251 results on '"β-arrestins"'
Search Results
52. A Comprehensive View of the β-Arrestinome.
- Author
-
Crépieux, Pascale, Poupon, Anne, Langonné-Gallay, Nathalie, Reiter, Eric, Delgado, Javier, Schaefer, Martin H., Bourquard, Thomas, Serrano, Luis, and Kiel, Christina
- Subjects
G protein coupled receptors ,EXTRACELLULAR matrix proteins ,NEUROTRANSMITTERS - Abstract
G protein-coupled receptors (GPCRs) are membrane receptors critically involved in sensing the environment and orchestrating physiological processes. As such, they transduce extracellular signals such as hormone, neurotransmitters, ions, and light into an integrated cell response. The intracellular trafficking, internalization, and signaling ability of ligand-activated GPCRs are controlled by arrestins, adaptor proteins that they interact with upon ligand binding. β-arrestins 1 and 2 in particular are now considered as hub proteins assembling multiprotein complexes to regulate receptor fate and transduce diversified cell responses. While more than 400 β-arrestin interaction partners have been identified so far, much remains to be learnt on how discrimination between so many binding partners is accomplished. Here, we gathered the interacting partners of β-arrestins through database mining and manual curation of the literature to map the β-arrestin interactome (β-arrestinome). We discussed several parameters that determine compatible (AND) or mutually exclusive (XOR) binding of β-arrestin interactors, such as structural constraints, intracellular abundance, or binding affinity. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
53. Genetically encoded intrabody sensors report the interaction and trafficking of β-arrestin 1 upon activation of G-protein–coupled receptors
- Author
-
Punita Kumari, Debarati Roy, Madhu Chaturvedi, Arun K. Shukla, Hemlata Dwivedi-Agnihotri, Shubhi Pandey, Aylin C. Hanyaloglu, Ashish Srivastava, Michel Bouvier, Silvia Sposini, Badr Sokrat, Mithu Baidya, Society for Endocrinology, and Genesis Research Trust
- Subjects
0301 basic medicine ,Biochemistry & Molecular Biology ,Cell signaling ,G protein-coupled receptor (GPCR) ,confocal microscopy ,Endocytosis ,Biochemistry ,Intrabody ,Receptors, G-Protein-Coupled ,Immunoglobulin Fab Fragments ,03 medical and health sciences ,synthetic antibody ,trafficking ,Functional selectivity ,Arrestin ,Humans ,cell signaling ,Molecular Biology ,G-protein–coupled receptors (GPCRs) ,11 Medical and Health Sciences ,β-arrestins ,G protein-coupled receptor ,bioluminescence resonance energy transfer (BRET) ,030102 biochemistry & molecular biology ,biology ,arrestin ,Chemistry ,Cell Biology ,06 Biological Sciences ,biosensors ,protein phosphorylation ,Synthetic antibody ,Cell biology ,Protein Transport ,intrabody ,HEK293 Cells ,beta-Arrestin 1 ,030104 developmental biology ,biased agonism ,biology.protein ,cellular signaling ,Signal transduction ,03 Chemical Sciences ,signal transduction ,hormones, hormone substitutes, and hormone antagonists - Abstract
Agonist stimulation of G protein-coupled receptors (GPCRs) typically leads to phosphorylation of GPCRs and binding to multifunctional proteins called β-arrestins (βarrs). The GPCR-βarr interaction critically contributes to GPCR desensitization, endocytosis, and downstream signaling, and GPCR-βarr complex formation can be used as a generic readout of GPCR and βarr activation. Although several methods are currently available to monitor GPCR-βarr interactions, additional sensors to visualize them may expand the toolbox and complement existing methods. We have previously described antibody fragments (FABs) that recognize activated βarr1 upon its interaction with the vasopressin V2 receptor C-terminal phosphopeptide (V2Rpp). Here, we demonstrate that these FABs efficiently report the formation of a GPCR-βarr1 complex for a broad set of chimeric GPCRs harboring the V2R C terminus. We adapted these FABs to an intrabody format by converting them to single-chain variable fragments (ScFvs) and used them to monitor the localization and trafficking of βarr1 in live cells. We observed that upon agonist simulation of cells expressing chimeric GPCRs, these intrabodies first translocate to the cell surface, followed by trafficking into intracellular vesicles. The translocation pattern of intrabodies mirrored that of βarr1, and the intrabodies co-localized with βarr1 at the cell surface and in intracellular vesicles. Interestingly, we discovered that intrabody sensors can also report βarr1 recruitment and trafficking for several unmodified GPCRs. Our characterization of intrabody sensors for βarr1 recruitment and trafficking expands currently available approaches to visualize GPCR-βarr1 binding, which may help decipher additional aspects of GPCR signaling and regulation.
- Published
- 2020
54. Corrigendum: Biased signaling of protease-activated receptors
- Author
-
Peishen eZhao, Matthew eMetcalf, and Nigel W. Bunnett
- Subjects
Signal Transduction ,G proteins ,Proteases ,biased signaling ,PARs ,β-arrestins ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Published
- 2015
- Full Text
- View/download PDF
55. Carbetocin is a Functional Selective Gq Agonist That Does Not Promote Oxytocin Receptor Recycling After Inducing β-Arrestin-Independent Internalisation.
- Author
-
Passoni, I., Leonzino, M., Gigliucci, V., Chini, B., and Busnelli, M.
- Subjects
- *
OXYTOCIN receptors , *ARRESTINS , *MOLECULAR pharmacology , *BIOLUMINESCENCE , *BIOSENSORS - Abstract
Carbetocin, a long-acting oxytocin analogue, has been reported to elicit interesting and peculiar behavioural effects. The present study investigated the molecular pharmacology of carbetocin, aiming to better understand the molecular basis of its action in the brain. Using bioluminescence resonance energy transfer biosensors, we characterised the effects of carbetocin on the three human oxytocin/vasopressin receptors expressed in the nervous system: the oxytocin receptor ( OXTR) and the vasopressin V1a (V1aR) and V1b (V1bR) receptors. Our results indicate that (i) carbetocin activates the OXTR but not the V1aR and V1bR at which it may act as an antagonist; (ii) carbetocin selectively activates only the OXTR/Gq pathway displaying a strong functional selectivity; (iii) carbetocin is a partial agonist at the OXTR/Gq coupling; (iv) carbetocin promotes OXTR internalisation via a previously unreported β-arrestin-independent pathway; and (v) carbetocin does not induce OXTR recycling to the plasma membrane. Altogether, these molecular pharmacology features identify carbetocin as a substantially different analogue compared to the endogenous oxytocin and, consequently, carbetocin is not expected to mimic oxytocin in the brain. Whether these unique features of carbetocin could be exploited therapeutically remains to be established. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
56. Beta-arrestin1 and 2 differently modulate metabotropic glutamate receptor 7 signaling in rat developmental sevoflurane-induced neuronal apoptosis.
- Author
-
Wang, W.-Y., Wu, X.-M., Jia, L.-J., Zhang, H.-H., Cai, F., Mao, H., Xu, W.-C., Chen, L., Zhang, J., and Hu, S.-F.
- Subjects
- *
GLUTAMATE receptors , *APOPTOSIS , *SEVOFLURANE , *GLUTAMIC acid , *CELL death - Abstract
Beta-arrestins (β-arrs) are initially known as negative regulators of G protein-coupled receptors (GPCRs). Recently, there is increasing evidence suggesting that β-arrs also serve as scaffolds and adapters that mediate distinct intracellular signal transduction initiated by GPCR activation. In the previous study, we have shown that metabotropic glutamate receptor 7 (mGluR7) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling may be involved in the developmental sevoflurane neurotoxicity. In the present study, we showed that activation of mGluR7 with a group III mGluRs orthosteric agonist LAP4 or an atypical mGluR7 allosteric agonist N,N′-bis(diphenylmethyl)-1,2-ethanediamine dihydrochloride (AMN082) significantly attenuated sevoflurane-induced neuronal apoptosis. Interestingly, this neuroprotective role of LAP4 could be partially reduced by β-arr1 small interfering RNA (siRNA) or β-arr2 siRNA transfection. In contrast, β-arr2 siRNA transfection alone abolished the effects of AMN082 on sevoflurane neurotoxicity. In addition, administration of LAP4 or AMN082 significantly enhanced Phospho-ERK1/2 in sevoflurane neurotoxicity, which could be abrogated by β-arr2 siRNA transfection, but not by β-arr1 siRNA transfection. Increased β-arr2-dependent Phospho-ERK1/2 signaling alleviated sevoflurane neurotoxicity by inhibiting bad phosphorylation. We also found that the neuroprotective role of AMN082 was completely reversed by ERK1/2 inhibitor 1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butadiene (U0126). Alternatively, treatment with U0126 partially suppressed the neuroprotective of LAP4, suggesting that other mechanisms may be implicated in this process. Further investigation indicated that, in the scenario of sevoflurane neurotoxicity, application of LAP4 (but not AMN082) increased the interaction of β-arrs with transcriptional factors CREB binding protein (CBP) and p300. LAP4 also enhanced the β-arr1-dependent H3 and H4 acetylation in sevoflurane neurotoxicity. For the behavior study, treatment with LAP4 or AMN082 significantly improved the emotional and spatial learning and memory disorders induced by postnatal sevoflurane exposure. These results suggested that β-arr1 and 2 may differently modulate mGluR7 signaling in developmental sevoflurane neurotoxicity. This study also reveals a β-arr-biased agonism at GPCRs (e.g. mGluR7). [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
57. Metabolic Functions of G Protein-Coupled Receptors and β-Arrestin-Mediated Signaling Pathways in the Pathophysiology of Type 2 Diabetes and Obesity
- Author
-
Xuenan Sun, Da Young Oh, and Camila Oliveira de Souza
- Subjects
obesity ,Gs alpha subunit ,G protein ,Endocrinology, Diabetes and Metabolism ,Review ,Diseases of the endocrine glands. Clinical endocrinology ,GPCRs ,Receptors, G-Protein-Coupled ,Endocrinology ,Heterotrimeric G protein ,Arrestin ,Animals ,Humans ,Receptor ,beta-Arrestins ,β-arrestins ,biased signaling ,G protein-coupled receptor ,biology ,RC648-665 ,Cell biology ,Diabetes Mellitus, Type 2 ,Gq alpha subunit ,biology.protein ,type 2 diabetes ,Signal transduction ,Signal Transduction - Abstract
Seven transmembrane receptors (7TMRs), often termed G protein-coupled receptors (GPCRs), are the most common target of therapeutic drugs used today. Many studies suggest that distinct members of the GPCR superfamily represent potential targets for the treatment of various metabolic disorders including obesity and type 2 diabetes (T2D). GPCRs typically activate different classes of heterotrimeric G proteins, which can be subgrouped into four major functional types: Gαs, Gαi, Gαq/11, and G12/13, in response to agonist binding. Accumulating evidence suggests that GPCRs can also initiate β-arrestin-dependent, G protein-independent signaling. Thus, the physiological outcome of activating a certain GPCR in a particular tissue may also be modulated by β-arrestin-dependent, but G protein-independent signaling pathways. In this review, we will focus on the role of G protein- and β-arrestin-dependent signaling pathways in the development of obesity and T2D-related metabolic disorders.
