalling. Such contributions may be exploited to develop arrestin-biased ligands. Here, we examine the effect of different GRK subtypes on the ability to promote translocation of arrestin-2 and arrestin-3 to the cannabinoid CB1 receptor (CB1) with a range of ligands. We find that most GRK subtypes (including visual GRK1) can enhance arrestin-2 and -3 translocation to CB1, and that GRK-dependent changes in arrestin-2 and arrestin-3 translocation were broadly shared for most agonists tested. GRK2/3 generally enhanced arrestin translocation more than the other GRK subtypes, with some small differences between ligands. We also explore the interplay between G protein activity and GRK2/3-dependent arrestin translocation, highlighting that high-efficacy G protein agonists will cause GRK2/3 dependent arrestin translocation. This study supports the hypothesis that arrestin-biased ligands for CB1 must engage GRK5/6 rather than GRK2/3, and G protein-biased ligands must have inherently low efficacy.


Subject(s)
Arrestins , Protein Transport , Receptor, Cannabinoid, CB1 , Signal Transduction , Humans , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/agonists , Signal Transduction/physiology , HEK293 Cells , Arrestins/metabolism , Protein Transport/physiology , GTP-Binding Proteins/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , Animals , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics
7.
Hypertension ; 81(6): 1332-1344, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38629290

ABSTRACT

BACKGROUND: ANG (angiotensin II) elicits dipsogenic and pressor responses via activation of the canonical Gαq (G-protein component of the AT1R [angiotensin type 1 receptor])-mediated AT1R in the subfornical organ. Recently, we demonstrated that ARRB2 (ß-arrestin 2) global knockout mice exhibit a higher preference for salt and exacerbated pressor response to deoxycorticosterone acetate salt. However, whether ARRB2 within selective neuroanatomical nuclei alters physiological responses to ANG is unknown. Therefore, we hypothesized that ARRB2, specifically in the subfornical organ, counterbalances maladaptive dipsogenic and pressor responses to the canonical AT1R signaling. METHODS: Male and female Arrb2FLOX mice received intracerebroventricular injection of either adeno-associated virus (AAV)-Cre-GFP (green fluorescent protein) to induce brain-specific deletion of ARRB2 (Arrb2ICV-Cre). Arrb2FLOX mice receiving ICV-AAV-GFP were used as control (Arrb2ICV-Control). Infection with ICV-AAV-Cre primarily targeted the subfornical organ with few off targets. Fluid intake was evaluated using the 2-bottle choice paradigm with 1 bottle containing water and 1 containing 0.15 mol/L NaCl. RESULTS: Arrb2ICV-Cre mice exhibited a greater pressor response to acute ICV-ANG infusion. At baseline conditions, Arrb2ICV-Cre mice exhibited a significant increase in saline intake compared with controls, resulting in a saline preference. Furthermore, when mice were subjected to water-deprived or sodium-depleted conditions, which would naturally increase endogenous ANG levels, Arrb2ICV-Cre mice exhibited elevated saline intake. CONCLUSIONS: Overall, these data indicate that ARRB2 in selective cardiovascular nuclei in the brain, including the subfornical organ, counterbalances canonical AT1R responses to both exogenous and endogenous ANG. Stimulation of the AT1R/ARRB axis in the brain may represent a novel strategy to treat hypertension.


Subject(s)
Blood Pressure , Homeostasis , Subfornical Organ , beta-Arrestin 2 , Animals , Female , Male , Mice , Angiotensin II/pharmacology , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Blood Pressure/physiology , Blood Pressure/genetics , Brain/metabolism , Homeostasis/physiology , Mice, Knockout , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Subfornical Organ/metabolism
8.
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
9.
Eur J Pharmacol ; 968: 176419, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38360293