- Published
- 2021
58. Biased Signaling of Protease-activated Receptors
- Author
-
Peishen eZhao, Matthew eMetcalf, and Nigel William Bunnett
- Subjects
Signal Transduction ,G proteins ,Proteases ,biased signaling ,PARs ,β-arrestins ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
In addition to their role in protein degradation and digestion, proteases can also function as hormone-like signaling molecules that regulate vital patho-physiological processes, including inflammation, hemostasis, pain and repair mechanisms. Certain proteases can signal to cells by cleaving protease-activated receptors (PARs), a family of four G protein-coupled receptors. PARs are expressed by almost all cell types, control important physiological and disease-relevant processes, and are an emerging therapeutic target for major diseases. Most information about PAR activation and function derives from studies of a few proteases, for example thrombin in the case of PAR1, PAR3 and PAR4, and trypsin in the case of PAR2 and PAR4. These proteases cleave PARs at established sites with the extracellular N-terminal domains, and expose tethered ligands that stabilize conformations of the cleaved receptors that activate the canonical pathways of G protein- and/or β-arrestin-dependent signaling. However, a growing number of proteases have been identified that cleave PARs at divergent sites to activate distinct patterns of receptor signaling and trafficking. The capacity of these proteases to trigger distinct signaling pathways is referred to as biased signaling, and can lead to unique patho-physiological outcomes. Given that a different repertoire of proteases are activated in various patho-physiological conditions that may activate PARs by different mechanisms, signaling bias may account for the divergent actions of proteases and PARs. Moreover, therapies that target disease-relevant biased signaling pathways may be more effective and selective approaches for the treatment of protease- and PAR-driven diseases. Thus, rather than mediating the actions of a few proteases, PARs may integrate the biological actions of a wide spectrum of proteases in different patho-physiological conditions.Keywords: PARs, proteases, biased signaling, G proteins, β-arrestins, signal transduction
- Published
- 2014
- Full Text
- View/download PDF
59. Differential Involvement of ACKR3 C-Tail in β-Arrestin Recruitment, Trafficking and Internalization
- Author
-
Henry F. Vischer, Thierry Durroux, Rianna J F de Jonker, Martine J. Smit, J.P. Bebelman, Claudia Perez, Marco Siderius, Jelle van den Bor, Aurélien Zarca, Joyce Heuninck, Vrije Universiteit Amsterdam [Amsterdam] (VU), Institut de Génomique Fonctionnelle (IGF), Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS), Guerineau, Nathalie C., Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Medicinal chemistry, and AIMMS
- Subjects
ACKR3 ,G-Protein-Coupled Receptor Kinase 3 ,G-Protein-Coupled Receptor Kinase 2 ,[SDV]Life Sciences [q-bio] ,Biosensing Techniques ,GRKs ,Chemokine receptor ,0302 clinical medicine ,GPCR ,Fluorescence Resonance Energy Transfer ,Protein phosphorylation ,Phosphorylation ,Internalization ,lcsh:QH301-705.5 ,media_common ,β-arrestins ,0303 health sciences ,protein recruitment ,Chemistry ,chemokine receptor ,General Medicine ,beta-Arrestin 2 ,Endocytosis ,Cell biology ,[SDV] Life Sciences [q-bio] ,homogeneous time resolved fluorescence (HTRF) ,Protein Transport ,beta-Arrestin 1 ,030220 oncology & carcinogenesis ,Protein Binding ,media_common.quotation_subject ,education ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Article ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Arrestin ,Humans ,Protein Interaction Domains and Motifs ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,030304 developmental biology ,G protein-coupled receptor ,Receptors, CXCR ,G protein-coupled receptor kinase ,HEK 293 cells ,Chemokine CXCL12 ,protein phosphorylation ,internalization ,Kinetics ,HEK293 Cells ,bioluminescence energy transfer (BRET) ,lcsh:Biology (General) - 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 β-arrestins upon activation. ACKR3 plays an important role in cancer and vascular diseases. As recruitment of β-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 β-arrestins and G-protein-coupled receptor kinases (GRKs), receptor internalization and trafficking. Results: Upon CXCL12 stimulation, ACKR3 recruits both β-arrestin 1 and 2 with equivalent kinetic profiles. We identified interactions with GRK2, 3 and 5, with GRK2 and 3 being important for β-arrestin recruitment. Upon activation, ACKR3 internalizes and recycles back to the cell membrane. We demonstrate that β-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 β-arrestin1 recruitment. Phosphorylation of the C-tail appears essential for ligand-induced internalization and important for differential β-arrestin recruitment. GRK2 and 3 play a key role in receptor internalization. Moreover, ACKR3 can still internalize when β-arrestin recruitment is impaired or in the absence of β-arrestins, using alternative internalization pathways. Our data indicate that distinct residues within the C-tail of ACKR3 differentially regulate CXCL12-induced β-arrestin recruitment, ACKR3 trafficking and internalization.
- Published
- 2021
60. A streamlined protocol for expression and purification of wild-type β-arrestins.
- Author
-
Yadav MK, Singh V, Saha S, and Shukla AK
- Subjects
- beta-Arrestins metabolism, Protein Binding, Receptors, G-Protein-Coupled metabolism, Arrestins chemistry, Arrestins metabolism, Escherichia coli genetics, Escherichia coli metabolism
- Abstract
The two isoforms of β-arrestins namely β-arrestin 1 and 2 interact with, and regulate a broad repertoire of G protein-coupled receptors (GPCRs). While several protocols have been described in the literature for purification of β-arrestins for biochemical and biophysical studies, some of these protocols involve multiple complicated steps that prolong the process and yield relatively smaller amounts of purified proteins. Here, we describe a simplified and streamlined protocol for expression and purification of β-arrestins using E. coli as an expression host. This protocol is based on N-terminal fusion of GST tag and involves a two-step protocol involving GST-based affinity chromatography and size exclusion chromatography. The protocol described here yields sufficient amounts of high-quality purified β-arrestins suitable for biochemical and structural studies., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
61. Structures of the arginine-vasopressin and oxytocin receptor signaling complexes.
- Author
-
Bous J, Fouillen A, Orcel H, Granier S, Bron P, and Mouillac B
- Subjects
- Animals, Humans, Receptors, Oxytocin genetics, Cryoelectron Microscopy, Vasopressins, Arginine, Mammals, Arginine Vasopressin, Oxytocin
- Abstract
Arginine-vasopressin (AVP) and oxytocin (OT) are neurohypophysial hormones which share a high sequence and structure homology. These are two cyclic C-terminally amidated nonapeptides with different residues at position 3 and 8. In mammals, AVP and OT exert their multiple biological functions through a specific G protein-coupled receptor family: four receptors are identified, the V1a, V1b, V2 receptors (V1aR, V1bR and V2R) and the OT receptor (OTR). The chemical structure of AVP and OT was elucidated in the early 1950s. Thanks to X-ray crystallography and cryo-electron microscopy, it took however 70 additional years to determine the three-dimensional structures of the OTR and the V2R in complex with their natural agonist ligands and with different signaling partners, G proteins and β-arrestins. Today, the comparison of the different AVP/OT receptor structures gives structural insights into their orthosteric ligand binding pocket, their molecular mechanisms of activation, and their interfaces with canonical Gs, Gq and β-arrestin proteins. It also paves the way to future rational drug design and therapeutic compound development. Indeed, agonist, antagonist, biased agonist, or pharmacological chaperone analogues of AVP and OT are promising candidates to regulate different physiological functions and treat several pathologies., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
62. Internalization of somatostatin receptors in brain and periphery.
- Author
-
Csaba Z and Dournaud P
- Subjects
- Humans, Brain metabolism, Receptors, Somatostatin chemistry, Receptors, Somatostatin metabolism, Neoplasms
- Abstract
Somatostatin (SRIF) is a neuropeptide that acts as an important regulator of both endocrine and exocrine secretion and modulates neurotransmission in the central nervous system (CNS). SRIF also regulates cell proliferation in normal tissues and tumors. The physiological actions of SRIF are mediated by a family of five G protein-coupled receptors, called somatostatin receptor (SST) SST
1 , SST2 , SST3 , SST4 , SST5 . These five receptors share similar molecular structure and signaling pathways but they display marked differences in their anatomical distribution, subcellular localization and intracellular trafficking. The SST subtypes are widely distributed in the CNS and peripheral nervous system, in many endocrine glands and tumors, particularly of neuroendocrine origin. In this review, we focus on the agonist-dependent internalization and recycling of the different SST subtypes in vivo in the CNS, peripheral organs and tumors. We also discuss the physiological, pathophysiological and potential therapeutic effects of the intracellular trafficking of SST subtypes., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
63. The sequence Pro295-Thr311 of the hinge region of oestrogen receptor α is involved in ERK1/2 activation via GPR30 in leiomyoma cells.
- Author
-
Leiber, Denis, Burlina, Fabienne, Byrne, Cillian, Robin, Philippe, Piesse, Christophe, Gonzalez, Lucie, Leclercq, Guy, Tanfin, Zahra, and Jacquot, Yves
- Subjects
- *
NUCLEOTIDE sequence , *ESTROGEN , *SMOOTH muscle tumors , *BREAST cancer , *STEROIDS , *ENZYME inhibitors - Abstract
The ERα (oestrogen receptor α)-derived peptide ERα17p activates rapid signalling events in breast carcinoma cells under steroid-deprived conditions. In the present study, we investigated its effects in ELT3 leiomyoma cells under similar conditions. We show that it activates ERK1/2 (extracellular-signal-regulated kinase 1/2), the Gαi protein, the trans-activation of EGFR (epidermal growth factor receptor) and, finally, cell proliferation. It is partially internalized in cells and induces membrane translocation of β-arrestins. The activation of ERK1/2 is abolished by the GPR30 (G-protein-coupled receptor 30) antagonist G15 and GPR30 siRNA. When ERα is down-regulated by prolonged treatment with E2 (oestradiol) or specific ERα siRNA, the peptide response is blunted. Thus the simultaneous presence of GPR30 and ERα is required for the action of ERα17p. In addition, its PLM sequence, which interferes with the formation of the ERα-calmodulin complex, appears to be requisite for the phosphorylation of ERK1/2 and cell proliferation. Hence ERα17p is, to our knowledge, the first known peptide targeting ERα-GPR30 membrane cross-talk and the subsequent receptor-mediated biological effects. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
64. Intracellular signaling mechanisms of the melanocortin receptors: current state of the art.
- Author
-
Rodrigues, Adriana, Almeida, Henrique, and Gouveia, Alexandra
- Subjects
- *
CELLULAR signal transduction , *MELANOCORTIN receptors , *ADRENOCORTICOTROPIC hormone , *LIGANDS (Biochemistry) , *AGOUTI-related peptide - Abstract
The melanocortin system is composed by the agonists adrenocorticotropic hormone and α, β and γ-melanocyte-stimulating hormone, and two naturally occurring antagonists, agouti and agouti-related protein. These ligands act by interaction with a family of five melanocortin receptors (MCRs), assisted by MCRs accessory proteins (MRAPs). MCRs stimulation activates different signaling pathways that mediate a diverse array of physiological processes, including pigmentation, energy metabolism, inflammation and exocrine secretion. This review focuses on the regulatory mechanisms of MCRs signaling, highlighting the differences among the five receptors. MCRs signal through G-dependent and independent mechanisms and their functional coupling to agonists at the cell surface is regulated by interacting proteins, namely MRAPs and β-arrestins. The knowledge of the distinct modulation pattern of MCRs signaling and function may be helpful for the future design of novel drugs able to combine specificity, safety and effectiveness in the course of their therapeutic use. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
65. Gamma-Aminobutyric Acid Induces Tumor Cells Apoptosis via GABAR1·β-Arrestins·JNKs Signaling Module.
- Author
-
Tian, Hui, Wu, Jin-Xia, Shan, Feng-Xiao, Zhang, Shang-Nuan, Cheng, Qian, Zheng, Jun-Nian, and Pei, Dong-Sheng
- Abstract
Gamma-aminobutyric acid (GABA), an inhibitory neurotransmitter in central nervous system, has yet been found to widely exist in tumor tissues to regulate tumor cells growth. However, the function of GABA on inducing tumor cells apoptosis and the potential mechanism are still unclear. In order to detect whether GABA via GABA receptor GABAR1 would activate c-Jun N-terminal kinases (JNKs) to promote tumor cells apoptosis, co-immunoprecipitation assay was used to investigate the association of β-arrestins with GABAR1 and JNKs in the different four cancer cell lines. Our observation demonstrated that β-arrestins, in addition to their role in G protein-coupled receptors desensitization, had an additional function as adapter proteins to recruit JNKs to GABAR1, thereby conferring distinct enzymatic activities upon the receptor, which may trigger JNKs signal pathway involved in the regulation of cellular growth. Activated JNKs subsequently phosphorylated downstream c-Jun to transcribe a wide variety of pro-apoptotic genes. Additionally, GABA up-regulated the ratio of pro-apoptotic protein Bax to anti-apoptotic protein Bcl-2, and thus facilitated caspase-3 cleavage, leading to tumor cells apoptosis in a mitochondrial-dependent pathway. In contrast, GABAR antagonist CGP35348 reversed GABA-induced JNKs phosphorylation and its downstream proteins activation, which consequently restrained tumor cells apoptosis. Taken together, our study suggested that GABA via its receptor GABAR1 recruited β-arrestins to facilitate the activation of JNKs cascade, resulting in tumor cells growth inhibition. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
66. Nitric Oxide and S-Nitrosylation in Cardiac Regulation: G Protein-Coupled Receptor Kinase-2 and β-Arrestins as Targets
- Author
-
Walter J. Koch and Gizem Kayki-Mutlu
- Subjects
0301 basic medicine ,G-Protein-Coupled Receptor Kinase 2 ,GRK2 ,Review ,030204 cardiovascular system & hematology ,Catalysis ,Nitric oxide ,Inorganic Chemistry ,lcsh:Chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Downregulation and upregulation ,nitric oxide ,Receptors, Adrenergic, beta ,Cyclic GMP-Dependent Protein Kinases ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,Cyclic GMP ,lcsh:QH301-705.5 ,Spectroscopy ,beta-Arrestins ,G protein-coupled receptor ,β-arrestins ,G protein-coupled receptor kinase ,S-Nitrosothiols ,biology ,Kinase ,Chemistry ,Beta adrenergic receptor kinase ,Organic Chemistry ,General Medicine ,S-Nitrosylation ,S-nitrosylation ,Computer Science Applications ,Cell biology ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,biology.protein ,Nitric Oxide Synthase ,cGMP-dependent protein kinase ,Signal Transduction - Abstract
Cardiac diseases including heart failure (HF), are the leading cause of morbidity and mortality globally. Among the prominent characteristics of HF is the loss of β-adrenoceptor (AR)-mediated inotropic reserve. This is primarily due to the derangements in myocardial regulatory signaling proteins, G protein-coupled receptor (GPCR) kinases (GRKs) and β-arrestins (β-Arr) that modulate β-AR signal termination via receptor desensitization and downregulation. GRK2 and β-Arr2 activities are elevated in the heart after injury/stress and participate in HF through receptor inactivation. These GPCR regulators are modulated profoundly by nitric oxide (NO) produced by NO synthase (NOS) enzymes through S-nitrosylation due to receptor-coupled NO generation. S-nitrosylation, which is NO-mediated modification of protein cysteine residues to generate an S-nitrosothiol (SNO), mediates many effects of NO independently from its canonical guanylyl cyclase/cGMP/protein kinase G signaling. Herein, we review the knowledge on the NO system in the heart and S-nitrosylation-dependent modifications of myocardial GPCR signaling components GRKs and β-Arrs.
- Published
- 2021
67. TRPV1 Activation Promotes β-arrestin2 Interaction with the Ribosomal Biogenesis Machinery in the Nucleolus: Implications for p53 Regulation and Neurite Outgrowth
- Author
-
Robyn Flynn, Daniel Young, Manon Defaye, Mircea Iftinca, Christophe Altier, Mark G.H. Scott, Nasser Abdullah, Antoine Dufour, Ahmed Hassan, Francina Agosti, University of Calgary, [Institut Cochin] Département Endocrinologie, métabolisme, diabète (EMD) (EMD), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), and SCOTT, Mark
- Subjects
Proteomics ,Nucleolus ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,lcsh:Chemistry ,0302 clinical medicine ,Treacle protein ,RNA Polymerase I ,Ganglia, Spinal ,lcsh:QH301-705.5 ,Spectroscopy ,β-arrestins ,Neurons ,0303 health sciences ,Chemistry ,General Medicine ,beta-Arrestin 2 ,Computer Science Applications ,Cell biology ,Protein Transport ,Signal transduction ,chronic pain ,Nucleophosmin ,Cell Nucleolus ,Protein Binding ,Neurite ,Neuronal Outgrowth ,neuroplasticity ,TRPV Cation Channels ,Article ,Catalysis ,ribosomal biogenesis ,Inorganic Chemistry ,03 medical and health sciences ,RNA polymerase I ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,030304 developmental biology ,Organic Chemistry ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Mice, Inbred C57BL ,TRPV1 ,HEK293 Cells ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cytoplasm ,Tumor Suppressor Protein p53 ,Ribosomes ,030217 neurology & neurosurgery ,Biogenesis - Abstract
International audience; Transient receptor potential vanilloids (TRPV1) are non-selective cation channels that sense and transduce inflammatory pain signals. We previously reported that activation of TRPV1 induced the translocation of β-arrestin2 (ARRB2) from the cytoplasm to the nucleus, raising questions about the functional role of ARRB2 in the nucleus. Here, we determined the ARRB2 nuclear signalosome by conducting a quantitative proteomic analysis of the nucleus-sequestered L395Q ARRB2 mutant, compared to the cytosolic wild-type ARRB2 (WT ARRB2), in a heterologous expression system. We identified clusters of proteins that localize to the nucleolus and are involved in ribosomal biogenesis. Accordingly, L395Q ARRB2 or WT ARRB2 after capsaicin treatment were found to co-localize and interact with the nucleolar marker nucleophosmin (NPM1), treacle protein (TCOF1) and RNA polymerase I (POL I). We further investigated the role of nuclear ARRB2 signaling in regulating neuroplasticity. Using neuroblastoma (neuro2a) cells and dorsal root ganglia (DRG) neurons, we found that L395Q ARRB2 mutant increased POL I activity, inhibited the tumor suppressorp53 (p53) level and caused a decrease in the outgrowth of neurites. Together, our results suggest that the activation of TRPV1 promotes the ARRB2-mediated regulation of ribosomal biogenesis in the nucleolus. The ARRB2-TCOF1-p53 checkpoint signaling pathway might be involved in regulating neurite outgrowth associated with pathological pain conditions.
- Published
- 2021
68. Nodal signals via β-arrestins and RalGTPases to regulate trophoblast invasion.
- Author
-
Law, Jeffrey, Guihua Zhang, Dragan, Magdalena, Postovit, Lynne-Marie, and Bhattacharya, Moshmi
- Subjects
- *
CELLULAR signal transduction , *ARRESTINS , *GUANOSINE triphosphatase , *TROPHOBLAST , *GENETIC regulation , *PLACENTA - Abstract
Placentation is critical for establishing a healthy pregnancy. Trophoblasts mediate implantation and placentation and certain subtypes, most notably extravillous cytotrophoblast, are highly invasive. Trophoblast invasion is tightly regulated by microenvironmental cues that dictate placental morphology and depth. In choriocarcinomas, malignant trophoblast cells become hyperinvasive, breaching the myometrium and leading to major complications. Nodal, a member of the TGF-β superfamily, is expressed throughout the endometrium during the peri-implantation period and in invasive trophoblast cells. Nodal promotes the invasion of numerous types of cancer cells. However, Nodal's role in trophoblast and choriocarcinoma cell invasion is unclear. Here we show that Nodal stimulates the invasion of both the non-malignant HTR-8SV/neo trophoblast and JAR choriocarcinoma cells in a dose-dependent manner. We found that endogenous β-arrestins and Ral GTPases, key regulators of the cell cytoskeleton, are constitutively associated with Nodal receptors (ALK4 and ALK7) in trophoblast cells and that RalA is colocalized with ALK4 in endocytic vesicles. Nodal stimulates endogenous β-arrestin2 to associate with phospho-ERK1/2, and knockdown of β-arrestin or Ral proteins impairs Nodal-induced trophoblast and choriocarcinoma cell invasion. These results demonstrate, for the first time, that β-arrestins and RalGTPases are important regulators of Nodal-induced invasion. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
69. β-Arrestins 1 and 2 are critical regulators of inflammation.
- Author
-
Fan, Hongkuan
- Subjects
- *
ARRESTINS , *INFLAMMATION , *MEMBRANE proteins , *GENETIC regulation , *G protein coupled receptors , *ENDOCYTOSIS , *UBIQUITINATION - Abstract
β-Arrestins 1 and 2 couple to seven trans-membrane receptors and regulate G protein-dependent signaling, receptor endocytosis and ubiquitylation. Recent studies have uncovered several unanticipated functions of β-arrestins, suggesting that the role of β-arrestins in cell signaling is much broader than originally thought. It is now recognized that β-arrestins can transduce receptor signaling independent of G proteins. The expression of β-arrestins is differentially regulated in immune cells and tissues in response to specific inflammatory stimuli, and β-arrestins are critical regulators of the inflammatory response. This review will focus on β-arrestins in immune cells and the impact of altered expression on the pathogenesis of specific inflammatory diseases. Understanding the role of β-arrestins in inflammation may lead to new strategies to treat inflammatory diseases, such as sepsis, rheumatoid arthritis, asthma, multiple sclerosis, inflammatory bowel disease and atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
70. Biased signaling of protease-activated receptors.
- Author
-
Zhao, Peishen, Metcalf, Matthew, and Bunnett, Nigel W.
- Subjects
PROTEOLYTIC enzymes ,ARRESTINS ,INFLAMMATION ,HOMEOSTASIS ,PAIN - Abstract
In addition to their role in protein degradation and digestion, proteases can also function as hormone-like signaling molecules that regulate vital patho-physiological processes, including inflammation, hemostasis, pain, and repair mechanisms. Certain proteases can signal to cells by cleaving protease-activated receptors (PARs), a family of four G protein-coupled receptors. PARs are expressed by almost all cell types, control important physiological and disease-relevant processes, and are an emerging therapeutic target for major diseases. Most information about PAR activation and function derives from studies of a few proteases, for example thrombin in the case of PAR
1 , PAR3 , and PAR4 , and trypsin in the case of PAR2 and PAR4 . These proteases cleave PARs at established sites with the extracellular N-terminal domains, and expose tethered ligands that stabilize conformations of the cleaved receptors that activate the canonical pathways of G protein- and/or β-arrestindependent signaling. However, a growing number of proteases have been identified that cleave PARs at divergent sites to activate distinct patterns of receptor signaling and trafficking. The capacity of these proteases to trigger distinct signaling pathways is referred to as biased signaling, and can lead to unique patho-physiological outcomes. Given that a different repertoire of proteases are activated in various patho-physiological conditions that may activate PARs by different mechanisms, signaling bias may account for the divergent actions of proteases and PARs. Moreover, therapies that target disease-relevant biased signaling pathways may be more effective and selective approaches for the treatment of protease- and PAR-driven diseases. Thus, rather than mediating the actions of a few proteases, PARs may integrate the biological actions of a wide spectrum of proteases in different patho-physiological conditions. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
71. Homology modeling, binding site identification, and docking study of human β-arrestin: an adaptor protein involved in apoptosis.