ABSTRACT

Glucagon-like peptide-1 receptor (GLP-1R) is a prime drug target for type 2 diabetes and obesity. The ligand initiated GLP-1R interaction with G protein has been well studied, but not with ß-arrestin 1/2. Therefore, bioluminescence resonance energy transfer (BRET), mutagenesis and an operational model were used to evaluate the roles of 85 extracellular surface residues on GLP-1R in ß-arrestin 1/2 recruitment triggered by three representative GLP-1R agonists (GLP-1, exendin-4 and oxyntomodulin). Residues selectively regulated ß-arrestin 1/2 recruitment for diverse ligands, and ß-arrestin isoforms were identified. Mutation of residues K130-S136, L142 and Y145 on the transmembrane helix 1 (TM1)-extracellular domain (ECD) linker decreased ß-arrestin 1 recruitment but increased ß-arrestin 2 recruitment. Other extracellular loop (ECL) mutations, including P137A, Q211A, D222A and M303A selectively affected ß-arrestin 1 recruitment while D215A, L217A, Q221A, S223A, Y289A, S301A, F381A and I382A involved more in ß-arrestin 2 recruitment for the ligands. Oxyntomodulin engaged more broadly with GLP-1R extracellular surface to drive ß-arrestin 1/2 recruitment than GLP-1 and exendin-4; I147, W214 and L218 involved in ß-arrestin 1 recruitment, while L141, D215, L218, D293 and F381 in ß-arrestin 2 recruitment for oxyntomodulin particularly. Additionally, the non-conserved residues on ß-arrestin 1/2 C-domains contributed to interaction with GLP-1R. Further proteomic profiling of GLP-1R stably expressed cell line upon ligand stimulation with or without ß-arrestin 1/2 overexpression demonstrated both commonly and biasedly regulated proteins and pathways associated with cognate ligands and ß-arrestins. Our study offers valuable information about ligand induced ß-arrestin recruitment mediated by GLP-1R and consequent intracellular signaling events.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , beta-Arrestin 1/metabolism , Exenatide/pharmacology , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Ligands , Oxyntomodulin/pharmacology , Proteomics , Glucagon-Like Peptide 1/metabolism , beta-Arrestins/metabolism
10.
J Clin Periodontol ; 51(6): 742-753, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38267365

ABSTRACT

AIM: To investigate the specific role of arrestin beta-2 (ARRB2) in the progression of periodontitis and the underlying mechanisms. MATERIALS AND METHODS: Single-cell RNA sequencing data were used to analyse gene expression in periodontal tissues from healthy controls and patients with periodontitis. Real-time quantitative polymerase chain reaction, Western blotting and immunohistochemical staining were performed to detect the expression of ARRB2. Furthermore, a ligature-induced periodontitis model was created. Using radiographic and histological methods, RNA sequencing and luciferase assay, the role of ARRB2 in periodontitis and the underlying mechanisms were explored. Finally, the therapeutic effect of melatonin, an inhibitor of activating transcription factor 6 (ATF6), on periodontitis in mice was assessed in both in vivo and in vitro experiments. RESULTS: ARRB2 expression was up-regulated in inflammatory periodontal tissue. In the ligature-induced mouse model, Arrb2 knockout exacerbated alveolar bone loss (ABL) and extracellular matrix (ECM) degradation. ARRB2 exerted a negative regulatory effect on ATF6, an essential targeted gene. Melatonin ameliorated ABL and an imbalance in ECM remodelling in Arrb2-deficient periodontitis mice. CONCLUSIONS: ARRB2 mediates ECM remodelling via inhibition of the ATF6 signalling pathway, which ultimately exerts a protective effect on periodontal tissues.