- Author
-
Chintha, Chetan, Gupta, Nirzari, Ghate, Manjunath, and Vyas, Vivek
- Abstract
β-Arrestins are well-known negative regulators of G-protein-coupled receptors (GPCRs) signaling. The present study is aimed to build human β -arrestins 3D structures by homology modeling followed by identification and characterization of binding sites and there by assessing druggability of the proteins. Based upon BLAST results, bovine β-arrestin (PDB ID:1G4M) was considered as a template for homology modeling. Homology models were constructed using MODELLER program, refined and validated using PROCHECK in which >97 % of residues present in the favored regions of the Ramachandran plots. Since there are no co-ordinates of ligands available in the template, we used various binding site prediction algorithms to predict different binding pockets on the modeled proteins. To examine the response of proteins toward various apoptosis inducers, molecular docking study was carried out. The identification of 3D structures, binding sites for various signaling molecules will guide us designing molecular tools for therapeutic intervention that may prove useful in numerous disorders associated with β-arrestin-GPCRs signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
72. Exploring GPCR‐arrestin interfaces with genetically encoded crosslinkers
- Author
-
Christian Ihling, Stefan Ernicke, Robert Serfling, Vsevolod V. Gurevich, Andrea Sinz, Thore Böttke, Edyta Burda, and Irene Coin
- Subjects
live cells ,genetic structures ,Arrestins ,Cell ,G protein‐coupled receptor ,genetically encoded crosslinkers ,Methods & Resources ,Biochemistry ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,DOCK ,Genetics ,medicine ,Arrestin ,Receptor ,Molecular Biology ,beta-Arrestins ,030304 developmental biology ,G protein-coupled receptor ,Vasopressin receptor ,chemistry.chemical_classification ,0303 health sciences ,Parathyroid Hormone Receptor 1 ,Articles ,β‐arrestins ,eye diseases ,Amino acid ,Cell biology ,medicine.anatomical_structure ,chemistry ,GPCR–arrestin complexes ,sense organs ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
β‐arrestins (βarr1 and βarr2) are ubiquitous regulators of G protein‐coupled receptor (GPCR) signaling. Available data suggest that β‐arrestins dock to different receptors in different ways. However, the structural characterization of GPCR‐arrestin complexes is challenging and alternative approaches to study GPCR‐arrestin complexes are needed. Here, starting from the finger loop as a major site for the interaction of arrestins with GPCRs, we genetically incorporate non‐canonical amino acids for photo‐ and chemical crosslinking into βarr1 and βarr2 and explore binding topologies to GPCRs forming either stable or transient complexes with arrestins: the vasopressin receptor 2 (rhodopsin‐like), the corticotropin‐releasing factor receptor 1, and the parathyroid hormone receptor 1 (both secretin‐like). We show that each receptor leaves a unique footprint on arrestins, whereas the two β‐arrestins yield quite similar crosslinking patterns. Furthermore, we show that the method allows defining the orientation of arrestin with respect to the GPCR. Finally, we provide direct evidence for the formation of arrestin oligomers in the cell., Chemical and photo‐crosslinkers genetically incorporated into β‐arrestins allow the mapping of footprints of different receptors on the arrestin surface in living cells, and to identify intermolecular pairs of proximal amino acids in GPCR‐arrestin complexes.
- Published
- 2020
73. β 2 -Adrenoceptor agonist profiling reveals biased signalling phenotypes for the β 2 -adrenoceptor with possible implications for the treatment of asthma.
- Author
-
De Pascali F, Ippolito M, Wolfe E, Komolov KE, Hopfinger N, Lemenze D, Kim N, Armen RS, An SS, Scott CP, and Benovic JL
- Subjects
- Adrenergic beta-Agonists pharmacology, GTP-Binding Protein alpha Subunits, Gs metabolism, Humans, Phenotype, Signal Transduction, beta-Arrestin 1 metabolism, beta-Arrestins metabolism, beta-Arrestins pharmacology, Asthma drug therapy, Receptors, Adrenergic, beta-2 metabolism
- Abstract
Background and Purpose: β-Adrenoceptor agonists relieve airflow obstruction by activating β
2 -adrenoceptors, which are G protein-coupled receptors (GPCRs) expressed on human airway smooth muscle (HASM) cells. The currently available β-adrenoceptor agonists are balanced agonists, however, and signal through both the stimulatory G protein (Gs )- and β-arrestin-mediated pathways. While Gs signalling is beneficial and promotes HASM relaxation, β-arrestin activation is associated with reduced Gs efficacy. In this context, biased ligands that selectively promote β2 -adrenoceptor coupling to Gs signalling represent a promising strategy to treat asthma. Here, we examined several β-adrenoceptor agonists to identify Gs -biased ligands devoid of β-arrestin-mediated effects., Experimental Approach: Gs -biased ligands for the β2 -adrenoceptor were identified by high-throughput screening and then evaluated for Gs interaction, Gi interaction, cAMP production, β-arrestin interaction, GPCR kinase (GRK) phosphorylation of the receptor, receptor trafficking, ERK activation, and functional desensitization of the β2 -adrenoceptor., Key Results: We identified ractopamine, dobutamine, and higenamine as Gs -biased agonists that activate the Gs /cAMP pathway upon β2 -adrenoceptor stimulation while showing minimal Gi or β-arrestin interaction. Furthermore, these compounds did not induce any receptor trafficking and had reduced GRK5-mediated phosphorylation of the β2 -adrenoceptor. Finally, we observed minimal physiological desensitization of the β2 -adrenoceptor in primary HASM cells upon treatment with biased agonists., Conclusion and Implications: Our work demonstrates that Gs -biased signalling through the β2 -adrenoceptor may prove to be an effective strategy to promote HASM relaxation in the treatment of asthma. Such biased compounds may also be useful in identifying the molecular mechanisms that determine biased signalling and in design of safer drugs., (© 2022 British Pharmacological Society.)- Published
- 2022
- Full Text
- View/download PDF
74. The Two β-Arrestins Regulate Distinct Metabolic Processes: Studies with Novel Mutant Mouse Models
- Author
-
Wess, Jürgen
- Subjects
obesity ,genetic structures ,QH301-705.5 ,Catalysis ,Receptors, G-Protein-Coupled ,Inorganic Chemistry ,Mice ,G protein-coupled receptors ,Diabetes Mellitus ,Animals ,Biology (General) ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,Spectroscopy ,β-arrestins ,diabetes ,Organic Chemistry ,General Medicine ,mutant mice ,beta-Arrestin 2 ,eye diseases ,Computer Science Applications ,Disease Models, Animal ,Chemistry ,Glucose ,beta-Arrestin 1 ,Perspective ,sense organs ,Energy Metabolism ,metabolism ,Signal Transduction - Abstract
The two β-arrestins (β-arrestin-1 and -2; alternative names: arrestin-2 and -3, respectively) are well known for their ability to inhibit signaling via G protein-coupled receptors. However, β-arrestins can also act as signaling molecules in their own right. Although the two proteins share a high degree of sequence and structural homology, early studies with cultured cells indicated that β-arrestin-1 and -2 are not functionally redundant. Recently, the in vivo metabolic roles of the two β-arrestins have been studied using mutant mice selectively lacking either β-arrestin-1 or -2 in cell types that are of particular relevance for regulating glucose and energy homeostasis. These studies demonstrated that the β-arrestin-1 and -2 mutant mice displayed distinct metabolic phenotypes in vivo, providing further evidence for the functional heterogeneity of these two highly versatile signaling proteins.
- Published
- 2022
75. Cannabinoid 2 receptor- and beta Arrestin 2-dependent upregulation of serotonin 2A receptors.
- Author
-
Franklin, J.M., Vasiljevik, T., Prisinzano, T.E., and Carrasco, G.A.
- Subjects
- *
CANNABINOID receptors , *ARRESTINS , *SEROTONIN receptors , *GENETIC regulation , *NEURAL transmission , *AFFECTIVE disorders - Abstract
Abstract: Recent evidence suggests that cannabinoid receptor agonists may regulate serotonin 2A (5-HT2A) receptor neurotransmission in the brain, although no molecular mechanism has been identified. Here, we present experimental evidence that sustained treatment with a non-selective cannabinoid agonist (CP55,940) or selective CB2 receptor agonists (JWH133 or GP1a) upregulate 5-HT2A receptors in a neuronal cell line. Furthermore, this cannabinoid receptor agonist-induced upregulation of 5-HT2A receptors was prevented in cells stably transfected with either CB2 or β-Arrestin 2 shRNA lentiviral particles. Additionally, inhibition of clathrin-mediated endocytosis also prevented the cannabinoid receptor-induced upregulation of 5-HT2A receptors. Our results indicate that cannabinoid agonists might upregulate 5-HT2A receptors by a mechanism that requires CB2 receptors and β-Arrestin 2 in cells that express both CB2 and 5-HT2A receptors. 5-HT2A receptors have been associated with several physiological functions and neuropsychiatric disorders such as stress response, anxiety and depression, and schizophrenia. Therefore, these results might provide a molecular mechanism by which activation of cannabinoid receptors might be relevant to some cognitive and mood disorders in humans. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
76. Angiotensin II activates NF-ĸB through AT1A receptor recruitment of β-arrestin in cultured rat vascular smooth muscle cells.
- Author
-
Morinelli, Thomas A., Mi-Hye Lee, Kendal, Ryan T., Luttrel, Louis M., Walker, Linda P., and Ullian, Michael E.
- Subjects
- *
VASCULAR smooth muscle physiology , *ANGIOTENSIN II , *NF-kappa B , *ARRESTINS , *PROTEIN receptors , *LABORATORY rats , *MUSCLE cells - Abstract
Activation of the angiotensin type 1A receptor (AT1AR) in rat aorta vascular smooth muscle cells (RASMC) results in increased synthesis of the proinflammatory enzyme cyclooxygenase-2 (COX-2). We previously showed that nuclear localization of internalized AT1AR results in activation of transcription of the gene for COX-2, i.e., prostaglandinendoperoxide synthase-2. Others have suggested that ANG II stimulation of COX-2 protein synthesis is mediated by NF-ĸB. The purpose of the present study was to examine the interrelationship between AT1AR activation, β-arrestin recruitment, and NF-ĸB activation in the ability of ANG II to increase COX-2 protein synthesis in RASMC. In the present study we utilized RASMC, inhibitors of the NF-ĸB pathway, β-arrestin knockdown, radioligand binding, immunoblotting, and immunofluorescence to characterize the roles of AT1AR internalization, NF-ĸB activation, and β-arrestin in ANG II-induced COX-2 synthesis. Ro-106-9920 or parthenolide, agents that inhibit the initial steps of NF-ĸB activation, blocked ANG II-induced p65 NF-ĸB nuclear localization, COX-2 protein expression, β-arrestin recruitment, and AT1AR internalization without inhibiting ANG II-induced p42/44 ERK activation. Curcumin, an inhibitor of NF-ĸB-induced transcription, blocked ANG II-induced COX-2 protein expression without altering AT1AR internalization, ANG II-induced p65 NF-ĸB nuclear localization, or p42/44 ERK activation. Small interfering RNA-induced knockdown of β-arrestin-1 and -2 inhibited ANG II-induced p65 NF-ĸB nuclear localization. In vascular smooth muscle cells, internalization of the activated AT1AR mediated by β-arrestins activates the NF-ĸB pathway, producing nuclear localization of the transcription factor and initiation of COX-2 protein synthesis, thereby linking internalization of the receptor with the NF-ĸB pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
77. Effects of the Ras homolog Rhes on Akt/protein kinase B and glycogen synthase kinase 3 phosphorylation in striatum
- Author
-
Harrison, L.M., Muller, S.H., and Spano, D.
- Subjects
- *
PROTEIN kinase B , *GLYCOGEN synthase kinase-3 , *PHOSPHORYLATION , *CORPUS striatum , *RAS proteins , *G protein coupled receptors , *CELLULAR signal transduction , *G proteins , *DOPAMINE receptors - Abstract
Abstract: G protein-coupled receptors (GPCR) signal not only through heterotrimeric G proteins, but also through alternate pathways. Thus, dopamine D2 receptors in the striatum signal through Gαi/o and also by promoting formation of a multi-protein complex containing β-arrestin2, protein phosphatase 2A (PP2A), and Akt in order to dephosphorylate Akt. Lithium, on the other hand, disrupts this complex to increase Akt phosphorylation. Rhes is a striatally enriched GTP-binding protein that has been shown to inhibit dopamine receptor-mediated behavior and signaling through heterotrimeric G proteins. Therefore, our objective was to test whether Rhes similarly affects signaling through the Akt/GSK3 pathway in the striatum. Rhes−/− mice showed basally increased Akt and GSK3β phosphorylation relative to rhes+/+ mice that was not further enhanced by lithium treatment. Furthermore, they responded to the D1/D2 agonist apomorphine with increased Akt and GSK3 phosphorylation. Co-immunoprecipitation experiments revealed that apomorphine treatment recruits PP 2A-C to Akt in both rhes+/+ and rhes−/− mice. Lithium did not disrupt their interaction in rhes−/− mice as there was little basal interaction. Rhes co-immunoprecipitated with β-arrestins, suggesting that it is integral to the multi-protein complex. Thus, Rhes is necessary for Akt dephosphorylation by the striatal multi-protein complex, and in its absence, a lithium-treated phenotype results. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
78. Roles of proteolysis in regulation of GPCR function.
- Author
-
Cottrell, GS
- Subjects
- *
PROTEOLYSIS , *G protein coupled receptors , *ENZYME activation , *PEPTIDASE , *PROTEIN-tyrosine kinases , *CELLULAR signal transduction - Abstract
The enzymatic activity of peptidases must be tightly regulated to prevent uncontrolled hydrolysis of peptide bonds, which could have devastating effects on biological systems. Peptidases are often generated as inactive propeptidases, secreted with endogenous inhibitors, or they are compartmentalized. Propeptidases become active after proteolytic removal of N-terminal activation peptides by other peptidases. Some peptidases only become active towards substrates only at certain p Hs, thus confining activity to specific compartments or conditions. This review discusses the different roles proteolysis plays in regulating GPCRs. At the cell-surface, certain GPCRs are regulated by the hydrolytic inactivation of bioactive peptides by membrane-anchored peptidases, which prevent signalling. Conversely, cell-surface peptidases can also generate bioactive peptides, which directly activate GPCRs. Alternatively, cell-surface peptidases activated by GPCRs, can generate bioactive peptides to cause transactivation of receptor tyrosine kinases, thereby promoting signalling. Certain peptidases can signal directly to cells, by cleaving GPCR to initiate intracellular signalling cascades. Intracellular peptidases also regulate GPCRs; lysosomal peptidases destroy GPCRs in lysosomes to permanently terminate signalling and mediate down-regulation; endosomal peptidases cleave internalized peptide agonists to regulate GPCR recycling, resensitization and signalling; and soluble intracellular peptidases also participate in GPCR function by regulating the ubiquitination state of GPCRs, thereby altering GPCR signalling and fate. Although the use of peptidase inhibitors has already brought success in the treatment of diseases such as hypertension, the discovery of new regulatory mechanisms involving proteolysis that control GPCRs may provide additional targets to modulate dysregulated GPCR signalling in disease. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
79. Involvement of β-arrestins in cancer progression.
- Author
-
Hu, Shanshan, Wang, Di, Wu, Jingjing, Jin, Juan, Wei, Wei, and Sun, Wuyi
- Abstract
β-arrestins, including β-arrestin1 and β-arrestin2, are ubiquitous cytosolic proteins which localize in the cytoplasm and plasma membrane, initially be regarded as an potential character in G protein-coupled receptors (GPCR) desensitization, sequestration, and internalization. Besides, recent many studies increasingly revealed that β-arrestins served widely as versatile adapter proteins for scaffolding many intracellular signaling networks to modulate the strength and duration of signaling by diverse types of receptors and downstream kinases. As we known, the biologic and clinical behaviors of many tumors are largely determined by multiple molecular signal pathways. More recently, accumulating evidences established that β-arrestins got widely involved in many cancer developmental signaling events which responsible for tumor viability and metastasis, suggesting an impressive role of β-arrestins in tumor progression. Because of the regulation and biological output of β-arrestins is so complex, the role of β-arrestins in cancer development still remains enigmatic. However, the further understanding with the clinical prognosis and oncogenic potential of β-arrestins might facilitate the identification of diagnosis biomarkers and development of drug targets in cancer. In this article, we reviewed a comprehensive summary of the β-arrestins-mediated functions in human cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
80. Melanocortin 5 receptor signaling and internalization: Role of MAPK/ERK pathway and β-arrestins 1/2
- Author
-
Rodrigues, Adriana R., Almeida, Henrique, and Gouveia, Alexandra M.
- Subjects
- *
MELANOCORTIN receptors , *CELLULAR signal transduction , *MITOGEN-activated protein kinases , *ARRESTINS , *G protein coupled receptors , *CELL membranes , *MSH (Hormone) - Abstract
Abstract: The Melanocortin 5 receptor (MC5R) is a G-protein coupled receptor (GPCR) that exhibits high affinity for α-MSH. Here we present evidence for MC5R-GFP internalization and subsequent recycling to cell surface, in α-MSH-stimulated HeLa cells. This melanocortin induces a biphasic activation of ERK1/2 with an early peak at 15min, a Gi-protein driven, β-arrestins 1/2 independent process, and a late sustained activation that is regulated by β-arrestins 1/2. ERK1/2 lead to downstream phosphorylation of 90-kDa ribosomal S6 kinases (p90RSK) and mitogen- and stress-activated protein kinase 1 (MSK1). Only a small fraction (10%) of phosphorylated p90RSK and ERK1/2 translocates to the nucleus inducing c-Fos expression. α-MSH also activates CREB through cAMP/PKA pathway. In 3T3-L1 adipocytes, where MC5R is endogenously expressed, α-MSH also induces phosphorylation and cytosolic retention of the same signaling molecules. These findings provide new evidence on the signaling mechanisms underlying MC5R biological response to α-MSH. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
81. Activated human hydroxy-carboxylic acid receptor-3 signals to MAP kinase cascades via the PLC-dependent PKC and MMP-mediated EGFR pathways.
- Author
-
Zhou, Q, Li, G, Deng, XY, He, XB, Chen, LJ, Wu, C, Shi, Y, Wu, KP, Mei, LJ, Lu, JX, and Zhou, NM
- Subjects
- *
HYDROXY carboxylic acids , *MITOGEN-activated protein kinases , *G protein coupled receptors , *ATHEROSCLEROSIS treatment , *PHOSPHOLIPASE C , *PROTEIN kinase C , *MATRIX metalloproteinases , *EPIDERMAL growth factor receptors - Abstract
BACKGROUND AND PURPOSE 3-Hydroxy-octanoate, recently identified as a ligand for, the orphan GPCR, HCA3, is of particular interest given its ability to treat lipid disorders and atherosclerosis. Here we demonstrate the pathway of HCA3-mediated activation of ERK1/2. EXPERIMENTAL APPROACH Using CHO-K1 cells stably expressing HCA3 receptors and A431 cells, a human epidermoid cell line with high levels of endogenous expression of functional HCA3 receptors, HCA3-mediated activation of ERK1/2 was measured by Western blot. KEY RESULTS HCA3-mediated activation of ERK1/2 was rapid, peaking at 5 min, and was Pertussis toxin sensitive. Our data, obtained by time course analyses in combination with different kinase inhibitors, demonstrated that on agonist stimulation, HCA3 receptors evoked ERK1/2 activation via two distinct pathways, the PLC/PKC pathway at early time points (≤2 min) and the MMP/ epidermal growth factor receptor (EGFR) transactivation pathway with a maximum response at 5 min. Furthermore, our present results also indicated that the βγ-subunits of the Gi protein play a critical role in HCA3-activated ERK1/2 phosphorylation, whereas β-arrestins and Src were not required for ERK1/2 activation. CONCLUSIONS AND IMPLICATIONS We have described the molecular mechanisms underlying the coupling of human HCA3 receptors to the ERK1/2 MAP kinase pathway in CHO-K1 and A431 cells, which implicate the Gi protein-initiated, PLC/PKC- and platelet-derived growth factor receptor/EGFR transactivation-dependent pathways. These observations may provide new insights into the pharmacological effects and the physiological functions modulated by the HCA3-mediated activation of ERK1/2. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
82. A Dopamine D1 Receptor-Dependent β-Arrestin Signaling Complex Potentially Regulates Morphine-Induced Psychomotor Activation but not Reward in Mice.
- Author
-
Urs, Nikhil M., Daigle, Tanya L., and Caron, Marc G.
- Subjects
- *
DOPAMINE receptors , *MORPHINE abuse , *DRUGS of abuse , *BRAIN stimulation , *LOCOMOTOR ataxia , *LABORATORY mice - Abstract
Morphine is a widely used analgesic in humans that is associated with multiple untoward effects, such as addiction and physical dependence. In rodent models, morphine also induces locomotor activity. These effects likely involve functionally selective mechanisms. Indeed, G protein-coupled receptor desensitization and adaptor protein β-arrestin 2 (βarr2) through its interaction with the μ-opioid receptor regulates the analgesic but not the rewarding properties of morphine. However, βarr2 is also required for morphine-induced locomotor activity in mice, but the exact cellular and molecular mechanisms that mediate this arrestin-dependent behavior are not understood. In this study, we show that βarr2 is required for morphine-induced locomotor activity in a dopamine D1 receptor (D1R)-dependent manner and that a βarr2/phospho-ERK (βarr2/pERK) signaling complex may mediate this behavior. Systemic administration of SL327, an MEK inhibitor, inhibits morphine-induced locomotion in wild-type mice in a dose-dependent manner. Acute morphine administration to mice promotes the formation of a βarr2/pERK signaling complex. Morphine-induced locomotor activity and formation of the βarr2/pERK signaling complex is blunted in D1R knockout (D1-KO) mice and is presumably independent of D2 dopamine receptors. However, D1Rs are not required for morphine-induced reward as D1-KO mice show the same conditioned place preference for morphine as do control mice. Taken together, these results suggest a potential role for a D1R-dependent βarr2/pERK signaling complex in selectively mediating the locomotor-stimulating but not the rewarding properties of morphine. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
83. GPCR signalling to the translation machinery
- Author
-
Musnier, Astrid, Blanchot, Benoît, Reiter, Eric, and Crépieux, Pascale
- Subjects
- *
G proteins , *CELLULAR signal transduction , *CELL differentiation , *PROTEIN-tyrosine kinases , *GENETIC translation , *MESSENGER RNA , *CELL metabolism , *EPIDERMAL growth factor - Abstract
Abstract: G protein-coupled receptors (GPCRs) are involved in most physiological processes, many of them being engaged in fully differentiated cells. These receptors couple to transducers of their own, primarily G proteins and β-arrestins, which launch intracellular signalling cascades. Some of these signalling events regulate the translational machinery to fine-tune general cell metabolism or to alter protein expression pattern. Though extensively documented for tyrosine kinase receptors, translational regulation by GPCRs is still poorly appreciated. The objective of this review paper is to address the following questions: i) is there a “GPCR signature” impacting on the translational machinery, and ultimately on the type of mRNA translated? ii) are the regulatory networks involved similar as those utilized by tyrosine kinase receptors? In particular, we will discuss the specific features of translational control mediated by GPCRs and highlight the intrinsic properties of GPCRs these mechanisms could rely on. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
84. Inhibition of EGF-induced ERK/MAP kinase-mediated astrocyte proliferation by μ opioids: integration of G protein and β-arrestin 2-dependent pathways.
- Author
-
Miyatake, Mayumi, Rubinstein, Tal J., McLennan, Gregory P., Belcheva, Mariana M., and Coscia, Carmine J.
- Subjects
- *
ASTROCYTES , *PROTEIN kinases , *G proteins , *OPIOID receptors , *EPIDERMAL growth factor , *PHOSPHORYLATION - Abstract
Although μ, κ, and δ opioids activate extracellular signal-regulated kinase (ERK)/mitogen-activated protein (MAP) kinase, the mechanisms involved in their signaling pathways and the cellular responses that ensue differ. Here we focused on the mechanisms by which μ opioids rapidly (min) activate ERK and their slower (h) actions to inhibit epidermal growth factor (EGF)-induced ERK-mediated astrocyte proliferation. The μ-opioid agonists ([d-ala2, mephe4, gly-ol5] enkephalin and morphine) promoted the phosphorylation of ERK/MAP kinase within 5 min via Gi/o protein, calmodulin (CaM), and β-arrestin2-dependent signaling pathways in immortalized and primary astrocytes. This was based on the attenuation of the μ-opioid activation of ERK by pertussis toxin (PTX), the CaM antagonist, W-7, and siRNA silencing of β-arrestin2. All three pathways were shown to activate ERK via an EGF receptor transactivation-mediated mechanism. This was disclosed by abolishment of μ-opioid-induced ERK phosphorylation with the EGF receptor-specific tyrosine phosphorylation inhibitor, AG1478, and μ-opioid-induced reduction of EGF receptor tyrosine phosphorylation by PTX, and β-arrestin2 targeting siRNA in the present studies and formerly by CaM antisense. Long-term (h) treatment of primary astrocytes with [d-ala2,mephe4,gly-ol5] enkephalin or morphine, attenuated EGF-induced ERK phosphorylation and proliferation (as measured by 5′-bromo-2′-deoxy-uridine labeling). PTX and β-arrestin2 siRNA but not W-7 reversed the μ-opioid inhibition. Unexpectedly, β-arrestin-2 siRNA diminished both EGF-induced ERK activation and primary astrocyte proliferation suggesting that this adaptor protein plays a novel role in EGF signaling as well as in the opioid receptor phase of this pathway. The results lend insight into the integration of the different μ-opioid signaling pathways to ERK and their cellular responses. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
85. β-Arrestins facilitate ubiquitin-dependent degradation of apoptosis signal-regulating kinase 1 (ASK1) and attenuate H2O2-induced apoptosis
- Author
-
Zhang, Zhengping, Hao, Jiaying, Zhao, Zhihui, Ben, Peiling, Fang, Fang, Shi, Lijun, Gao, Yanhong, Liu, Junhong, Wen, Chuanjun, Luo, Lan, and Yin, Zhimin
- Subjects
- *
CELLULAR signal transduction , *APOPTOSIS , *ENDOCYTOSIS , *JNK mitogen-activated protein kinases , *PROTEIN binding , *UBIQUITIN - Abstract
Abstract: β-Arrestins are ubiquitously expressed proteins that play important roles in receptor desensitization, endocytosis, proteosomal degradation, apoptosis and signaling. It has been reported that β-Arrestin2 acts as a scaffold by directly interacting with the JNK3 isoform and recruiting MKK4 and the apoptosis-signaling kinase-1 (ASK1). Here, we report a novel function of β-Arrestins in regulating H2O2-induced apoptosis. Our study demonstrates that β-Arrestins physically associate with C-terminal domain of ASK1, and moreover, both over-expression and RNA interference (RNAi) experiments indicate that β-Arrestins down-regulate ASK1 protein. In detail, β-Arrestin-induced reduction of ASK1 protein is due to ubiquitination and proteasome-dependent degradation of ASK1 in response to association of β-Arrestins and ASK1. Upon H2O2 stimulation, the protein binding between β-Arrestins and ASK1 increases and ASK1 degradation is expedited. In consequence, β-Arrestins prevent ASK1-JNK signaling and as a result attenuate H2O2-induced apoptosis. Structurally, C-terminal domain of ASK1 is essential for β-Arrestins and ASK1 association. We also found that CHIP is required for β-Arrestins-induced ASK1 degradation, which suggested that β-Arrestins function as a scaffold of ASK1 and CHIP, leading to CHIP-mediated ASK1 degradation. All these findings indicate that β-Arrestins play a negative regulatory role in H2O2-induced apoptosis signaling through associating with ASK1 and CHIP and facilitating ASK1 degradation, which provides a new insight for analyzing the effects of β-Arrestins on protecting cells from oxidative stress-induced apoptosis. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
86. Tolerance to the antinociceptive effects of peripherally administered opioids: Expression of β-arrestins
- Author
-
Hernández, Laura, Romero, Asunción, Almela, Pilar, García-Nogales, Paula, Laorden, M. Luisa, and Puig, Margarita M.
- Subjects
- *
DRUG tolerance , *OPIOID receptors , *ANALGESICS , *GENE expression , *MORPHINE , *INFLAMMATION , *PLACEBOS , *LABORATORY mice - Abstract
Abstract: Tolerance to peripheral antinociception after chronic exposure to systemic morphine was assessed in mice with chronic CFA-inflammation; cross-tolerance to locally administered μ, δ and κ-opioid agonists and levels of β-arrestins in the injured paw, were also evaluated. Tolerance was induced by the subcutaneous implantation of a 75 mg morphine-pellet, and antinociception evaluated with the Randall–Selitto test, 5 min after the subplantar injection of morphine, fentanyl, buprenorphine, DPDPE, U-50488H or CRF. Experiments were performed in the absence and presence of CFA-inflammation, in animals implanted with a morphine or placebo pellet. Beta-arrestin protein levels were determined by western blot. In mice without inflammation, subplantar opioids did not induce antinociception, while during CFA-inflammation, all drugs generated dose–response curves with an order of potency of: U-50488H < DPDPE < morphine < buprenorphine < fentanyl << CRF. During CFA-inflammation plus morphine-pellet, the potency of fentanyl decreased 1.25 times, while that of DPDPE, U-50488H and CRF diminished approximately 2.5–4.3 times. For each drug, the ratio between the ED50''s in tolerant and naive animals, was significantly higher than 1 (except for buprenorphine and fentanyl), demonstrating partial cross-tolerance to systemic morphine. Inflammation induced a twofold increase in β-arrestin expression (p <0.01), and the levels decreased after acute morphine exposure (p <0.05). Tolerance did not alter β-arrestins, but partially prevented the increase induced by inflammation. The results suggest that peripheral β-arrestins could facilitate peripheral OR-desensitization and tolerance development. Clinically, the experiments could be useful to establish the effectiveness of local opioid administration in patients with musculoskeletal pain, chronically receiving morphine analgesia. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
87. Kappa opioids promote the proliferation of astrocytes via Gβγ and β-arrestin 2-dependent MAPK-mediated pathways.
- Author
-
McLennan, Gregory P., Kiss, Alexi, Miyatake, Mayumi, Belcheva, Mariana M., Chambers, Kari T., Pozek, John J., Mohabbat, Yasmin, Moyer, Robert A., Bohn, Laura M., and Coscia, Carmine J.
- Subjects
- *
OPIOIDS , *ASTROCYTES , *OPIOID receptors , *MITOGEN-activated protein kinases , *G proteins , *PROTEIN kinases - Abstract
GTP binding regulatory protein (G protein)-coupled receptors can activate MAPK pathways via G protein-dependent and -independent mechanisms. However, the physiological outcomes correlated with the cellular signaling events are not as well characterized. In this study, we examine the involvement of G protein and β-arrestin 2 pathways in kappa opioid receptor-induced, extracellular signal-regulated kinase 1/2 (ERK1/2)-mediated proliferation of both immortalized and primary astrocyte cultures. As different agonists induce different cellular signaling pathways, we tested the prototypic kappa agonist, U69593 as well as the structurally distinct, non-nitrogenous agonist, C(2)-methoxymethyl salvinorin B (MOM-Sal-B). In immortalized astrocytes, U69593, activated ERK1/2 by a rapid (min) initial stimulation that was sustained over 2 h and increased proliferation. Sequestration of activated Gβγ subunits attenuated U69593 stimulation of ERK1/2 and suppressed proliferation in these cells. Furthermore, small interfering RNA silencing of β-arrestin 2 diminished sustained ERK activation induced by U69593. In contrast, MOM-Sal-B induced only the early phase of ERK1/2 phosphorylation and did not affect proliferation of immortalized astrocytes. In primary astrocytes, U69593 produced the same effects as seen in immortalized astrocytes. MOM-Sal-B elicited sustained ERK1/2 activation which was correlated with increased primary astrocyte proliferation. Proliferative actions of both agonists were abolished by either inhibition of ERK1/2, Gβγ subunits or β-arrestin 2, suggesting that both G protein-dependent and -independent ERK pathways are required for this outcome. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
88. Differences in motilin receptor desensitization after stimulation with motilin or motilides are due to alternative receptor trafficking
- Author
-
Mitselos, Anna, Berghe, Pieter Vanden, Peeters, Theo L., and Depoortere, Inge
- Subjects
- *
MOTILIN , *DESENSITIZATION (Psychotherapy) , *FLUORESCENCE microscopy , *INTRACELLULAR pathogens - Abstract
Abstract: Backgrounds & aims: The motilin receptor (MTLR) is an important therapeutic target for treatment of hypomotility disorders. The negative outcome in clinical trials with the motilin agonist, ABT-229, indicated that desensitization may limit the therapeutic usefulness of motilides. We therefore compared the mechanisms involved in the intracellular trafficking of the MTLR after stimulation with motilin, erythromycin-A (EM-A) or ABT-229. Methods: Desensitization was studied by measuring changes in Ca2+ rises and by receptor binding studies in CHO cells co-expressing the Ca2+ indicator apoaequorin and the MTLR, C-terminally tagged with EGFP. Receptor phosphorylation was studied by immunoprecipitation. MTLR-EGFP trafficking to organelles and translocation of β-arrestins were visualized by fluorescence microscopy. Results: Agonist-induced desensitization of the MTLR was due to receptor internalization with potencies (p-int50) in the order of: ABT-229 (8.3)>motilin (7.86)>EM-A (4.77) but with no differences in the internalization kinetics (t 1/2: ∼25min). The percentage cell surface receptor loss was more profound after exposure to ABT-229 (88±1%) than to motilin (63±10%) or EM-A (34±2%). For motilin and EM-A MTLR phosphorylation probably occurs via G protein-coupled receptor kinases while for ABT-229 phosphorylation was also protein kinase C dependent. All agonists translocated cytosolic β-arrestin-2 with greater affinity to the plasma membrane than β-arrestin-1. After internalization the MTLR co-localized with transferrin but not with cathepsin D. After stimulation with motilin and EM-A the t 1/2 for MTLR resensitization was 3h and 1h, respectively but amounted 26h for ABT-229. Conclusion: Our results suggest that the resensitization kinetics determine the desensitization properties of the motilin agonists. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
89. Internalization and desensitization of adenosine receptors.
- Author
-
Klaasse, Elisabeth, IJzerman, Adriaan, Grip, Willem, and Beukers, Margot
- Abstract
Until now, more than 800 distinct G protein-coupled receptors (GPCRs) have been identified in the human genome. The four subtypes of the adenosine receptor (A
1 , A2A , A2B and A3 receptor) belong to this large family of GPCRs that represent the most widely targeted pharmacological protein class. Since adenosine receptors are widespread throughout the body and involved in a variety of physiological processes and diseases, there is great interest in understanding how the different subtypes are regulated, as a basis for designing therapeutic drugs that either avoid or make use of this regulation. The major GPCR regulatory pathway involves phosphorylation of activated receptors by G protein-coupled receptor kinases (GRKs), a process that is followed by binding of arrestin proteins. This prevents receptors from activating downstream heterotrimeric G protein pathways, but at the same time allows activation of arrestin-dependent signalling pathways. Upon agonist treatment, adenosine receptor subtypes are differently regulated. For instance, the A1 Rs are not (readily) phosphorylated and internalize slowly, showing a typical half-life of several hours, whereas the A2A R and A2B R undergo much faster downregulation, usually shorter than 1 h. The A3 R is subject to even faster downregulation, often a matter of minutes. The fast desensitization of the A3 R after agonist exposure may be therapeutically equivalent to antagonist occupancy of the receptor. This review describes the process of desensitization and internalization of the different adenosine subtypes in cell systems, tissues and in vivo studies. In addition, molecular mechanisms involved in adenosine receptor desensitization are discussed. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
90. The role and mechanism of β-arrestins in cancer invasion and metastasis (Review)
- Author
-
Qi Li, Qing Ji, and Qing Song
- Subjects
0301 basic medicine ,Scaffold protein ,mechanism ,Apoptosis ,Biology ,Metastasis ,03 medical and health sciences ,Cell Movement ,Neoplasms ,Genetics ,medicine ,cancer ,metastasis ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Protein kinase A ,Protein kinase B ,beta-Arrestins ,G protein-coupled receptor ,β-arrestins ,Cell Proliferation ,Wnt signaling pathway ,Signal transducing adaptor protein ,General Medicine ,Articles ,medicine.disease ,invasion ,Cell biology ,030104 developmental biology ,Signal transduction ,Signal Transduction - Abstract
β‑arrestins are a family of adaptor proteins that regulate the signaling and trafficking of various G protein‑coupled receptors (GPCRs). They consist of β‑arrestin1 and β‑arrestin2 and are considered to be scaffolding proteins. β‑arrestins regulate cell proliferation, promote cell invasion and migration, transmit anti‑apoptotic survival signals and affect other characteristics of tumors, including tumor growth rate, angiogenesis, drug resistance, invasion and metastatic potential. It has been demonstrated that β‑arrestins serve roles in various physiological and pathological processes and exhibit a similar function to GPCRs. β‑arrestins serve primary roles in cancer invasion and metastasis via various signaling pathways. The present review assessed the function and mechanism of β‑arrestins in cancer invasion and metastasis via multiple signaling pathways, including mitogen‑activated protein kinase/extracellular signal regulated kinase, Wnt/β‑catenin, nuclear factor‑κB and phosphoinositide‑3 kinase/Akt.
- Published
- 2017
91. A synthetic intrabody-based selective and generic inhibitor of GPCR endocytosis
- Author
-
Punita Kumari, Eshan Ghosh, Prem N. Yadav, Sachdev S. Sidhu, Ashish Srivastava, Mithu Baidya, Ravi Ranjan, Arun K. Shukla, Hemlata Dwivedi, Akiko Koide, Shalini Dogra, Kumari Nidhi, and Shohei Koide
- Subjects
0301 basic medicine ,Phage display ,media_common.quotation_subject ,Allosteric regulation ,synthetic antibody fragments ,Biomedical Engineering ,Bioengineering ,Endocytosis ,Clathrin ,Article ,GPCRs ,Intrabody ,Receptors, G-Protein-Coupled ,Immunoglobulin Fab Fragments ,03 medical and health sciences ,ERK MAP kinase ,Phage Display ,Peptide Library ,Humans ,General Materials Science ,Electrical and Electronic Engineering ,Extracellular Signal-Regulated MAP Kinases ,Internalization ,β-arrestins ,G protein-coupled receptor ,media_common ,biology ,Chemistry ,intrabodies ,Condensed Matter Physics ,Atomic and Molecular Physics, and Optics ,Cell biology ,Synthetic antibody ,Enzyme Activation ,HEK293 Cells ,030104 developmental biology ,biology.protein ,cellular signaling ,Single-Chain Antibodies - Abstract
Beta-arrestins (βarrs) critically mediate desensitization, endocytosis and signalling of G protein-coupled receptors (GPCRs), and they scaffold a large number of interaction partners. However, allosteric modulation of their scaffolding abilities and direct targeting of their interaction interfaces to modulate GPCR functions selectively have not been fully explored yet. Here we identified a series of synthetic antibody fragments (Fabs) against different conformations of βarrs from phage display libraries. Several of these Fabs allosterically and selectively modulated the interaction of βarrs with clathrin and ERK MAP kinase. Interestingly, one of these Fabs selectively disrupted βarr–clathrin interaction, and when expressed as an intrabody, it robustly inhibited agonist-induced endocytosis of a broad set of GPCRs without affecting ERK MAP kinase activation. Our data therefore demonstrate the feasibility of selectively targeting βarr interactions using intrabodies and provide a novel framework for fine-tuning GPCR functions with potential therapeutic implications. A synthetic antibody fragment can disrupt the interaction of a GPCR regulatory protein β-arrestin with clathrin, and thereby inhibit induced internalization of GPCRs.
- Published
- 2017
92. Tracking the opioid receptors on the way of desensitization
- Author
-
Marie, Nicolas, Aguila, Benjamin, and Allouche, Stéphane
- Subjects
- *
OPIOID receptors , *CHEMICAL reactions , *G proteins , *MEMBRANE proteins - Abstract
Abstract: Opioid receptors belong to the super family of G-protein coupled receptors (GPCRs) and are the targets of numerous opioid analgesic drugs. Prolonged use of these drugs results in a reduction of their effectiveness in pain relief also called tolerance, a phenomenon well known by physicians. Opioid receptor desensitization is thought to play a major role in tolerance and a lot of work has been dedicated to elucidate the molecular basis of desensitization. As described for most of GPCRs, opioid receptor desensitization involves their phosphorylation by kinases and their uncoupling from G-proteins realized by arrestins. More recently, opioid receptor trafficking was shown to contribute to desensitization. In this review, our knowledge on the molecular mechanisms of desensitization and recent progress on the role of opioid receptor internalization, recycling or degradation in desensitization will be reported. A better understanding of these regulatory mechanisms would be helpful to develop new analgesic drugs or new strategies for pain treatment by limiting opioid receptor desensitization and tolerance. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
93. Changes in the expression of G protein-coupled receptor kinases and β-arrestins in mouse brain during cannabinoid tolerance.
- Author
-
Rubino, Tiziana, Viganò, Daniela, Premoli, Fabrizio, Castiglioni, Chiara, Bianchessi, Silvia, Zippel, Renata, and Parolaro, Daniela
- Abstract
The focus of our study was to determine the role of G protein-coupled receptor kinases (GRKs) and β-arrestins in agonist-induced CB1 receptor modulation during cannabinoid tolerance and their dependence from the extracellular signal-regulated kinase (ERK) cascade. In wild-type mice, chronic Δ
9 -tetrahydrocannabinol (THC) exposure significantly activated specific GRK and β-arrestin subunits in all the considered brain areas (striatum, cerebellum, hippocampus, and prefrontal cortex), suggesting their involvement in the adaptive processes underlying CB1 receptor downregulation and desensitization. These events were ERK-dependent in the striatum and cerebellum, because they were prevented in the genetic (Ras-GRF1 knockout mice) and pharmacological (SL327-pretreated mice) models of ERK activation inhibition, whereas in the hippocampus and prefrontal cortex, they appeared to be mostly ERK-independent. In the latter areas, ERK activation after chronic THC increased the transcription factors cyclic adenosine monophosphate response element-binding protein and Fos B as well as a downstream protein known as brain-derived neurotrophic factor. As a whole, our data suggest that in the striatum and cerebellum, THC-induced ERK activation could represent a key signaling event to initiate homologous desensitization of CB1 receptor, accounting for the development of tolerance to THC-induced hypolocomotion. In the prefrontal cortex and hippocampus, THC-induced alteration in GRKs and β-arrestins primarily depends on other kinases, whereas ERK activation could be part of the molecular adaptations that underlie the complex behavioral phenotype that defines the addicted state. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
94. G-protein-coupled receptor kinase activity in human heart failure: Effects of β-adrenoceptor blockade
- Author
-
Leineweber, Kirsten, Rohe, Patrick, Beilfuß, Anja, Wolf, Christina, Sporkmann, Heike, Bruck, Heike, Jakob, Heinz-Günther, Heusch, Gerd, Philipp, Thomas, and Brodde, Otto-Erich
- Subjects
- *
G proteins , *HEART failure , *ANIMALS , *ADRENERGIC receptors - Abstract
Abstract: Objectives: In human end-stage heart failure as well as in experimental animal models of heart failure, G-protein-coupled receptor kinase activity (GRK) is increased while β-adrenoceptor responsiveness is diminished. In animal studies, β-adrenoceptor blockers reverse the GRK-mediated desensitization and down-regulation of myocardial β-adrenoceptors. The aim of this study was to investigate whether alterations in GRK activity are an early or late accompaniment of human heart failure and whether also in humans β-adrenoceptor blocker treatment is able to influence myocardial GRK activity. Methods: We assessed in right atria, obtained from patients at different stages of heart failure, treated with or not treated with β-adrenoceptor blockers, and in the four chambers of explanted hearts, obtained from patients with end-stage heart failure, β-adrenoceptor density (by (-)-[125I]-iodocyanopindolol binding) and GRK activity (by an in vitro rhodopsin phosphorylation assay). Results: With increasing severity of heart failure, plasma noradrenaline levels increased while myocardial β-adrenoceptor density decreased with a maximum in GRK activity in end-stage heart failure. However, in relation to the progression of heart failure, we found that GRK activity transiently increased at an early stage of heart failure (NYHA I and II) but decreased back to control values in patients at NYHA III and IV. β-Adrenoceptor blockers were able to reduce the early increase in GRK activity at NYHA I and II to control levels, whereas in those patients who did not have increased GRK activity (NYHA III and IV), they had only a marginal effect. Conclusion: According to our results, an increase in GRK activity is an early and transient event in the course of heart failure that can be prevented by β-adrenoceptor blocker treatment. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
95. Regulated expression of G protein-coupled receptor kinases (GRK's) and beta-arrestins in osteoblasts.
- Author
-
Spurney, R. F.
- Subjects
- *
CELL culture , *PROTEINS , *CELLS , *ENZYMES , *TISSUES , *BIOMOLECULES - Abstract
Desensitization of G-protein coupled receptors (GPCR's) is largely mediated by a family of enzymes and protein co-factors termed GRKs and arrestins, respectively. In the present studies, we investigated expression of GRKs and arrestins in osteoblastic cell lines concentrating on the enzymes (GRK2 and GRK3) and protein co-factors (beta-arrestint 1 and beta-arrestin 2) that play dominant roles in regulating GPCR responsiveness in most tissues and cell types. We found that osteoblastic cells express similar amounts of GRK2 with either undetectable or lesser amounts of GRK3. In contrast, expression of beta-arrestin 1 and beta-arrestin 2 by osteoblastic cells varied between cell lines. To determine if GRK2 or beta-arrestin expression is modulated during osteoblast development, we assessed expression of GRK2 and beta-arrestin proteins during differentiation of the mouse osteoblastic cell line MC3T3-E1 cells over a 21-day period. We found that expression of GRK2 and beta-arrestin 2 increased to maximal levels by day 7 and then decreased 4-fold by day 21. In contrast, expression of beta-arrestin 1 increased to maximal levels by day 14 and then decreased 2-fold by day 21. Over this same time period (days 7-21), PTH/PTHrP receptor number decreased to a greater extent than the decrease in PTH(1-34)-induced cAMP generation, suggesting that responsiveness of individual PTH/PTHrP receptors was enhanced in differentiated cells. We conclude that (1) osteoblastic cell lines differentially express the enzymes and protein co-factors that modulate GPCR responsiveness and (2) expression of both GRK2 and beta-arrestins is temporally regulated during osteoblast development. These data are consistent with the notion that GPCR responsiveness may be differentially regulated in osteoblastic cell lines and during osteoblast development. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
96. WORKFLOW DESCRIPTION TO DYNAMICALLY MODEL β-ARRESTIN SIGNALING NETWORKS
- Author
-
Francesco De Pascali, Pascale Crépieux, Mohammed Akli Ayoub, Romain Yvinec, Anne Poupon, Eric Reiter, Physiologie de la reproduction et des comportements [Nouzilly] (PRC), Institut National de la Recherche Agronomique (INRA)-Institut Français du Cheval et de l'Equitation [Saumur]-Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS), United Arab Emirates University (UAEU), Centre National de la Recherche Scientifique (CNRS)-Université de Tours-Institut Français du Cheval et de l'Equitation [Saumur]-Institut National de la Recherche Agronomique (INRA), and Institut National de la Recherche Agronomique (INRA)-Institut Français du Cheval et de l'Equitation [Saumur]-Université de Tours-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Biological data ,model selection ,Computer science ,Distributed computing ,Energy transfer ,Model selection ,[MATH.MATH-DS]Mathematics [math]/Dynamical Systems [math.DS] ,data fitting ,System dynamics ,biochemical reaction network ,Dynamic models ,03 medical and health sciences ,parameter identification ,030104 developmental biology ,0302 clinical medicine ,Workflow ,Arrestin ,Receptor ,030217 neurology & neurosurgery ,Topology (chemistry) ,β-arrestins - Abstract
International audience; Dynamic models of signaling networks allow the formulation of hypotheses on the topology and kinetic rate laws characterizing a given molecular network, in-depth exploration and confrontation with kinetic biological data. Despite its standardization, dynamic modeling of signaling networks still requires successive technical steps that need to be carefully performed. Here, we detail these steps by going through the mathematical and statistical framework. We explain how it can be applied to the understanding of β-arrestin-dependent signaling networks. We illustrate our methodology through the modeling of β-arrestin recruitment kinetics at the Follicle Stimulating Hormone (FSH) receptor supported by in-house Bioluminescence Resonance Energy Transfer (BRET) data.
- Published
- 2019
97. Direct evidence that the GPCR CysLTR2 mutant causative of uveal melanoma is constitutively active with highly biased signaling
- Author
-
Jordan M. Mattheisen, Amanda R. Moore, Yu Chen, Ping Chi, Manija A. Kazmi, Tyler D. Hitchman, Mizuho Horioka, Thomas P. Sakmar, Emilie Ceraudo, and Thomas Huber
- Subjects
Uveal Neoplasms ,0301 basic medicine ,Receptors, Vasopressin ,Glutamine ,MV, missense variant ,Mutant ,HTRF, homogeneous time-resolved fluorescence immunoassay ,UVM, uveal melanoma ,Biochemistry ,Malignant transformation ,CysLTR2 ,Receptor ,Melanoma ,β-arrestins ,biased signaling ,CA, constitutive activity ,BRET, bioluminescent resonance energy transfer ,LTD4, leukotriene D4 ,beta-Arrestin 2 ,CAM, constitutively active mutant ,Cell biology ,Gene Expression Regulation, Neoplastic ,EPAC, exchange protein activated by cAMP ,LiCl, lithium chloride ,uveal melanoma ,Signal transduction ,signaling ,Protein Binding ,Signal Transduction ,Research Article ,PLCβ, phospholipase C-β ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,V2(A)6, hexa-alanine variant ,Biology ,Models, Biological ,RLuc3, Renilla Luciferase ,DPBS, Dulbecco’s PBS ,03 medical and health sciences ,FBS, fetal bovine serum ,Downregulation and upregulation ,Arrestin ,Humans ,BRET2, bioluminescent resonance energy transfer 2 ,constitutive activity ,Molecular Biology ,G protein-coupled receptor ,Receptors, Leukotriene ,CysLTR2, cysteinyl-leukotriene receptor 2 ,030102 biochemistry & molecular biology ,Lysine ,G protein ,Cell Biology ,CI, confidence interval ,Kinetics ,HEK293 Cells ,030104 developmental biology ,Cysteinyl leukotriene receptor 2 ,Amino Acid Substitution ,G protein–coupled receptor ,Mutation ,GTP-Binding Protein alpha Subunits, Gq-G11 ,GPCR, G protein–coupled receptor ,IP1, d-myo-inositol-1-phosphate ,BSA, bovine serum albumin - Abstract
Uveal melanoma is the most common eye cancer in adults and is clinically and genetically distinct from skin cutaneous melanoma. In a subset of cases, the oncogenic driver is an activating mutation in CYSLTR2, the gene encoding the G protein–coupled receptor cysteinyl-leukotriene receptor 2 (CysLTR2). The mutant CYSLTR2 encodes for the CysLTR2–L129Q receptor, with the substitution of Leu to Gln at position 129 (3.43). The ability of CysLTR2–L129Q to cause malignant transformation has been hypothesized to result from constitutive activity, but how the receptor could escape desensitization is unknown. Here, we characterize the functional properties of CysLTR2–L129Q. We show that CysLTR2–L129Q is a constitutively active mutant that strongly drives Gq/11 signaling pathways. However, CysLTR2–L129Q only poorly recruits β-arrestin. Using a modified Slack–Hall operational model, we quantified the constitutive activity for both pathways and conclude that CysLTR2–L129Q displays profound signaling bias for Gq/11 signaling pathways while escaping β-arrestin–mediated downregulation. CYSLTR2 is the first known example of a G protein–coupled receptor driver oncogene that encodes a highly biased constitutively active mutant receptor. These results provide new insights into the mechanism of CysLTR2–L129Q oncoprotein signaling and suggest CYSLTR2 as a promising potential therapeutic target in uveal melanoma.
- Published
- 2021
98. G protein-coupled receptors as therapeutic target.
- Author
-
Alcántara-Hernández R, Hernández-Espinosa DA, Medina LDC, and García-Sáinz JA
- Subjects
- Hormones, Humans, Ligands, Models, Molecular, Amino Acids, Receptors, G-Protein-Coupled
- Abstract
Receptors are proteins coded by DNA, some of which have already been crystalized, thus allowing the details of their structure at the atomic level and some aspects of their function to be known. This review focuses on the most diverse and abundant family of receptors, G protein-coupled receptors. This family of receptors recognizes and mediates the action of several endogenous ligands (hormones, neurotransmitters, growth factors and local hormones) and also intervenes in the pathogenesis of various diseases, which is why they are targeted by approximately 30 to 40% of medications that are used in daily clinical practice and of various illegal drugs as well. X-ray crystallography is one of the essential tools that has allowed to observe the structure of these receptors in the amino acids that participate in this interaction, which allows to know the binding site of the endogenous ligand and of synthetic molecules that act on them to modulate their action. Molecular modeling or "docking" is also a computational bioinformatics tool that supports research on receptor-ligand binding, which allows the design and development of increasingly specific drugs. These developments have brought along significant changes in fundamental pharmacodynamic concepts., (Copyright: © 2022 Permanyer.)
- Published
- 2022
- Full Text
- View/download PDF
99. Acute and chronic metabolic effects of carvedilol in high-fructose, high-fat diet-fed mice: implication of β-arrestin2 pathway.
- Author
-
Ahmed HMS, Mohamed SG, Ibrahim WS, Rezk AM, Mahmoud AAA, Mahmoud MF, and Ibrahim IAAE
- Subjects
- Animals, Dietary Carbohydrates administration & dosage, Diglycerides metabolism, Dyslipidemias metabolism, Fructose administration & dosage, Homeostasis drug effects, Lipid Metabolism drug effects, Liver metabolism, Male, Mice, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction physiology, Up-Regulation drug effects, Carvedilol administration & dosage, Carvedilol pharmacology, Diet, High-Fat adverse effects, Dietary Carbohydrates adverse effects, Dyslipidemias drug therapy, Dyslipidemias etiology, Fructose adverse effects, Glucose metabolism, Insulin Resistance physiology, Signal Transduction drug effects, Signal Transduction genetics, beta-Arrestin 2 metabolism
- Abstract
We aimed to investigate the acute and chronic effects of carvedilol on insulin resistance in high-fructose, high-fat diet (HFrHFD) - fed mice and the implication of the β-arrestin2 pathway. The acute effect of carvedilol (10 mg/kg, i.p.) on glucose tolerance and hepatic lipid signaling in normal and insulin resistant mice was investigated. Then, the chronic effect of carvedilol on insulin resistance and dyslipidemia in HFrHFD-fed mice was examined. Changes in β-arrestin2 and its downstream signals in liver, skeletal muscle, and adipose tissue were measured. This involved measuring phosphatidylinositol 4,5-bisphosphate (PIP2) and diacylglycerol (DAG) levels and protein kinase B (AKT) activity. Carvedilol acutely reduced fasting blood glucose levels in both normal and insulin resistant mice without significantly affecting the glucose tolerance. These acute effects were associated with increased hepatic PIP2 but decreased hepatic DAG levels. Chronic administration of carvedilol significantly ameliorated insulin resistance and dyslipidemia in HFrHFD-fed mice. These chronic effects were associated with increased β-arrestin2, PIP2, and AKT activity levels but decreased DAG levels in the classical insulin target tissues. In conclusion, carvedilol acutely maintains glucose homeostasis and chronically ameliorates insulin resistance and dyslipidemia in HFrHFD-fed mice. The insulin sensitizing effects of carvedilol are highly correlated with the upregulation of β-arrestin2 pathway.
- Published
- 2022
- Full Text
- View/download PDF
100. In-cellulo chemical cross-linking to visualize protein-protein interactions.
- Author
-
Saha S, Ranjan A, Godara M, and Shukla AK
- Subjects
- Immunoprecipitation, beta-Arrestins, Signal Transduction
- Abstract
Reversible protein-protein interaction in cells is an integral and central aspect of intracellular signaling mechanisms. This allows distinct signaling cascades to become active upon stimulation with external signal resulting in cellular and physiological responses. Several distinct methods are currently available and utilized routinely to monitor protein-protein interactions including co-immunoprecipitation (co-IP). An inherent limitation associated with co-IP assay however is the inability to efficiently capture transient and short-lived interactions in cells. Chemical cross-linking of such transient interactions in cellular context using cell permeable reagents followed by co-IP overcomes this limitation, and allows a simplified approach without requiring any sophisticated instrumentation. In this chapter, we present a step-by-step protocol for monitoring protein-protein interaction by combining chemical cross-linking and co-immunoprecipitation using GPCR-β-arrestin complex as a case example. This protocol is based on previously validated method that can potentially be adapted to capture and visualize transient protein-protein interactions in general., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.