Subject(s)
Activating Transcription Factor 6 , Disease Models, Animal , Extracellular Matrix , Periodontitis , beta-Arrestin 2 , Animals , Humans , Male , Mice , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/genetics , Alveolar Bone Loss/metabolism , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Disease Progression , Extracellular Matrix/metabolism , Melatonin/metabolism , Melatonin/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Periodontitis/metabolism , Periodontitis/genetics , Signal Transduction
11.
Hypertension ; 81(1): 6-16, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37449411

ABSTRACT

ß-arrestins are a family of intracellular signaling proteins that play a key role in regulating the activity of G protein-coupled receptors. The angiotensin-II type 1 receptor is an important G protein-coupled receptor involved in the regulation of cardiovascular function and has been implicated in the progression of cardiovascular diseases. In addition to canonical G protein signaling, G protein-coupled receptors including the angiotensin-II type 1 receptor can signal via ß-arrestin. Dysregulation of ß-arrestin signaling has been linked to several cardiovascular diseases including hypertension, atherosclerosis, and heart failure. Understanding the role of ß-arrestins in these conditions is critical to provide new therapeutic targets for the treatment of cardiovascular disease. In this review, we will discuss the beneficial and maladaptive physiological outcomes of angiotensin-II type 1 receptor-dependent ß-arrestin activation in different cardiovascular diseases.


Subject(s)
Cardiovascular Diseases , Humans , beta-Arrestins , Arrestins/metabolism , Signal Transduction , Receptor, Angiotensin, Type 1/metabolism , Angiotensins/metabolism , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , beta-Arrestin 1/metabolism , Angiotensin II/metabolism
12.
Biochem Pharmacol ; 228: 115988, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38159685

ABSTRACT

The histamine H3 receptor (H3R) is a neurotransmitter receptor that is primarily found in the brain, where it controls the release and synthesis of histamine, as well as the release of other neurotransmitters (e.g. dopamine, serotonin). Notably, 20 H3R isoforms are differentially expressed in the human brain as a consequence of alternative gene splicing. The hH3R-445, -415, -365 and -329 isoforms contain the prototypical GPCR (7TM) structure, yet exhibit deletions in the third intracellular loop, a structural domain that is pivotal for G protein-coupling, signaling and regulation. To date, the physiological relevance underlying the individual and combinatorial function of hH3R isoforms remains poorly understood. Nevertheless, given their significant implication in physiological processes (e.g. cognition, homeostasis) and neurological disorders (e.g. Alzheimer's and Parkinson's disease, schizophrenia), widespread targeting of hH3R isoforms by drugs may lead to on-target side effects in brain regions that are unaffected by disease. To this end, isoform- and/or pathway-selective targeting of hH3R isoforms by biased agonists could be of therapeutic relevance for the development of region- and disease-specific drugs. Hence, we have evaluated ligand biased signaling at the hH3R-445, -415, -365 and -329 isoforms across various Gαi/o-mediated (i.e. [35S]GTPγS accumulation, cAMP inhibition, pERK1/2 activation, pAKT T308/S473 activation) and non Gαi/o-mediated (i.e. ß-arrestin2 recruitment) endpoints that are relevant to neurological diseases. Our findings indicate that H3R agonists display significantly altered patterns in their degree of ligand bias, in a pathway- and isoform-dependent manner, underlining the significance to investigate GPCRs with multiple isoforms to improve development of selective drugs. SUBJECT CATEGORY: Neuropharmacology.


Subject(s)
Protein Isoforms , Receptors, Histamine H3 , Humans , Receptors, Histamine H3/metabolism , Ligands , Protein Isoforms/metabolism , Protein Isoforms/agonists , HEK293 Cells , Signal Transduction/drug effects , Signal Transduction/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Histamine Agonists/pharmacology
13.
J Biol Chem ; 299(11): 105293, 2023 11.
Article in English | MEDLINE | ID: mdl-37774973

ABSTRACT

ß-arrestins play a key role in G protein-coupled receptor (GPCR) internalization, trafficking, and signaling. Whether ß-arrestins act independently of G protein-mediated signaling has not been fully elucidated. Studies using genome-editing approaches revealed that whereas G proteins are essential for mitogen-activated protein kinase activation by GPCRs., ß-arrestins play a more prominent role in signal compartmentalization. However, in the absence of G proteins, GPCRs may not activate ß-arrestins, thereby limiting the ability to distinguish G protein from ß-arrestin-mediated signaling events. We used ß2-adrenergic receptor (ß2AR) and its ß2AR-C tail mutant expressed in human embryonic kidney 293 cells wildtype or CRISPR-Cas9 gene edited for Gαs, ß-arrestin1/2, or GPCR kinases 2/3/5/6 in combination with arrestin conformational sensors to elucidate the interplay between Gαs and ß-arrestins in controlling gene expression. We found that Gαs is not required for ß2AR and ß-arrestin conformational changes, ß-arrestin recruitment, and receptor internalization, but that Gαs dictates the GPCR kinase isoforms involved in ß-arrestin recruitment. By RNA-Seq analysis, we found that protein kinase A and mitogen-activated protein kinase gene signatures were activated by stimulation of ß2AR in wildtype and ß-arrestin1/2-KO cells but absent in Gαs-KO cells. These results were validated by re-expressing Gαs in the corresponding KO cells and silencing ß-arrestins in wildtype cells. These findings were extended to cellular systems expressing endogenous levels of ß2AR. Overall, our results support that Gs is essential for ß2AR-promoted protein kinase A and mitogen-activated protein kinase gene expression signatures, whereas ß-arrestins initiate signaling events modulating Gαs-driven nuclear transcriptional activity.


Subject(s)
GTP-Binding Proteins , Gene Expression Regulation , Receptors, Adrenergic, beta-2 , beta-Arrestins , Humans , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , beta-Arrestins/genetics , beta-Arrestins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/genetics , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , HEK293 Cells , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Protein Structure, Tertiary , Protein Isoforms , Enzyme Activation/genetics
14.
Cell Death Dis ; 14(7): 422, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443143

ABSTRACT

ß-arrestin 2 (ARRB2) is functionally implicated in cancer progression via various signaling pathways. However, its role in lung cancer remains unclear. To obtain clinical insight on its function in lung cancer, microarray data from lung tumor tissues (LTTs) and matched lung normal tissues (mLNTs) of primary non-small cell lung cancer (NSCLC) patients (n = 37) were utilized. ARRB2 expression levels were markedly decreased in all 37 LTTs compared to those in matched LNTs of NSCLC patients. They were significantly co-related to enrichment gene sets associated with oncogenic and cancer genes. Importantly, Gene Set Enrichment Analysis (GSEA) between three LTTs with highly down-regulated ARRB2 and three LTTs with lowly down-regulated ARRB2 revealed significant enrichments related to toll-like receptor (TLR) signaling and autophagy genes in three LTTs with highly down-regulated ARRB2, suggesting that ARRB2 was negatively involved in TLR-mediated signals for autophagy induction in lung cancer. Biochemical studies for elucidating the molecular mechanism revealed that ARRB2 interacted with TNF receptor-associated factor 6 (TRAF6) and Beclin 1 (BECN1), thereby inhibiting the ubiquitination of TRAF6-TAB2 to activate NF-κB and TRAF6-BECN1 for autophagy stimulated by TLR3 and TLR4, suggesting that ARRB2 could inhibit the TRAF6-TAB2 signaling axis for NF-κB activation and TRAF6-BECN1 signaling axis for autophagy in response to TLR3 and TLR4. Notably, ARRB2-knockout (ARRB2KO) lung cancer cells exhibited marked enhancements of cancer migration, invasion, colony formation, and proliferation in response to TLR3 and TLR4 stimulation. Altogether, our current data suggest that ARRB2 can negatively regulate lung cancer progression by inhibiting TLR3- and TLR4-induced autophagy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , NF-kappa B/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Lung Neoplasms/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 3/metabolism , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Toll-Like Receptors/metabolism , Lung/metabolism , Autophagy/genetics , Adaptor Proteins, Signal Transducing/metabolism
15.
Sci Adv ; 9(18): eadf7737, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37134170

ABSTRACT

The glucagon-like peptide-1 receptor (GLP-1R) is a major type 2 diabetes therapeutic target. Stimulated GLP-1Rs are rapidly desensitized by ß-arrestins, scaffolding proteins that not only terminate G protein interactions but also act as independent signaling mediators. Here, we have assessed in vivo glycemic responses to the pharmacological GLP-1R agonist exendin-4 in adult ß cell-specific ß-arrestin 2 knockout (KO) mice. KOs displayed a sex-dimorphic phenotype consisting of weaker acute responses that improved 6 hours after agonist injection. Similar effects were observed for semaglutide and tirzepatide but not with biased agonist exendin-phe1. Acute cyclic adenosine 5'-monophosphate increases were impaired, but desensitization reduced in KO islets. The former defect was attributed to enhanced ß-arrestin 1 and phosphodiesterase 4 activities, while reduced desensitization co-occurred with impaired GLP-1R recycling and lysosomal targeting, increased trans-Golgi network signaling, and reduced GLP-1R ubiquitination. This study has unveiled fundamental aspects of GLP-1R response regulation with direct application to the rational design of GLP-1R-targeting therapeutics.


Subject(s)
Diabetes Mellitus, Type 2 , Animals , Mice , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Mice, Knockout
16.
Hum Hered ; 88(1): 68-78, 2023.
Article in English | MEDLINE | ID: mdl-37100034

ABSTRACT

INTRODUCTION: The role of ARRB2 in cardiovascular disease has recently gained increasing attention. However, the association between ARRB2 polymorphisms and heart failure (HF) has not yet been investigated. METHODS: A total of 2,386 hospitalized patients with chronic HF were enrolled as the first cohort and followed up for a mean period of 20.2 months. Meanwhile, ethnically and geographically matched 3,000 individuals without evidence of HF were included as healthy controls. We genotyped the common variant in ARRB2 gene to identify the association between variant and HF. A replicated independent cohort enrolling 837 patients with chronic HF was applied to validate the observed association. A series of function analyses were conducted to illuminate the underlying mechanism. RESULTS: We identified a common variant rs75428611 associated with the prognosis of HF in two-stage population: adjusted p = 0.001, hazard ratio (HR) = 1.31 (1.11-1.54) in additive model and adjusted p = 0.001, HR = 1.39 (1.14-1.69) in dominant model in first-stage population; adjusted p = 0.04, HR = 1.41 (1.02-1.95) in additive model and adjusted p = 0.03, HR = 1.51 (1.03-2.20) in dominant model in replicated stage. However, rs75428611 did not significantly associate with the risk of HF. Functional analysis indicated that rs75428611-G allele increased the promoter activity and the mRNA expression level of ARRB2 by facilitating transcription factor SRF binding but not the A allele. CONCLUSIONS: Our findings demonstrated that rs75428611 in promoter of ARRB2 was associated with the risk of HF mortality. It is a promising potential treatment target for HF.


Subject(s)
Cardiovascular Diseases , Heart Failure , Humans , Prognosis , Heart Failure/genetics , Heart Failure/therapy , Polymorphism, Genetic , Cardiovascular Diseases/genetics , Chronic Disease , Promoter Regions, Genetic/genetics , beta-Arrestin 2/genetics
17.
Neuroscience ; 519: 120-130, 2023 05 21.
Article in English | MEDLINE | ID: mdl-36796753

ABSTRACT

The cytoplasmic multifunctional adaptor protein ß-arrestin 2 (Arrb2) is involved in the occurrence of various nervous system diseases, such as Alzheimer's disease and Parkinson's disease. Previous laboratory studies have shown that the expression and function of the Arrb2 gene was increased in valproic acid-induced autistic mice models. However, few reports have examined the possible role of Arrb2 in the pathogenesis of autism spectrum disorder. Therefore, Arrb2-deficient (Arrb2-/-) mice were further studied to uncover the physiological function of Arrb2 in the nervous system. In this study, we found that Arrb2-/- mice had normal behavioral characteristics compared with wild-type mice. The autophagy marker protein LC3B was decreased in the hippocampus of Arrb2-/- mice compared to wild-type mice. Western blot analysis revealed that deletion of Arrb2 caused hyperactivation of Akt-mTOR signaling in the hippocampus. In addition, abnormal mitochondrial dysfunction was observed in Arrb2-/- hippocampal neurons, which was characterized by a reduction in mitochondrial membrane potential and adenosine triphosphate production and an increase in reactive oxygen species levels. Therefore, this study elucidates the interaction between Arrb2 and the Akt-mTOR signaling pathway and provides insights into the role of Arrb2 in hippocampal neuron autophagy.


Subject(s)
Autism Spectrum Disorder , Proto-Oncogene Proteins c-akt , Mice , Animals , beta-Arrestin 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Autism Spectrum Disorder/pathology , TOR Serine-Threonine Kinases/metabolism , Autophagy/physiology , Hippocampus/metabolism , beta-Arrestin 2/genetics
18.
J Pharmacol Exp Ther ; 385(1): 17-34, 2023 04.
Article in English | MEDLINE | ID: mdl-36669876

ABSTRACT

Tolerance to compounds that target G protein-coupled receptors (GPCRs), such as the cannabinoid type-1 receptor (CB1R), is in part facilitated by receptor desensitization. Processes that mediate CB1R desensitization include phosphorylation of CB1R residues S426 and S430 by a GPCR kinase and subsequent recruitment of the ß-arrestin2 scaffolding protein. Tolerance to cannabinoid drugs is reduced in S426A/S430A mutant mice and ß-arrestin2 knockout (KO) mice according to previous work in vivo. However, the presence of additional phosphorylatable residues on the CB1R C-terminus made it unclear as to whether recruitment to S426 and S430 accounted for all desensitization and tolerance by ß-arrestin2. Therefore, we assessed acute response and tolerance to the cannabinoids delta-9-tetrahydrocannabinol (Δ9-THC) and CP55,940 in S426A/S430A x ß-arrestin2 KO double-mutant mice. We observed both delayed tolerance and increased sensitivity to the antinociceptive and hypothermic effects of CP55,940 in male S426A/S430A single- and double-mutant mice compared with wild-type littermates, but not with Δ9-THC. Female S426A/S430A single- and double-mutant mice were more sensitive to acute antinociception (CP55,940 and Δ9-THC) and hypothermia (CP55,940 only) exclusively after chronic dosing and did not differ in the development of tolerance. These results indicate that phosphorylation of S426 and S430 are likely responsible for ß-arrestin2-mediated desensitization as double-mutant mice did not differ from the S426A/S430A single-mutant model in respect to cannabinoid tolerance and sensitivity. We also found antinociceptive and hypothermic effects from cannabinoid treatment demonstrated by sex-, agonist-, and duration-dependent features. SIGNIFICANCE STATEMENT: A better understanding of the molecular mechanisms involved in tolerance will improve the therapeutic potential of cannabinoid drugs. This study determined that further deletion of ß-arrestin2 does not enhance the delay in cannabinoid tolerance observed in CB1R S426A/S430A mutant mice.


Subject(s)
Cannabinoids , Mice , Male , Female , Animals , Cannabinoids/pharmacology , Dronabinol/pharmacology , beta-Arrestin 2/genetics , Mice, Knockout , Receptors, Cannabinoid , Analgesics/pharmacology , Receptor, Cannabinoid, CB1/genetics
19.
Am J Physiol Cell Physiol ; 324(1): C133-C141, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36440855

ABSTRACT

Stress causes a rapid spike in norepinephrine (NE) levels, leading to gastrointestinal dysfunction. NE reduces the expression of tight junctions (TJs) and aggravates intestinal mucosal damage, but the regulatory mechanism is still unclear. The present study aimed to investigate the molecular mechanisms underlying the regulation of stress-associated duodenal hyperpermeability by NE. Fluorescein isothiocyanate-dextran permeability, transepithelial resistance, immunofluorescence, Western blot, and high-performance liquid chromatography analysis were used in water-immersion restraint stress (WIRS) rats in this study. The results indicate that the duodenal permeability, degradation of TJs, mucosal NE, and ß2-adrenergic receptor (ß2-AR) increased in WIRS rats. The duodenal intracellular cyclic adenosine monophosphate levels were decreased, whereas the expression of ß-arrestin 2 negatively regulates G protein-coupled receptors signaling, was significantly increased. Src recruitment was mediated by ß-arrestin; thus, the levels of Src kinase activation were enhanced in WIRS rats. NE depletion, ß2-AR, or ß-arrestin 2 blockade significantly decreased mucosal permeability and increased TJs expression, suggesting improved mucosal barrier function. Moreover, NE induced an increased duodenal permeability of normal rats with activated ß-arrestin 2/Src signaling, which was significantly inhibited by ß2-AR blockade. The present findings demonstrate that the enhanced NE induced an increased duodenal permeability in WIRS rats through the activated ß2-AR/ß-arrestin 2/Src pathway. This study provides novel insight into the molecular mechanism underlying the regulation of NE on the duodenal mucosal barrier and a new target for treating duodenal ulcers induced by stress.


Subject(s)
Duodenum , Norepinephrine , Animals , Rats , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Water/metabolism , Stress, Physiological , Duodenum/pathology , Duodenum/physiology
20.
J Neuroinflammation ; 19(1): 240, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36183107

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is a prevalent and devastating psychiatric illness. Unfortunately, the current therapeutic practice, generally depending on the serotonergic system for drug treatment is unsatisfactory and shows intractable side effects. Multiple evidence suggests that dopamine (DA) and dopaminergic signals associated with neuroinflammation are highly involved in the pathophysiology of depression as well as in the mechanism of antidepressant drugs, which is still in the early stage of study and well worthy of investigation. METHODS: We established two chronic stress models, including chronic unpredictable mild stress (CUMS), and chronic social defeat stress (CSDS), to complementarily recapitulate depression-like behaviors. Then, hippocampal tissues were used to detect inflammation-related molecules and signaling pathways. Pathological changes in depressive mouse hippocampal astrocytes were examined by RNA sequencing. After confirming the dopamine receptor 2 (Drd2)/ß-arrestin2 signaling changes in the depressive mice brain, we then established the depressive mouse model using the ß-arrestin2 knockout mice or administrating the ß-arrestin2-biased Drd2 agonist to investigate the roles. Label-free mass spectrometry was used to identify the ß-arrestin2-binding proteins as the underlying mechanisms. We modeled neuroinflammation with interleukin-6 (IL-6) and corticosterone treatment and characterized astrocytes using multiple methods including cell viability assay, flow cytometry, and confocal immunofluorescence. RESULTS: Drd2-biased ß-arrestin2 pathway is significantly changed in the progression of depression, and genetic deletion of ß-arrestin2 aggravates neuroinflammation and depressive-like phenotypes. Mechanistically, astrocytic ß-arrestin2 retains STAT3 in the cytoplasm by structural combination with STAT3, therefore, inhibiting the JAK-STAT3 pathway-mediated inflammatory activation. Furtherly, pharmacological activation of Drd2/ß-arrestin2 pathway by UNC9995 abolishes the inflammation-induced loss of astrocytes and ameliorates depressive-like behaviors in mouse model for depression. CONCLUSIONS: Drd2/ß-arrestin2 pathway is a potential therapeutic target for depression and ß-arrestin2-biased Drd2 agonist UNC9995 is identified as a potential anti-depressant strategy for preventing astrocytic dysfunctions and relieving neuropathological manifestations in mouse model for depression, which provides insights for the therapy of depression.


Subject(s)
Astrocytes , Depressive Disorder, Major , Animals , Astrocytes/metabolism , Corticosterone/metabolism , Depression/drug therapy , Depression/etiology , Depressive Disorder, Major/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopamine Agonists/therapeutic use , Hippocampus/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Mice , Mice, Knockout , Receptors, Dopamine D2/metabolism , Stress, Psychological/complications , Stress, Psychological/drug therapy , Stress, Psychological/pathology , beta-Arrestin 1/metabolism , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL