101
|
Kim DK, Yun Y, Kim HR, Seo MD, Chung KY. Different conformational dynamics of various active states of β-arrestin1 analyzed by hydrogen/deuterium exchange mass spectrometry. J Struct Biol 2015; 190:250-9. [DOI: 10.1016/j.jsb.2015.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/26/2015] [Accepted: 04/07/2015] [Indexed: 01/14/2023]
|
102
|
Mörl K, Beck-Sickinger AG. Intracellular Trafficking of Neuropeptide Y Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:73-96. [PMID: 26055055 DOI: 10.1016/bs.pmbts.2015.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The multireceptor multiligand system of neuropeptide Y receptors and their ligands is involved in the regulation of a multitude of physiological and pathophysiological processes. Specific expression patterns, ligand-binding modes, and signaling properties contribute to the complex network regulating distinct cellular responses. Intracellular trafficking processes are important key steps that are regulated in context with accessory proteins. These proteins exert their influence by interacting directly or indirectly with the receptors, causing modification of the receptors, or operating as scaffolds for the assembly of larger signaling complexes. On the intracellular receptor faces, sequence-specific motifs have been identified that play an important role in this process. Interestingly, it is also possible to influence the receptor internalization by modification of the peptide ligand.
Collapse
Affiliation(s)
- Karin Mörl
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany.
| | - Annette G Beck-Sickinger
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
103
|
de Munnik SM, Smit MJ, Leurs R, Vischer HF. Modulation of cellular signaling by herpesvirus-encoded G protein-coupled receptors. Front Pharmacol 2015; 6:40. [PMID: 25805993 PMCID: PMC4353375 DOI: 10.3389/fphar.2015.00040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/12/2015] [Indexed: 12/22/2022] Open
Abstract
Human herpesviruses (HHVs) are widespread infectious pathogens that have been associated with proliferative and inflammatory diseases. During viral evolution, HHVs have pirated genes encoding viral G protein-coupled receptors (vGPCRs), which are expressed on infected host cells. These vGPCRs show highest homology to human chemokine receptors, which play a key role in the immune system. Importantly, vGPCRs have acquired unique properties such as constitutive activity and the ability to bind a broad range of human chemokines. This allows vGPCRs to hijack human proteins and modulate cellular signaling for the benefit of the virus, ultimately resulting in immune evasion and viral dissemination to establish a widespread and lifelong infection. Knowledge on the mechanisms by which herpesviruses reprogram cellular signaling might provide insight in the contribution of vGPCRs to viral survival and herpesvirus-associated pathologies.
Collapse
Affiliation(s)
- Sabrina M de Munnik
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| |
Collapse
|
104
|
Crudden C, Ilic M, Suleymanova N, Worrall C, Girnita A, Girnita L. The dichotomy of the Insulin-like growth factor 1 receptor: RTK and GPCR: friend or foe for cancer treatment? Growth Horm IGF Res 2015; 25:2-12. [PMID: 25466906 DOI: 10.1016/j.ghir.2014.10.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/06/2014] [Accepted: 10/20/2014] [Indexed: 01/14/2023]
Abstract
The prime position of the insulin-like growth factor 1 receptor (IGF-1R), at the head of the principle mitogenic and anti-apoptotic signalling cascades, along with the resilience to transformation of IGF-1R deficient cells fuelled great excitement for its anti-cancer targeting. Yet its potential has not been fulfilled, as clinical trial results fell far short of expectations. Advancements in understanding of other receptors' function have now begun to shed light on this incongruity, with the now apparent parallels highlighting the immaturity of our understanding of IGF-1R biology, with the model used for drug development now recognised as having been too simplistic. Gathering together the many advancements of the field of IGF-1R research over the past decade, alongside those in the GPCR field, advocates for a major paradigm shift in our appreciation of the subtle workings of this receptor. This review will emphasise the updating of the IGF-1R's classification from an RTK, to an RTK/GPCR functional hybrid, which integrates both canonical kinase signalling with many functions characteristic of a GPCR. Recognition of the shortcomings of IGF-1R inhibitor drug development programs and the models used not only allows us to reignite the initial interest in the IGF-1R as an anti-cancer therapeutic target, but also points to the possibility of biased ligand therapeutics, which together may hold a very powerful key to unlocking the true potential of IGF-1R modulation.
Collapse
Affiliation(s)
- Caitrin Crudden
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Marina Ilic
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Naida Suleymanova
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Claire Worrall
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Ada Girnita
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden; Dermatology Department, Karolinska University Hospital, Stockholm, Sweden
| | - Leonard Girnita
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden.
| |
Collapse
|
105
|
Morinelli TA, Walker LP, Velez JCQ, Ullian ME. Clathrin-dependent internalization of the angiotensin II AT₁A receptor links receptor internalization to COX-2 protein expression in rat aortic vascular smooth muscle cells. Eur J Pharmacol 2014; 748:143-8. [PMID: 25542758 DOI: 10.1016/j.ejphar.2014.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 01/14/2023]
Abstract
The major effects of Angiotensin II (AngII) in vascular tissue are mediated by AngII AT1A receptor activation. Certain effects initiated by AT1A receptor activation require receptor internalization. In rat aortic vascular smooth muscle cells (RASMC), AngII stimulates cyclooxygenase 2 protein expression. We have previously shown this is mediated by β-arrestin-dependent receptor internalization and NF-κB activation. In this study, a specific inhibitor of clathrin-mediated endocytosis (CME), pitstop-2, was used to test the hypothesis that clathrin-dependent internalization of activated AT1A receptor mediates NF-κB activation and subsequent cyclooxygenase 2 expression. Radioligand binding assays, real time qt-PCR and immunoblotting were used to document the effects of pitstop-2 on AngII binding and signaling in RASMC. Laser scanning confocal microscopy (LSCM) was used to image pitstop-2׳s effects on AT1 receptor/GFP internalization in HEK-293 cells and p65 NF-κB nuclear localization in RASMC. Pitstop-2 significantly inhibited internalization of AT1A receptor (44.7% ± 3.1% Control vs. 13.2% ± 8.3% Pitstop-2; n=3) as determined by radioligand binding studies in RASMC. Studies utilizing AT1A receptor/GFP expressed in HEK 293 cells and LSCM confirmed these findings. Pitstop-2 significantly inhibited AngII-induced p65 NF-κB phosphorylation and nuclear localization, COX-2 message and protein expression in RASMC without altering activation of p42/44 ERK or TNFα signaling. Pitstop-2, a specific inhibitor of clathrin-mediated endocytosis, confirms that internalization of activated AT1A receptor mediates AngII activation of cyclooxygenase 2 expression in RASMC. These data provide support for additional intracellular signaling pathways activated through β-arrestin mediated internalization of G protein-coupled receptors, such as AT1A receptors.
Collapse
Affiliation(s)
- Thomas A Morinelli
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, United States.
| | - Linda P Walker
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, United States
| | - Juan Carlos Q Velez
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| | - Michael E Ullian
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| |
Collapse
|
106
|
Evolutionary pattern in the OXT-OXTR system in primates: coevolution and positive selection footprints. Proc Natl Acad Sci U S A 2014; 112:88-93. [PMID: 25535371 DOI: 10.1073/pnas.1419399112] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Oxytocin is a nonapeptide involved in a wide range of physiologic and behavioral functions. Until recently, it was believed that an unmodified oxytocin sequence was present in all placental mammals. This study analyzed oxytocin (OXT) in 29 primate species and the oxytocin receptor (OXTR) in 21 of these species. We report here three novel OXT forms in the New World monkeys, as well as a more extensive distribution of a previously described variant (Leu8Pro). In structural terms, these OXTs share the same three low-energy conformations in solution during molecular dynamic simulations, with subtle differences in their side chains. A consistent signal of positive selection was detected in the Cebidae family, and OXT position 8 showed a statistically significant (P = 0.013) correlation with litter size. Several OXTR changes were identified, some of them promoting gain or loss of putative phosphorylation sites, with possible consequences for receptor internalization and desensitization. OXTR amino acid sites are under positive selection, and intramolecular and intermolecular coevolutionary processes with OXT were also detected. We suggest that some New World monkey OXT-OXTR forms can be correlated to male parental care through the increase of cross-reactivity with its correlated vasopressin system.
Collapse
|
107
|
Ostermaier MK, Schertler GFX, Standfuss J. Molecular mechanism of phosphorylation-dependent arrestin activation. Curr Opin Struct Biol 2014; 29:143-51. [PMID: 25484000 DOI: 10.1016/j.sbi.2014.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/16/2014] [Accepted: 07/18/2014] [Indexed: 12/31/2022]
Abstract
The past years have seen tremendous progress towards understanding how arrestins recognize phosphorylated G protein-coupled receptors (GPCRs). Two arrestin crystal structures, one of a pre-activated splice variant and one bound to a GPCR phosphopeptide, provided insights into the conformational changes upon phosphate recognition. Scanning mutagenesis and spectroscopic studies complete the picture of arrestin activation and receptor binding. Most perspicuous is the C-tail exchange mechanism, by which the C-tail of arrestin is released from its basal conformation and replaced by the phosphorylated GPCR C-terminus. Three positively charged clusters could act as conserved arrestin phosphosensors. Variations in the pattern of phosphorylation in a GPCR and variations within the C-terminus of different GPCRs may encode specificity to arrestin subtypes and particular physiological responses.
Collapse
Affiliation(s)
- Martin K Ostermaier
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Gebhard F X Schertler
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232 Villigen, Switzerland; Deparment of Biology, ETH Zurich, Wolfgang-Pauli-Str. 27, 8093 Zürich, Switzerland
| | - Joerg Standfuss
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232 Villigen, Switzerland.
| |
Collapse
|
108
|
Zindel D, Butcher AJ, Al-Sabah S, Lanzerstorfer P, Weghuber J, Tobin AB, Bünemann M, Krasel C. Engineered hyperphosphorylation of the β2-adrenoceptor prolongs arrestin-3 binding and induces arrestin internalization. Mol Pharmacol 2014; 87:349-62. [PMID: 25425623 DOI: 10.1124/mol.114.095422] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
G protein-coupled receptor phosphorylation plays a major role in receptor desensitization and arrestin binding. It is, however, unclear how distinct receptor phosphorylation patterns may influence arrestin binding and subsequent trafficking. Here we engineer phosphorylation sites into the C-terminal tail of the β2-adrenoceptor (β2AR) and demonstrate that this mutant, termed β2AR(SSS), showed increased isoprenaline-stimulated phosphorylation and differences in arrestin-3 affinity and trafficking. By measuring arrestin-3 recruitment and the stability of arrestin-3 receptor complexes in real time using fluorescence resonance energy transfer and fluorescence recovery after photobleaching, we demonstrate that arrestin-3 dissociated quickly and almost completely from the β2AR, whereas the interaction with β2AR(SSS) was 2- to 4-fold prolonged. In contrast, arrestin-3 interaction with a β2-adrenoceptor fused to the carboxyl-terminal tail of the vasopressin type 2 receptor was nearly irreversible. Further analysis of arrestin-3 localization revealed that by engineering phosphorylation sites into the β2-adrenoceptor the receptor showed prolonged interaction with arrestin-3 and colocalization with arrestin in endosomes after internalization. This is in contrast to the wild-type receptor that interacts transiently with arrestin-3 at the plasma membrane. Furthermore, β2AR(SSS) internalized more efficiently than the wild-type receptor, whereas recycling was very similar for both receptors. Thus, we show how the interaction between arrestins and receptors can be increased with minimal receptor modification and that relatively modest increases in receptor-arrestin affinity are sufficient to alter arrestin trafficking.
Collapse
Affiliation(s)
- Diana Zindel
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| | - Adrian J Butcher
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| | - Suleiman Al-Sabah
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| | - Peter Lanzerstorfer
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| | - Julian Weghuber
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| | - Andrew B Tobin
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| | - Moritz Bünemann
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| | - Cornelius Krasel
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany (D.Z., M.B., C.K.); MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.J.B., A.B.T.); Department of Pharmacology and Toxicology, Kuwait University, Kuwait (S.A.-S.); and University of Applied Sciences Upper Austria, Wels, Austria (P.L., J.W.)
| |
Collapse
|
109
|
Di Benedetto A, Sun L, Zambonin CG, Tamma R, Nico B, Calvano CD, Colaianni G, Ji Y, Mori G, Grano M, Lu P, Colucci S, Yuen T, New MI, Zallone A, Zaidi M. Osteoblast regulation via ligand-activated nuclear trafficking of the oxytocin receptor. Proc Natl Acad Sci U S A 2014; 111:16502-7. [PMID: 25378700 PMCID: PMC4246276 DOI: 10.1073/pnas.1419349111] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We report that oxytocin (Oxt) receptors (Oxtrs), on stimulation by the ligand Oxt, translocate into the nucleus of osteoblasts, implicating this process in the action of Oxt on osteoblast maturation. Sequential immunocytochemistry of intact cells or isolated nucleoplasts stripped of the outer nuclear membrane showed progressive nuclear localization of the Oxtr; this nuclear translocation was confirmed by monitoring the movement of Oxtr-EGFP as well as by immunogold labeling. Nuclear Oxtr localization was conclusively shown by Western immunoblotting and MS of nuclear lysate proteins. We found that the passage of Oxtrs into the nucleus was facilitated by successive interactions with β-arrestins (Arrbs), the small GTPase Rab5, importin-β (Kpnb1), and transportin-1 (Tnpo1). siRNA-mediated knockdown of Arrb1, Arrb2, or Tnpo1 abrogated Oxt-induced expression of the osteoblast differentiation genes osterix (Sp7), Atf4, bone sialoprotein (Ibsp), and osteocalcin (Bglap) without affecting Erk phosphorylation. Likewise and again, without affecting pErk, inhibiting Arrb recruitment by mutating Ser rich clusters of the nuclear localization signal to Ala abolished nuclear import and Oxtr-induced gene expression. These studies define a previously unidentified mechanism for Oxtr action on bone and open possibilities for direct transcriptional modulation by nuclear G protein-coupled receptors.
Collapse
Affiliation(s)
- Adriana Di Benedetto
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy; Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71122, Italy
| | - Li Sun
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Carlo G Zambonin
- Department of Chemistry, University of Bari Aldo Moro, Bari 70126, Italy; and
| | - Roberto Tamma
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Beatrice Nico
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Cosima D Calvano
- Department of Chemistry, University of Bari Aldo Moro, Bari 70126, Italy; and
| | - Graziana Colaianni
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Yaoting Ji
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71122, Italy
| | - Maria Grano
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Ping Lu
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Silvia Colucci
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Tony Yuen
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | | | - Alberta Zallone
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Mone Zaidi
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029; Structural and Chemical Biology, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
110
|
Balakumar P, Jagadeesh G. Structural determinants for binding, activation, and functional selectivity of the angiotensin AT1 receptor. J Mol Endocrinol 2014; 53:R71-92. [PMID: 25013233 DOI: 10.1530/jme-14-0125] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The renin-angiotensin system (RAS) plays an important role in the pathophysiology of cardiovascular disorders. Pharmacologic interventions targeting the RAS cascade have led to the discovery of renin inhibitors, angiotensin-converting enzyme inhibitors, and AT(1) receptor blockers (ARBs) to treat hypertension and some cardiovascular and renal disorders. Mutagenesis and modeling studies have revealed that differential functional outcomes are the results of multiple active states conformed by the AT(1) receptor upon interaction with angiotensin II (Ang II). The binding of agonist is dependent on both extracellular and intramembrane regions of the receptor molecule, and as a consequence occupies more extensive area of the receptor than a non-peptide antagonist. Both agonist and antagonist bind to the same intramembrane regions to interfere with each other's binding to exhibit competitive, surmountable interaction. The nature of interactions with the amino acids in the receptor is different for each of the ARBs given the small differences in the molecular structure between drugs. AT(1) receptors attain different conformation states after binding various Ang II analogues, resulting in variable responses through activation of multiple signaling pathways. These include both classical and non-classical pathways mediated through growth factor receptor transactivations, and provide cross-communication between downstream signaling molecules. The structural requirements for AT(1) receptors to activate extracellular signal-regulated kinases 1 and 2 through G proteins, or G protein-independently through β-arrestin, are different. We review the structural and functional characteristics of Ang II and its analogs and antagonists, and their interaction with amino acid residues in the AT(1) receptor.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Pharmacology UnitFaculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, MalaysiaDivision of Cardiovascular and Renal ProductsCenter for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Gowraganahalli Jagadeesh
- Pharmacology UnitFaculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, MalaysiaDivision of Cardiovascular and Renal ProductsCenter for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland 20993, USA
| |
Collapse
|
111
|
Chen X, Bai B, Tian Y, Du H, Chen J. Identification of serine 348 on the apelin receptor as a novel regulatory phosphorylation site in apelin-13-induced G protein-independent biased signaling. J Biol Chem 2014; 289:31173-87. [PMID: 25271156 DOI: 10.1074/jbc.m114.574020] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Phosphorylation plays vital roles in the regulation of G protein-coupled receptor (GPCR) functions. The apelin and apelin receptor (APJ) system is involved in the regulation of cardiovascular function and central control of body homeostasis. Here, using tandem mass spectrometry, we first identified phosphorylated serine residues in the C terminus of APJ. To determine the role of phosphorylation sites in APJ-mediated G protein-dependent and -independent signaling and function, we induced a mutation in the C-terminal serine residues and examined their effects on the interaction between APJ with G protein or GRK/β-arrestin and their downstream signaling. Mutation of serine 348 led to an elimination of both GRK and β-arrestin recruitment to APJ induced by apelin-13. Moreover, APJ internalization and G protein-independent ERK signaling were also abolished by point mutation at serine 348. In contrast, this mutant at serine residues had no demonstrable impact on apelin-13-induced G protein activation and its intracellular signaling. These findings suggest that mutation of serine 348 resulted in inactive GRK/β-arrestin. However, there was no change in the active G protein thus, APJ conformation was biased. These results provide important information on the molecular interplay and impact of the APJ function, which may be extrapolated to design novel drugs for cardiac hypertrophy based on this biased signal pathway.
Collapse
Affiliation(s)
- Xiaoyu Chen
- From the Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, People's Republic of China, the Department of physiology, Taishan Medical College, Taian, Shandong 271000, People's Republic of China
| | - Bo Bai
- the Neurobiology Institute, Jining Medical University, Jining, Shandong 272067, People's Republic of China,
| | - Yanjun Tian
- the Neurobiology Institute, Jining Medical University, Jining, Shandong 272067, People's Republic of China
| | - Hui Du
- the Department of physiology, Taishan Medical College, Taian, Shandong 271000, People's Republic of China
| | - Jing Chen
- the Neurobiology Institute, Jining Medical University, Jining, Shandong 272067, People's Republic of China, the Division of Translational and Systems Medicine, Warwick Medical School, University of Warwick, Coventry CV4 7AL, United Kingdom, and
| |
Collapse
|
112
|
Evron T, Peterson SM, Urs NM, Bai Y, Rochelle LK, Caron MG, Barak LS. G Protein and β-arrestin signaling bias at the ghrelin receptor. J Biol Chem 2014; 289:33442-55. [PMID: 25261469 DOI: 10.1074/jbc.m114.581397] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The G protein-coupled ghrelin receptor GHSR1a is a potential pharmacological target for treating obesity and addiction because of the critical role ghrelin plays in energy homeostasis and dopamine-dependent reward. GHSR1a enhances growth hormone release, appetite, and dopamine signaling through G(q/11), G(i/o), and G(12/13) as well as β-arrestin-based scaffolds. However, the contribution of individual G protein and β-arrestin pathways to the diverse physiological responses mediated by ghrelin remains unknown. To characterize whether a signaling bias occurs for GHSR1a, we investigated ghrelin signaling in a number of cell-based assays, including Ca(2+) mobilization, serum response factor response element, stress fiber formation, ERK1/2 phosphorylation, and β-arrestin translocation, utilizing intracellular second loop and C-tail mutants of GHSR1a. We observed that GHSR1a and β-arrestin rapidly form metastable plasma membrane complexes following exposure to an agonist, but replacement of the GHSR1a C-tail by the tail of the vasopressin 2 receptor greatly stabilizes them, producing complexes observable on the plasma membrane and also in endocytic vesicles. Mutations of the contiguous conserved amino acids Pro-148 and Leu-149 in the GHSR1a intracellular second loop generate receptors with a strong bias to G protein and β-arrestin, respectively, supporting a role for conformation-dependent signaling bias in the wild-type receptor. Our results demonstrate more balance in GHSR1a-mediated ERK signaling from G proteins and β-arrestin but uncover an important role for β-arrestin in RhoA activation and stress fiber formation. These findings suggest an avenue for modulating drug abuse-associated changes in synaptic plasticity via GHSR1a and indicate the development of GHSR1a-biased ligands as a promising strategy for selectively targeting downstream signaling events.
Collapse
Affiliation(s)
| | | | | | - Yushi Bai
- From the Departments of Cell Biology
| | | | - Marc G Caron
- From the Departments of Cell Biology, Neurobiology, and Medicine, Duke University, Medical Center, Durham, North Carolina 27710
| | | |
Collapse
|
113
|
Sood R, Minzel W, Rimon G, Tal S, Barki-Harrington L. Down-regulation of cyclooxygenase-2 by the carboxyl tail of the angiotensin II type 1 receptor. J Biol Chem 2014; 289:31473-9. [PMID: 25231994 DOI: 10.1074/jbc.m114.587576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The enzyme cyclooxygenase-2 (COX-2) plays an important role in the kidney by up-regulating the production of the vasoconstrictor hormone angiotensin II (AngII), which in turn down-regulates COX-2 expression via activation of the angiotensin II type 1 receptor (AT1) receptor. Chemical inhibition of the catalytic activity of COX-2 is a well-established strategy for treating inflammation but little is known of cellular mechanisms that dispose of the protein itself. Here we show that in addition to its indirect negative feedback on COX-2, AT1 also down-regulates the expression of the COX-2 protein via a pathway that does not involve G-protein or β-arrestin-dependent signaling. Instead, AT1 enhances the ubiquitination and subsequent degradation of the enzyme in the proteasome through elements in its cytosolic carboxyl tail (CT). We find that a mutant receptor that lacks the last 35 amino acids of its CT (Δ324) is devoid of its ability to reduce COX-2, and that expression of the CT sequence alone is sufficient to down-regulate COX-2. Collectively these results propose a new role for AT1 in regulating COX-2 expression in a mechanism that deviates from its canonical signaling pathways. Down-regulation of COX-2 by a short peptide that originates from AT1 may present as a basis for novel therapeutic means of eliminating excess COX-2 protein.
Collapse
Affiliation(s)
- Rapita Sood
- From the Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel 3498838 and
| | - Waleed Minzel
- From the Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel 3498838 and
| | - Gilad Rimon
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva 84105, Israel
| | - Sharon Tal
- From the Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel 3498838 and
| | - Liza Barki-Harrington
- From the Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel 3498838 and
| |
Collapse
|
114
|
Jaeger WC, Seeber RM, Eidne KA, Pfleger KDG. Molecular determinants of orexin receptor-arrestin-ubiquitin complex formation. Br J Pharmacol 2014; 171:364-74. [PMID: 24206104 PMCID: PMC3904257 DOI: 10.1111/bph.12481] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 09/11/2013] [Accepted: 10/15/2013] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE The orexin system regulates a multitude of key physiological processes, particularly involving maintenance of metabolic homeostasis. Consequently, there is considerable potential for pharmaceutical development for the treatment of disorders from narcolepsy to metabolic syndrome. It acts through the hormonal activity of two endogenous peptides, orexin A binding to orexin receptors 1 and 2 (OX₁ and OX₂) with similar affinity, and orexin B binding to OX₂ with higher affinity than OX₁ receptors. We have previously revealed data differentiating orexin receptor subtypes with respect to their relative stability in forming orexin receptor-arrestin-ubiquitin complexes measured by BRET. Recycling and cellular signalling distinctions were also observed. Here, we have investigated, using BRET, the molecular determinants involved in providing OX₂ receptors with greater β-arrestin-ubiquitin complex stability. EXPERIMENTAL APPROACH The contribution of the C-terminal tail of the OX receptors was investigated by bulk substitution and site-specific mutagenesis using BRET and inositol phosphate assays. KEY RESULTS Replacement of the OX₁ receptor C-terminus with that of the OX₂ receptor did not result in the expected gain of function, indicating a role for intracellular domain configuration in addition to primary structure. Furthermore, two out of the three putative serine/threonine clusters in the C-terminus were found to be involved in OX₂ receptor-β-arrestin-ubiquitin complex formation. CONCLUSIONS AND IMPLICATIONS This study provides fundamental insights into the molecular elements that influence receptor-arrestin-ubiquitin complex formation. Understanding how and why the orexin receptors can be functionally differentiated brings us closer to exploiting these receptors as drug targets.
Collapse
Affiliation(s)
- Werner C Jaeger
- Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research (WAIMR) and Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | | | | | | |
Collapse
|
115
|
Khoury E, Nikolajev L, Simaan M, Namkung Y, Laporte SA. Differential regulation of endosomal GPCR/β-arrestin complexes and trafficking by MAPK. J Biol Chem 2014; 289:23302-17. [PMID: 25016018 DOI: 10.1074/jbc.m114.568147] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
β-Arrestins are signaling adaptors that bind to agonist-occupied G protein-coupled receptors (GPCRs) and target them for endocytosis; however, the mechanisms regulating receptor/β-arrestin complexes and trafficking in endosomes, remain ill defined. Here we show, in live cells, differential dynamic regulation of endosomal bradykinin B2 receptor (B2R) complexes with either β-arrestin-1 or -2. We find a novel role for MAPK in the B2R/β-arrestin-2 complex formation, receptor trafficking and signaling mediated by an ERK1/2 regulatory motif in the hinge domain of the rat β-arrestin-2 (PET(178)P), but not rat β-arrestin-1 (PER(177)P). While the ERK1/2 regulatory motif is conserved between rat and mouse β-arrestin-2, it is surprisingly not conserved in human β-arrestin-2 (PEK(178)P). However, mutation of lysine 178 to threonine is sufficient to confer MAPK sensitivity to the human β-arrestin-2. Furthermore, substitution for a phosphomimetic residue in both the rat and the human β-arrestin-2 (T/K178D) significantly stabilizes B2R/β-arrestin complexes in endosomes, delays receptor recycling to the plasma membrane and maintains intracellular MAPK signaling. Similarly, the endosomal trafficking of β2-adrenergic, angiotensin II type 1 and vasopressin V2 receptors was altered by the β-arrestin-2 T178D mutant. Our findings unveil a novel subtype specific mode of MAPK-dependent regulation of β-arrestins in intracellular trafficking and signaling of GPCRs, and suggest differential endosomal receptor/β-arrestin-2 signaling roles among species.
Collapse
Affiliation(s)
| | | | | | | | - Stéphane A Laporte
- From the Departments of Medicine, Pharmacology and Therapeutics, and Anatomy and Cell Biology, McGill University Health Center Research Institute, McGill University, Strathcona Anatomy & Dentistry Bldg., Quebec H3A 2B2, Canada
| |
Collapse
|
116
|
Gatfield J, Monnier L, Studer R, Bolli MH, Steiner B, Nayler O. Sphingosine-1-phosphate (S1P) displays sustained S1P1 receptor agonism and signaling through S1P lyase-dependent receptor recycling. Cell Signal 2014; 26:1576-88. [DOI: 10.1016/j.cellsig.2014.03.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/26/2014] [Accepted: 03/30/2014] [Indexed: 10/25/2022]
|
117
|
Schuler B, Rieger G, Gubser M, Arras M, Gianella M, Vogel O, Jirkof P, Cesarovic N, Klohs J, Jakob P, Brock M, Gorr TA, Baum O, Hoppeler H, Samillan-Soto V, Gassmann M, Fischer JA, Born W, Vogel J. Endogenous α-calcitonin-gene-related peptide promotes exercise-induced, physiological heart hypertrophy in mice. Acta Physiol (Oxf) 2014; 211:107-21. [PMID: 24479375 DOI: 10.1111/apha.12244] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/10/2013] [Accepted: 01/24/2014] [Indexed: 11/29/2022]
Abstract
AIM It is unknown how the heart distinguishes various overloads, such as exercise or hypertension, causing either physiological or pathological hypertrophy. We hypothesize that alpha-calcitonin-gene-related peptide (αCGRP), known to be released from contracting skeletal muscles, is key at this remodelling. METHODS The hypertrophic effect of αCGRP was measured in vitro (cultured cardiac myocytes) and in vivo (magnetic resonance imaging) in mice. Exercise performance was assessed by determination of maximum oxygen consumption and time to exhaustion. Cardiac phenotype was defined by transcriptional analysis, cardiac histology and morphometry. Finally, we measured spontaneous activity, body fat content, blood volume, haemoglobin mass and skeletal muscle capillarization and fibre composition. RESULTS While αCGRP exposure yielded larger cultured cardiac myocytes, exercise-induced heart hypertrophy was completely abrogated by treatment with the peptide antagonist CGRP(8-37). Exercise performance was attenuated in αCGRP(-/-) mice or CGRP(8-37) treated wild-type mice but improved in animals with higher density of cardiac CGRP receptors (CLR-tg). Spontaneous activity, body fat content, blood volume, haemoglobin mass, muscle capillarization and fibre composition were unaffected, whereas heart index and ventricular myocyte volume were reduced in αCGRP(-/-) mice and elevated in CLR-tg. Transcriptional changes seen in αCGRP(-/-) (but not CLR-tg) hearts resembled maladaptive cardiac phenotype. CONCLUSIONS Alpha-calcitonin-gene-related peptide released by skeletal muscles during exercise is a hitherto unrecognized effector directing the strained heart into physiological instead of pathological adaptation. Thus, αCGRP agonists might be beneficial in heart failure patients.
Collapse
Affiliation(s)
- B. Schuler
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zürich; Zürich Switzerland
- Department of Physiology, Anatomy and Genetics; University of Oxford; Oxford UK
| | - G. Rieger
- Institute of Anatomy; University of Bern; Bern Switzerland
| | - M. Gubser
- Institute of Anatomy; University of Bern; Bern Switzerland
| | - M. Arras
- Division of Surgical Research; University Hospital Zürich; Zürich Switzerland
| | - M. Gianella
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zürich; Zürich Switzerland
| | - O. Vogel
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zürich; Zürich Switzerland
| | - P. Jirkof
- Division of Surgical Research; University Hospital Zürich; Zürich Switzerland
| | - N. Cesarovic
- Division of Surgical Research; University Hospital Zürich; Zürich Switzerland
| | - J. Klohs
- Institute for Biomedical Engineering; University of Zurich and Swiss Federal Institute of Technology; Zürich (ETHZ); Zürich Switzerland
| | - P. Jakob
- Institute of Physiology and Cardiovascular Research; University of Zürich; Zürich Switzerland
| | - M. Brock
- Division of Pulmonology; University Hospital Zürich; Zürich Switzerland
- Zürich Center for Integrative Human Physiology (ZIHP); Zürich Switzerland
| | - T. A. Gorr
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zürich; Zürich Switzerland
- Clinic IV; Division of Pediatric Hematology and Oncology; University Medical Center; Freiburg Germany
| | - O. Baum
- Institute of Anatomy; University of Bern; Bern Switzerland
| | - H. Hoppeler
- Institute of Anatomy; University of Bern; Bern Switzerland
| | - V. Samillan-Soto
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zürich; Zürich Switzerland
- Physiology Department; Medical School; Universidad Alas Peruanas; Lima Peru
| | - M. Gassmann
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zürich; Zürich Switzerland
- Zürich Center for Integrative Human Physiology (ZIHP); Zürich Switzerland
- Universidad Peruana Cayetano Heredia (UPCH); Lima Peru
| | - J. A. Fischer
- Former Research Laboratory for Calcium Metabolism; Orthopedic University Hospital Zürich; Zürich Switzerland
| | - W. Born
- Former Research Laboratory for Calcium Metabolism; Orthopedic University Hospital Zürich; Zürich Switzerland
| | - J. Vogel
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zürich; Zürich Switzerland
| |
Collapse
|
118
|
Tian X, Kang DS, Benovic JL. β-arrestins and G protein-coupled receptor trafficking. Handb Exp Pharmacol 2014; 219:173-86. [PMID: 24292830 DOI: 10.1007/978-3-642-41199-1_9] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nonvisual arrestins (β-arrestin-1 and β-arrestin-2) are adaptor proteins that function to regulate G protein-coupled receptor (GPCR) signaling and trafficking. β-arrestins are ubiquitously expressed and function to inhibit GPCR/G protein coupling, a process called desensitization, and promote GPCR trafficking and arrestin-mediated signaling. β-arrestin-mediated endocytosis of GPCRs requires the coordinated interaction of β-arrestins with clathrin, adaptor protein 2 (AP2), and phosphoinositides. These interactions are facilitated by a conformational change in β-arrestin that is thought to occur upon binding to a phosphorylated activated GPCR. In this review, we provide an overview of the key interactions involved in β-arrestin-mediated trafficking of GPCRs.
Collapse
Affiliation(s)
- Xufan Tian
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | | | | |
Collapse
|
119
|
Abstract
The two homologous mammalian proteins called β-arrestin1 (also known as arrestin2) and β-arrestin2 (also called arrestin3) are now widely accepted as endocytic and signaling adaptors for G protein-coupled receptors (GPCRs), growth factor receptors, and ion channels. The sustained interactions of β-arrestins with activated GPCRs have been shown to correlate with the agonist-induced ubiquitination on distinct domains in the β-arrestin molecule. Additionally, ubiquitination of β-arrestin promotes its interaction with proteins that mediate endocytosis (e.g., clathrin) and signaling (e.g., c-RAF). Recent studies have demonstrated that deubiquitination of β-arrestin by specific deubiquitinating enzymes (DUBs) acts as an important regulatory mechanism, which determines the stability of β-arrestin-GPCR binding and fine-tunes β-arrestin-dependent signaling to downstream kinases. Accordingly, ubiquitination/deubiquitination of β-arrestin can serve as an on/off switch for its signaling and endocytic functions. Moreover, by regulating the stability and localization of signalosomes, deubiquitination of β-arrestins by DUBs imparts spatial and temporal resolution in GPCR signaling.
Collapse
Affiliation(s)
- Sudha K Shenoy
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
120
|
Abstract
G-protein-coupled receptors (GPCRs) are the primary interaction partners for arrestins. The visual arrestins, arrestin1 and arrestin4, physiologically bind to only very few receptors, i.e., rhodopsin and the color opsins, respectively. In contrast, the ubiquitously expressed nonvisual variants β-arrestin1 and 2 bind to a large number of receptors in a fairly nonspecific manner. This binding requires two triggers, agonist activation and receptor phosphorylation by a G-protein-coupled receptor kinase (GRK). These two triggers are mediated by two different regions of the arrestins, the "phosphorylation sensor" in the core of the protein and a less well-defined "activation sensor." Binding appears to occur mostly in a 1:1 stoichiometry, involving the N-terminal domain of GPCRs, but in addition a second GPCR may loosely bind to the C-terminal domain when active receptors are abundant.Arrestin binding initially uncouples GPCRs from their G-proteins. It stabilizes receptors in an active conformation and also induces a conformational change in the arrestins that involves a rotation of the two domains relative to each other plus changes in the polar core. This conformational change appears to permit the interaction with further downstream proteins. The latter interaction, demonstrated mostly for β-arrestins, triggers receptor internalization as well as a number of nonclassical signaling pathways.Open questions concern the exact stoichiometry of the interaction, possible specificity with regard to the type of agonist and of GRK involved, selective regulation of downstream signaling (=biased signaling), and the options to use these mechanisms as therapeutic targets.
Collapse
Affiliation(s)
- Martin J Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Straße 9, 97078, Würzburg, Germany,
| | | |
Collapse
|
121
|
Snyder JC, Rochelle LK, Barak LS, Caron MG. The stem cell-expressed receptor Lgr5 possesses canonical and functionally active molecular determinants critical to β-arrestin-2 recruitment. PLoS One 2013; 8:e84476. [PMID: 24386388 PMCID: PMC3873998 DOI: 10.1371/journal.pone.0084476] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/14/2013] [Indexed: 01/08/2023] Open
Abstract
Lgr5 is a membrane protein related to G protein-coupled receptors (GPCR)s whose expression identifies stem cells in multiple tissues and is strongly correlated with cancer. Despite the recent identification of endogenous ligands for Lgr5, its mode of signaling remains enigmatic. The ability to couple to G proteins and βarrestins are classical molecular behaviors of GPCRs that have yet to be observed for Lgr5. Therefore, the goal of this study was to determine if Lgr5 can engage a classical GPCR behavior and elucidate the molecular determinants of this process. Structural analysis of Lgr5 revealed several motifs consistent with its ability to recruit βarr2. Among them, a "SSS" serine cluster located at amino acid position 873-875 within the C-terminal tail (C-tail), is in a region consistent with other GPCRs that bind βarr2 with high-affinity. To test its functionality, a ligand-independent βarr2 translocation assay was implemented. We show that Lgr5 recruits βarr2 and that the "SSS" amino acids (873-875) are absolutely critical to this process. We also demonstrate that for full efficacy, this cluster requires other Lgr5 C-tail serines that were previously shown to be important for constitutive and βarr2 independent internalization of Lgr5. These data are proof of principle that a classical GPCR behavior can be manifested by Lgr5. The existence of alternative ligands or missing effectors of Lgr5 that scaffold this classical GPCR behavior and the downstream signaling pathways engaged should be considered. Characterizing Lgr5 signaling will be invaluable for assessing its role in tissue maintenance, repair, and disease.
Collapse
Affiliation(s)
- Joshua C. Snyder
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Lauren K. Rochelle
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Larry S. Barak
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Marc G. Caron
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
122
|
Song YB, Park CO, Jeong JY, Huh WK. Monitoring G protein-coupled receptor activation using an adenovirus-based β-arrestin bimolecular fluorescence complementation assay. Anal Biochem 2013; 449:32-41. [PMID: 24361713 DOI: 10.1016/j.ab.2013.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 11/16/2013] [Accepted: 12/10/2013] [Indexed: 01/22/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell-surface receptors and are involved in a variety of pathological conditions including cancer and cardiovascular, metabolic, neurological, and autoimmune diseases. GPCRs are being intensively investigated as targets for therapeutic intervention, and the β-arrestin recruitment assay has become a popular tool for analyzing GPCR activation. Here, we report a high-throughput method for cloning GPCR cDNAs into adenoviral bimolecular fluorescence complementation (BiFC) vectors and performing the β-arrestin BiFC assay in cells transduced with recombinant adenoviruses. An analysis of the activation of somatostatin receptor 2 (SSTR2) with the adenovirus-based β-arrestin BiFC assay showed that the assay is suitable for quantifying SSTR2 activation in response to specific agonists or antagonists. Furthermore, the adenovirus-based β-arrestin BiFC assay was able to detect the activation of a broad range of GPCRs. Collectively, our data indicate that the adenovirus-based β-arrestin BiFC assay can serve as a simple and universal platform for studying GPCR activation and thus will be useful for high-throughput screening of drugs that target GPCRs.
Collapse
Affiliation(s)
- Yong Bhum Song
- Department of Biological Sciences and Research Center for Functional Cellulomics, Seoul National University, Seoul 151-747, Republic of Korea
| | - Chul O Park
- Department of Biological Sciences and Research Center for Functional Cellulomics, Seoul National University, Seoul 151-747, Republic of Korea
| | - Jae-Yeon Jeong
- Marine Biotechnology Research Division, Korea Institute of Ocean Science & Technology, Ansan 426-744, Republic of Korea.
| | - Won-Ki Huh
- Department of Biological Sciences and Research Center for Functional Cellulomics, Seoul National University, Seoul 151-747, Republic of Korea; Institute of Microbiology, Seoul National University, Seoul 151-747, Republic of Korea.
| |
Collapse
|
123
|
Sun WY, Song Y, Hu SS, Wang QT, Wu HX, Chen JY, Wei W. Depletion of β-arrestin2 in hepatic stellate cells reduces cell proliferation via ERK pathway. J Cell Biochem 2013. [PMID: 23192415 DOI: 10.1002/jcb.24458] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
β-Arrestins are multifunctional adaptor proteins. Recently, some new roles of β-arrestins in regulating intracellular signaling networks have been discovered, which regulate cell growth, proliferation, and apoptosis. Though, the role of β-arrestins expression in the pathology of hepatic fibrosis remains unclear. In this study, the possible relationship between the expression of β-arrestins with the experimental hepatic fibrosis and the proliferation of hepatic stellate cells (HSCs) were investigated. Porcine serum induced liver fibrosis was established in this study. At five time points, the dynamic expression of β-arrestin1, β-arrestin2, and α-smooth muscle actin (α-SMA) in rat liver tissues, was measured by immunohistochemical staining, double immunofluorescent staining, and Western blotting. This study showed that aggravation of hepatic fibrosis with gradually increasing expression of β-arrestin2 in the hepatic tissues, but not β-arrestin1. Further, as hepatic fibrosis worsens, β-arrestin2-expressing activated HSCs accounts for an increasingly larger percentage of all activated HSCs. And the expression of β-arrestin2 had a significant positive correlation with the expression of α-SMA, an activated HSCs marker. In vitro studies, the dynamic expression of β-arrestin1 and β-arrestin2 in platelet derived growth factor-BB (PDGF-BB) stimulated HSCs was assessed by Western blotting. The expression of β-arrestin2 was remarkably increased in PDGF-BB stimulated HSCs. Furthermore, the small interfering RNA (siRNA) technique was used to explore the effect of β-arrestins on the proliferation of HSCs and the activation of ERK1/2. Transfection of siRNA targeting β-arrestin2 mRNA (siβ-arrestin2) into HSCs led to a 68% and 70% reduction of β-arrestin2 mRNA and protein expression, respectively. siβ-arrestin2 abolished the effect of PDGF-BB on the proliferation of HSCs. In addition, siβ-arrestin2 exerted the inhibition of the activation of ERK1/2 in HSCs. The present study provided strong evidence for the participation of the β-arrestin2 in the pathogenesis of hepatic fibrosis. The β-arrestin2 depletion diminishes HSCs ERK1/2 signaling and proliferation stimulated by PDGF-BB. Selective targeting of β-arrestin2 inhibitors to HSCs might present as a novel strategy for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Engineering Technology Research Center of Anti-inflammatory and Immunodrugs in Anhui Province, Hefei, Anhui Province 230032, China
| | | | | | | | | | | | | |
Collapse
|
124
|
Zhao P, Canals M, Murphy JE, Klingler D, Eriksson EM, Pelayo JC, Hardt M, Bunnett NW, Poole DP. Agonist-biased trafficking of somatostatin receptor 2A in enteric neurons. J Biol Chem 2013; 288:25689-25700. [PMID: 23913690 DOI: 10.1074/jbc.m113.496414] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Somatostatin (SST) 14 and SST 28 activate somatostatin 2A receptors (SSTR2A) on enteric neurons to control gut functions. SST analogs are treatments of neuroendocrine and bleeding disorders, cancer, and diarrhea, with gastrointestinal side effects of constipation, abdominal pain, and nausea. How endogenous agonists and drugs differentially regulate neuronal SSTR2A is unexplored. We evaluated SSTR2A trafficking in murine myenteric neurons and neuroendocrine AtT-20 cells by microscopy and determined whether agonist degradation by endosomal endothelin-converting enzyme 1 (ECE-1) controls SSTR2A trafficking and association with β-arrestins, key regulators of receptors. SST-14, SST-28, and peptide analogs (octreotide, lanreotide, and vapreotide) stimulated clathrin- and dynamin-mediated internalization of SSTR2A, which colocalized with ECE-1 in endosomes and the Golgi. After incubation with SST-14, SSTR2A recycled to the plasma membrane, which required active ECE-1 and an intact Golgi. SSTR2A activated by SST-28, octreotide, lanreotide, or vapreotide was retained within the Golgi and did not recycle. Although ECE-1 rapidly degraded SST-14, SST-28 was resistant to degradation, and ECE-1 did not degrade SST analogs. SST-14 and SST-28 induced transient interactions between SSTR2A and β-arrestins that were stabilized by an ECE-1 inhibitor. Octreotide induced sustained SSTR2A/β-arrestin interactions that were not regulated by ECE-1. Thus, when activated by SST-14, SSTR2A internalizes and recycles via the Golgi, which requires ECE-1 degradation of SST-14 and receptor dissociation from β-arrestins. After activation by ECE-1-resistant SST-28 and analogs, SSTR2A remains in endosomes because of sustained β-arrestin interactions. Therapeutic SST analogs are ECE-1-resistant and retain SSTR2A in endosomes, which may explain their long-lasting actions.
Collapse
Affiliation(s)
- Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Meritxell Canals
- From the Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jane E Murphy
- the Department of Surgery, University of California, San Francisco, San Francisco, California 94143
| | - Diana Klingler
- the Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, Massachusetts 02142, and
| | - Emily M Eriksson
- the Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California 94110
| | - Juan-Carlos Pelayo
- the Department of Surgery, University of California, San Francisco, San Francisco, California 94143
| | - Markus Hardt
- the Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, Massachusetts 02142, and
| | - Nigel W Bunnett
- From the Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia,.
| | - Daniel P Poole
- From the Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia,.
| |
Collapse
|
125
|
Barak LS, Bai Y, Snyder JC, Wang J, Chen W, Caron MG. Triphenylmethane dye activation of beta-arrestin. Biochemistry 2013; 52:5403-14. [PMID: 23865508 PMCID: PMC3744129 DOI: 10.1021/bi400217r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
β-Arrestins regulate G protein-coupled receptor signaling as competitive inhibitors and protein adaptors. Low molecular weight biased ligands that bind receptors and discriminate between the G protein dependent arm and β-arrestin, clathrin-associated arm of receptor signaling are considered therapeutically valuable as a result of this distinctive pharmacological behavior. Other than receptor agonists, compounds that activate β-arrestins are not available. We show that within minutes of exposure to the cationic triphenylmethane dyes malachite green and brilliant green, tissue culture cells recruit β-arrestins to clathrin scaffolds in a receptor-activation independent manner. In the presence of these compounds, G protein signaling is inhibited, ERK and GSK3β signaling are preserved, and the recruitment of the beta2-adaptin, AP2 adaptor complex to clathrin as well as transferrin internalization is reduced. Moreover, malachite green binds β-arrestin2-GFP coated immunotrap beads relative to GFP only coated beads. Triphenylmethane dyes are FDA approved for topical use on newborns as components of triple-dye preparations and are not approved but used effectively as aqueous antibiotics in fish husbandry. As possible carcinogens, their chronic ingestion in food preparations, particularly through farmed fish, is discouraged in the U.S. and Europe. Our results indicate triphenylmethane dyes as a result of novel pharmacology may have additional roles as β-arrestin/clathrin pathway signaling modulators in both pharmacology research and clinical therapy.
Collapse
Affiliation(s)
- Larry S Barak
- Departments of Cell Biology, Duke University, Durham, NC 27710, USA.
| | | | | | | | | | | |
Collapse
|
126
|
Jensen DD, Godfrey CB, Niklas C, Canals M, Kocan M, Poole DP, Murphy JE, Alemi F, Cottrell GS, Korbmacher C, Lambert NA, Bunnett NW, Corvera CU. The bile acid receptor TGR5 does not interact with β-arrestins or traffic to endosomes but transmits sustained signals from plasma membrane rafts. J Biol Chem 2013; 288:22942-60. [PMID: 23818521 DOI: 10.1074/jbc.m113.455774] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
TGR5 is a G protein-coupled receptor that mediates bile acid (BA) effects on energy balance, inflammation, digestion, and sensation. The mechanisms and spatiotemporal control of TGR5 signaling are poorly understood. We investigated TGR5 signaling and trafficking in transfected HEK293 cells and colonocytes (NCM460) that endogenously express TGR5. BAs (deoxycholic acid (DCA), taurolithocholic acid) and the selective agonists oleanolic acid and 3-(2-chlorophenyl)-N-(4-chlorophenyl)-N, 5-dimethylisoxazole-4-carboxamide stimulated cAMP formation but did not induce TGR5 endocytosis or recruitment of β-arrestins, as assessed by confocal microscopy. DCA, taurolithocholic acid, and oleanolic acid did not stimulate TGR5 association with β-arrestin 1/2 or G protein-coupled receptor kinase (GRK) 2/5/6, as determined by bioluminescence resonance energy transfer. 3-(2-chlorophenyl)-N-(4-chlorophenyl)-N, 5-dimethylisoxazole-4-carboxamide stimulated a low level of TGR5 interaction with β-arrestin 2 and GRK2. DCA induced cAMP formation at the plasma membrane and cytosol, as determined using exchange factor directly regulated by cAMP (Epac2)-based reporters, but cAMP signals did not desensitize. AG1478, an inhibitor of epidermal growth factor receptor tyrosine kinase, the metalloprotease inhibitor batimastat, and methyl-β-cyclodextrin and filipin, which block lipid raft formation, prevented DCA stimulation of ERK1/2. Bioluminescence resonance energy transfer analysis revealed TGR5 and EGFR interactions that were blocked by disruption of lipid rafts. DCA stimulated TGR5 redistribution to plasma membrane microdomains, as localized by immunogold electron microscopy. Thus, TGR5 does not interact with β-arrestins, desensitize, or traffic to endosomes. TGR5 signals from plasma membrane rafts that facilitate EGFR interaction and transactivation. An understanding of the spatiotemporal control of TGR5 signaling provides insights into the actions of BAs and therapeutic TGR5 agonists/antagonists.
Collapse
Affiliation(s)
- Dane D Jensen
- Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Ma WJ, Hashii M, Munesue T, Hayashi K, Yagi K, Yamagishi M, Higashida H, Yokoyama S. Non-synonymous single-nucleotide variations of the human oxytocin receptor gene and autism spectrum disorders: a case-control study in a Japanese population and functional analysis. Mol Autism 2013; 4:22. [PMID: 23815867 PMCID: PMC3707786 DOI: 10.1186/2040-2392-4-22] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 06/18/2013] [Indexed: 12/21/2022] Open
Abstract
Background The human oxytocin receptor (hOXTR) is implicated in the etiology of autism spectrum disorders (ASDs) and is a potential target for therapeutic intervention. Several studies have reported single-nucleotide polymorphisms (SNPs) of the OXTR gene associated with ASDs. These SNPs, however, reside outside the protein-coding region. Not much is known about genetic variations that cause amino acid substitutions that alter receptor functions. Methods Variations in the OXTR gene were analyzed in 132 ASD patients at Kanazawa University Hospital in Japan and 248 unrelated healthy Japanese volunteers by re-sequencing and real-time polymerase chain reaction-based genotyping. Functional changes in variant OXTRs were assessed by radioligand binding assay and measurements of intracellular free calcium concentrations ([Ca2+]i) and inositol 1,4,5-trisphosphate (IP3) levels. Results Six subjects (4.5%) in the ASD group and two in the control group (0.8%) were identified as heterozygotes carrying the R376G variation (rs35062132; c.1126C>G); one individual from the ASD group (0.8%) and three members of the control group (1.2%) were found to be carrying R376C (c.1126C>T). The C/G genotype significantly correlated with an increased risk of ASDs (odds ratio (OR) = 5.83; 95% confidence interval (CI) = 1.16 to 29.33; P = 0.024, Fisher’s exact test). Consistently, the G allele showed a correlation with an increased likelihood of ASDs (OR = 5.73; 95% CI = 1.15 to 28.61; P = 0.024, Fisher’s exact test). The frequencies of the C/T genotype and the T allele in the ASD and control groups did not differ significantly. We also examined changes in agonist-induced cellular responses mediated by the variant receptors hOXTR-376G and hOXTR-376C. OXT-induced receptor internalization and recycling were faster in hOXTR-376G-expressing HEK-293 cells than in cells expressing hOXTR-376R or hOXTR-376C. In addition, the elevation in [Ca2+]i and IP3 formation decreased in the cells expressing hOXTR-376G and hOXTR-376C tagged with enhanced green fluorescent protein (EGFP), in comparison with the cells expressing the common-type hOXTR-376R tagged with EGFP. Conclusions These results suggest that the rare genetic variation rs35062132 might contribute to the pathogenesis of ASDs, and could provide a molecular basis of individual differences in OXTR-mediated modulation of social behavior.
Collapse
Affiliation(s)
- Wen-Jie Ma
- Department of Biophysical Genetics, Kanazawa University Graduate School of Medicine, 13-1 Takara-machi, Kanazawa 920-8640, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Hauger RL, Olivares-Reyes JA, Braun S, Hernandez-Aranda J, Hudson CC, Gutknecht E, Dautzenberg FM, Oakley RH. Desensitization of human CRF2(a) receptor signaling governed by agonist potency and βarrestin2 recruitment. ACTA ACUST UNITED AC 2013; 186:62-76. [PMID: 23820308 DOI: 10.1016/j.regpep.2013.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 04/18/2013] [Accepted: 06/20/2013] [Indexed: 01/04/2023]
Abstract
The primary goal was to determine agonist-specific regulation of CRF2(a) receptor function. Exposure of human retinoblastoma Y79 cells to selective (UCN2, UCN3 or stresscopins) and non-selective (UCN1 or sauvagine) agonists prominently desensitized CRF2(a) receptors in a rapid, concentration-dependent manner. A considerably slower rate and smaller magnitude of desensitization developed in response to the weak agonist CRF. CRF1 receptor desensitization stimulated by CRF, cortagine or stressin1-A had no effect on CRF2(a) receptor cyclic AMP signaling. Conversely, desensitization of CRF2(a) receptors by UCN2 or UCN3 did not cross-desensitize Gs-coupled CRF1 receptor signaling. In transfected HEK293 cells, activation of CRF2(a) receptors by UCN2, UCN3 or CRF resulted in receptor phosphorylation and internalization proportional to agonist potency. Neither protein kinase A nor casein kinases mediated CRF2(a) receptor phosphorylation or desensitization. Exposure of HEK293 or U2OS cells to UCN2 or UCN3 (100nM) produced strong βarrestin2 translocation and colocalization with membrane CRF2(a) receptors while CRF (1μM) generated only weak βarrestin2 recruitment. βarrestin2 did not internalize with the receptor, however, indicating that transient CRF2(a) receptor-arrestin complexes dissociate at or near the cell membrane. Since deletion of the βarrestin2 gene upregulated Gs-coupled CRF2(a) receptor signaling in MEF cells, a βarrestin2 mechanism restrains Gs-coupled CRF2(a) receptor signaling activated by urocortins. We further conclude that the rate and extent of homologous CRF2(a) receptor desensitization are governed by agonist-specific mechanisms affecting GRK phosphorylation, βarrestin2 recruitment, and internalization thereby producing unique signal transduction profiles that differentially affect the stress response.
Collapse
Affiliation(s)
- Richard L Hauger
- Center of Excellence for Stress and Mental Health, San Diego VA Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA; Department of Psychiatry, School of Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA.
| | | | | | | | | | | | | | | |
Collapse
|
129
|
Gyombolai P, Boros E, Hunyady L, Turu G. Differential β-arrestin2 requirements for constitutive and agonist-induced internalization of the CB1 cannabinoid receptor. Mol Cell Endocrinol 2013; 372:116-27. [PMID: 23541635 DOI: 10.1016/j.mce.2013.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/06/2013] [Accepted: 03/15/2013] [Indexed: 01/13/2023]
Abstract
CB1 cannabinoid receptor (CB1R) undergoes both constitutive and agonist-induced internalization, but the underlying mechanisms of these processes and the role of β-arrestins in the regulation of CB1R function are not completely understood. In this study, we followed CB1R internalization using confocal microscopy and bioluminescence resonance energy transfer measurements in HeLa and Neuro-2a cells. We found that upon activation CB1R binds β-arrestin2 (β-arr2), but not β-arrestin1. Furthermore, both the expression of dominant-negative β-arr2 (β-arr2-V54D) and siRNA-mediated knock-down of β-arr2 impaired the agonist-induced internalization of CB1R. In contrast, neither β-arr2-V54D nor β-arr2-specific siRNA had a significant effect on the constitutive internalization of CB1R. However, both constitutive and agonist-induced internalization of CB1R were impaired by siRNA-mediated depletion of clathrin heavy chain. We conclude that although clathrin is required for both constitutive and agonist-stimulated internalization of CB1R, β-arr2 binding is only required for agonist-induced internalization of the receptor suggesting that the molecular mechanisms underlying constitutive and agonist-induced internalization of CB1R are different.
Collapse
Affiliation(s)
- Pál Gyombolai
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| | | | | | | |
Collapse
|
130
|
Grotegut CA, Gunatilake RP, Feng L, Heine RP, Murtha AP. The influence of maternal body mass index on myometrial oxytocin receptor expression in pregnancy. Reprod Sci 2013; 20:1471-7. [PMID: 23653389 DOI: 10.1177/1933719113488446] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Obese pregnant women have higher rates of dysfunctional labor patterns, need for oxytocin augmentation, labor induction, postdates pregnancy, and cesarean delivery compared to normal weight pregnant women. We tested the hypothesis that myometrial oxytocin receptor (OXTR) gene and protein expression are affected by obesity in pregnancy. Myometrial samples were obtained at the time of cesarean delivery from the upper aspect of the uterine hysterotomy incision and processed for real-time quantitative polymerase chain reaction and Western blot. There were 63 myometrial samples available for analysis. The median body mass index (BMI) at delivery was 31.0 kg/m(2) (interquartile range, 26.0, 40.0 kg/m(2)), and the median gestational age at delivery was 38.0 weeks (interquartile range, 33.0, 39.1 weeks). The OXTR gene expression did not correlate with maternal BMI at delivery by linear regression, and the median OXTR gene expression did not differ between women with a BMI ≤ 30 kg/m(2) and those with a BMI ≥ 40 kg/m(2). The OXTR protein expression was also not affected by maternal BMI. Myometrial OXTR gene expression appears to be independent of BMI at the time of delivery. Dysfunctional labor patterns and increased oxytocin utilization seen in obese women may not be due to differences in OXTR expression, though functional studies are required.
Collapse
Affiliation(s)
- Chad A Grotegut
- 1Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | | | | | | | | |
Collapse
|
131
|
Snyder JC, Rochelle LK, Lyerly HK, Caron MG, Barak LS. Constitutive internalization of the leucine-rich G protein-coupled receptor-5 (LGR5) to the trans-Golgi network. J Biol Chem 2013; 288:10286-97. [PMID: 23439653 DOI: 10.1074/jbc.m112.447540] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
LGR5 is a Wnt pathway associated G protein-coupled receptor (GPCR) that serves as a molecular determinant of stem cells in numerous tissues including the intestine, stomach, hair follicle, eye, and mammary gland. Despite its importance as a marker for this critical niche, little is known about LGR5 signaling nor the biochemical mechanisms and receptor determinants that regulate LGR5 membrane expression and intracellular trafficking. Most importantly, in cells LGR5 is predominantly intracellular, yet the mechanisms underlying this behavior have not been determined. In this work we elucidate a precise trafficking program for LGR5 and identify the motif at its C terminus that is responsible for the observed constitutive internalization. We show that this process is dependent upon dynamin GTPase activity and find that wild-type full-length LGR5 rapidly internalizes into EEA1- and Rab5-positive endosomes. However, LGR5 fails to rapidly recycle to the plasmid membrane through Rab4-positive vesicles, as is common for other GPCRs. Rather, internalized LGR5 transits through Rab7- and Rab9-positive vesicles, co-localizes in vesicles with Vps26, a retromer complex component that regulates retrograde trafficking to the trans-Golgi network (TGN) and reaches a steady-state distribution in the TGN within 2 h. Using mutagenesis, particularly of putative phosphorylation sites, we show that the amino acid pair, serine 861 and 864, is the principal C-tail determinant that mediates LGR5 constitutive internalization. The constitutive internalization of LGR5 to the TGN suggests the existence of novel biochemical roles for its Wnt pathway related, but ill defined signaling program.
Collapse
Affiliation(s)
- Joshua C Snyder
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
132
|
Gimenez LED, Ghildyal P, Fischer KE, Hu H, Ja WW, Eaton BA, Wu Y, Austad SN, Ranjan R. Modulation of methuselah expression targeted to Drosophila insulin-producing cells extends life and enhances oxidative stress resistance. Aging Cell 2013; 12:121-9. [PMID: 23121290 DOI: 10.1111/acel.12027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2012] [Indexed: 01/14/2023] Open
Abstract
Ubiquitously reduced signaling via Methuselah (MTH), a G-protein-coupled receptor (GPCR) required for neurosecretion, has previously been reported to extend life and enhance stress resistance in flies. Whether these effects are due to reduced MTH signalling in specific tissues remains unknown. We determined that reduced expression of mth targeted to the insulin-producing cells (IPCs) of the fly brain was sufficient to extend life and enhance oxidative stress resistance. Paradoxically, we discovered that overexpression of mth targeted to the same cells has similar phenotypic effects to reduced expression due to MTH's interaction with β-arrestin, which uncouples GPCRs from their G-proteins. We confirmed the functional relationship between MTH and β-arrestin by finding that IPC-targeted overexpression of β-arrestin alone mimics the longevity phenotype of reduced MTH signaling. As reduced MTH signaling also inhibits insulin secretion from the IPCs, the most parsimonious mechanistic explanation of its longevity and stress-resistance enhancement might be through reduced insulin/IGF signaling (IIS). However, examination of phenotypic features of long-lived IPC-mth modulated flies as well as several downstream IIS targets implicates enhanced activity of the JNK stress-resistance pathway more directly than insulin signaling in the longevity and stress-resistance phenotypes.
Collapse
Affiliation(s)
- Luis E. D. Gimenez
- Department of Pharmacology; University of Texas Health Science Center San Antonio; San Antonio; TX; 78245; USA
| | - Parakashtha Ghildyal
- Department of Pharmacology; University of Texas Health Science Center San Antonio; San Antonio; TX; 78245; USA
| | - Kathleen E. Fischer
- Department of Physiology; University of Texas Health Science Center San Antonio; San Antonio; TX; 78229; USA
| | - Hongxiang Hu
- Department of Pharmacology; University of Texas Health Science Center San Antonio; San Antonio; TX; 78245; USA
| | - William W. Ja
- Department of Metabolism & Aging; The Scripps Research Institute; Jupiter; FL; 33458; USA
| | - Benjamin A. Eaton
- Department of Physiology; University of Texas Health Science Center San Antonio; San Antonio; TX; 78229; USA
| | - Yimin Wu
- Department of Physiology; University of Texas Health Science Center San Antonio; San Antonio; TX; 78229; USA
| | | | | |
Collapse
|
133
|
β-Arrestins in the Central Nervous System. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:267-95. [DOI: 10.1016/b978-0-12-394440-5.00011-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
134
|
Hattori M, Tanaka M, Takakura H, Aoki K, Miura K, Anzai T, Ozawa T. Analysis of temporal patterns of GPCR–β-arrestin interactions using split luciferase-fragment complementation. MOLECULAR BIOSYSTEMS 2013; 9:957-64. [DOI: 10.1039/c2mb25443c] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
135
|
Walther C, Ferguson SSG. Arrestins: role in the desensitization, sequestration, and vesicular trafficking of G protein-coupled receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:93-113. [PMID: 23764051 DOI: 10.1016/b978-0-12-394440-5.00004-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the years, β-arrestins have emerged as multifunctional molecular scaffolding proteins regulating almost every imaginable G protein-coupled receptor (GPCR) function. Originally discovered as GPCR-desensitizing molecules, they have been shown to also serve as important regulators of GPCR signaling, sequestration, and vesicular trafficking. This broad functional role implicates β-arrestins as key regulatory proteins for cellular function. Hence, this chapter summarizes the current understanding of the β-arrestin family's unique ability to control the kinetics as well as the extent of GPCR activity at the level of desensitization, sequestration, and subsequent intracellular trafficking.
Collapse
Affiliation(s)
- Cornelia Walther
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, Western University Canada, London, Ontario, Canada
| | | |
Collapse
|
136
|
Abstract
Arrestins belong to a family of multifunctional adaptor proteins that regulate internalization of diverse receptors including G-protein-coupled receptors (GPCRs). Defects associated with endocytosis of GPCRs have been linked to human diseases. We used enhanced green fluorescent protein-tagged arrestin 2 (Arr2) to monitor the turnover of the major rhodopsin (Rh1) in live Drosophila. We demonstrate that during degeneration of norpA(P24) photoreceptors the loss of Rh1 is parallel to the disappearance of rhabdomeres, the specialized visual organelle that houses Rh1. The cause of degeneration in norpA(P24) is the failure to activate CaMKII (Ca(2+)/calmodulin-dependent protein kinase II) and retinal degeneration C (RDGC) because of a loss of light-dependent Ca(2+) entry. A lack of activation in CaMKII, which phosphorylates Arr2, leads to hypophosphorylated Arr2, while a lack of activation of RDGC, which dephosphorylates Rh1, results in hyperphosphorylated Rh1. We investigated how reversible phosphorylation of Rh1 and Arr2 contributes to photoreceptor degeneration. To uncover the consequence underlying a lack of CaMKII activation, we characterized ala(1) flies in which CaMKII was suppressed by an inhibitory peptide, and showed that morphology of rhabdomeres was not affected. In contrast, we found that expression of phosphorylation-deficient Rh1s, which either lack the C terminus or contain Ala substitution in the phosphorylation sites, was able to prevent degeneration of norpA(P24) photoreceptors. This suppression is not due to a loss of Arr2 interaction. Importantly, co-expression of these modified Rh1s offered protective effects, which greatly delayed photoreceptor degeneration. Together, we conclude that phosphorylation of Rh1 is the major determinant that orchestrates its internalization leading to retinal degeneration.
Collapse
|
137
|
Scholten DJ, Canals M, Wijtmans M, de Munnik S, Nguyen P, Verzijl D, de Esch IJP, Vischer HF, Smit MJ, Leurs R. Pharmacological characterization of a small-molecule agonist for the chemokine receptor CXCR3. Br J Pharmacol 2012; 166:898-911. [PMID: 21883151 DOI: 10.1111/j.1476-5381.2011.01648.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The chemokine receptor CXCR3 is a GPCR found predominantly on activated T cells. CXCR3 is activated by three endogenous peptides; CXCL9, CXCL10 and CXCL11. Recently, a small-molecule agonist, VUF10661, has been reported in the literature and synthesized in our laboratory. The aim of the present study was to provide a detailed pharmacological characterization of VUF10661 by comparing its effects with those of CXCL11. EXPERIMENTAL APPROACH Agonistic properties of VUF10661 were assessed in a chemotaxis assay with murine L1.2 cells transiently transfected with cDNA encoding the human CXCR3 receptor and in binding studies, with [(125)I]-CXCL10 and [(125)I]-CXCL11, on membrane preparations from HEK293 cells stably expressing CXCR3. [(35)S]-GTPγS binding was used to determine its potency to induce CXCR3-mediated G protein activation and BRET-based assays to investigate its effects on intracellular cAMP levels and β-arrestin recruitment. KEY RESULTS VUF10661 acted as a partial agonist in CXCR3-mediated chemotaxis, bound to CXCR3 in an allosteric fashion in ligand binding assays and activated G(i) proteins with the same efficacy as CXCL11 in the [(35)S]-GTPγS binding and cAMP assay, while it recruited more β-arrestin1 and β-arrestin2 to CXCR3 receptors than the chemokine. CONCLUSIONS AND IMPLICATIONS VUF10661, like CXCL11, activates both G protein-dependent and -independent signalling via the CXCR3 receptor, but probably exerts its effects from an allosteric binding site that is different from that for CXCL11. It could stabilize different receptor and/or β-arrestin conformations leading to differences in functional output. Such ligand-biased signalling might offer interesting options for the therapeutic use of CXCR3 agonists.
Collapse
Affiliation(s)
- D J Scholten
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Li G, Zhou Q, Yu Y, Chen L, Shi Y, Luo J, Benovic J, Lu J, Zhou N. Identification and characterization of distinct C-terminal domains of the human hydroxycarboxylic acid receptor-2 that are essential for receptor export, constitutive activity, desensitization, and internalization. Mol Pharmacol 2012; 82:1150-61. [PMID: 22962331 DOI: 10.1124/mol.112.081307] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The human hydroxycarboxylic acid receptor 2 (HCA₂), also known as GPR109A and HM74a, was first identified as a niacin receptor and has recently received significant attention because of its potential to clinically modify plasma lipids in a favorable manner. Our recent studies have demonstrated that the niacin-induced internalization of HCA₂ receptors is regulated by G protein-coupled receptor kinase (GRK) 2 and arrestin3 and that internalized receptors rapidly recycle back to the cell surface. The investigation presented here used a combination of amino acid deletion and site-directed mutagenesis to identify structural and functional domains within the HCA₂ C terminus and explore their potential roles in receptor phosphorylation, desensitization, and internalization. We first constructed four mutants with deletions of 10 to 15 amino acids each that were distinct from truncated mutants. We successfully identified different domains responsible for receptor export, constitutive activity, desensitization, phosphorylation, and internalization. We also generated a comprehensive series of alanine substitution mutants, replacing conserved serine and threonine residues in the C terminus with alanine residues to pinpoint the key residues that are essential for GRK2-mediated phosphorylation and arrestin3 association. Moreover, we found that a sequence from residues 329 to 343 in the C-terminal tail of HCA₂ plays a crucial role in keeping HCA₂ in an inactive conformation. These data demonstrate the importance of distinct domains within the C terminus of HCA₂ for receptor cell surface expression, desensitization, and internalization and phosphorylation and stabilization of an inactive receptor conformation.
Collapse
Affiliation(s)
- Guo Li
- College of Life Sciences, Zhejiang University, Zijingang Campus, Hangzhou Zhejinag, China
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Heitzler D, Durand G, Gallay N, Rizk A, Ahn S, Kim J, Violin JD, Dupuy L, Gauthier C, Piketty V, Crépieux P, Poupon A, Clément F, Fages F, Lefkowitz RJ, Reiter E. Competing G protein-coupled receptor kinases balance G protein and β-arrestin signaling. Mol Syst Biol 2012; 8:590. [PMID: 22735336 PMCID: PMC3397412 DOI: 10.1038/msb.2012.22] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 05/23/2012] [Indexed: 01/14/2023] Open
Abstract
The molecular mechanisms and hidden dynamics governing ERK activation by the angiotensin II type 1A receptor are studied and deciphered, revealing a signal balancing mechanism that is found to be relevant to a range of other seven transmembrane receptors. ![]()
An ODE-based dynamical model of ERK activation by the prototypical angiotensin II type-1A seven transmembrane receptor has been built and validated. In order to deal with a limited number of experimental read-outs, unknown parameters have been inferred by simultaneously fitting control and perturbed conditions. In addition to its well-established function in G-protein uncoupling, G protein-coupled receptor kinase 2 has been shown to exert a strong negative effect on β-arrestin-dependent signaling and by doing so, to balance G-protein and β-arrestin signaling. This novel function of G protein-coupled receptor kinase 2 has also been evidenced in primary vascular smooth muscle cells naturally expressing the AT1AR and in HEK293 cells expressing other 7TMRs.
Seven-transmembrane receptors (7TMRs) are involved in nearly all aspects of chemical communications and represent major drug targets. 7TMRs transmit their signals not only via heterotrimeric G proteins but also through β-arrestins, whose recruitment to the activated receptor is regulated by G protein-coupled receptor kinases (GRKs). In this paper, we combined experimental approaches with computational modeling to decipher the molecular mechanisms as well as the hidden dynamics governing extracellular signal-regulated kinase (ERK) activation by the angiotensin II type 1A receptor (AT1AR) in human embryonic kidney (HEK)293 cells. We built an abstracted ordinary differential equations (ODE)-based model that captured the available knowledge and experimental data. We inferred the unknown parameters by simultaneously fitting experimental data generated in both control and perturbed conditions. We demonstrate that, in addition to its well-established function in the desensitization of G-protein activation, GRK2 exerts a strong negative effect on β-arrestin-dependent signaling through its competition with GRK5 and 6 for receptor phosphorylation. Importantly, we experimentally confirmed the validity of this novel GRK2-dependent mechanism in both primary vascular smooth muscle cells naturally expressing the AT1AR, and HEK293 cells expressing other 7TMRs.
Collapse
Affiliation(s)
- Domitille Heitzler
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Mustafa S, See HB, Seeber RM, Armstrong SP, White CW, Ventura S, Ayoub MA, Pfleger KDG. Identification and profiling of novel α1A-adrenoceptor-CXC chemokine receptor 2 heteromer. J Biol Chem 2012; 287:12952-65. [PMID: 22371491 PMCID: PMC3340001 DOI: 10.1074/jbc.m111.322834] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/20/2012] [Indexed: 01/14/2023] Open
Abstract
We have provided the first evidence for specific heteromerization between the α(1A)-adrenoceptor (α(1A)AR) and CXC chemokine receptor 2 (CXCR2) in live cells. α(1A)AR and CXCR2 are both expressed in areas such as the stromal smooth muscle layer of the prostate. By utilizing the G protein-coupled receptor (GPCR) heteromer identification technology on the live cell-based bioluminescence resonance energy transfer (BRET) assay platform, our studies in human embryonic kidney 293 cells have identified norepinephrine-dependent β-arrestin recruitment that was in turn dependent upon co-expression of α(1A)AR with CXCR2. These findings have been supported by co-localization observed using confocal microscopy. This norepinephrine-dependent β-arrestin recruitment was inhibited not only by the α(1)AR antagonist Terazosin but also by the CXCR2-specific allosteric inverse agonist SB265610. Furthermore, Labetalol, which is marketed for hypertension as a nonselective β-adrenoceptor antagonist with α(1)AR antagonist properties, was identified as a heteromer-specific-biased agonist exhibiting partial agonism for inositol phosphate production but essentially full agonism for β-arrestin recruitment at the α(1A)AR-CXCR2 heteromer. Finally, bioluminescence resonance energy transfer studies with both receptors tagged suggest that α(1A)AR-CXCR2 heteromerization occurs constitutively and is not modulated by ligand. These findings support the concept of GPCR heteromer complexes exhibiting distinct pharmacology, thereby providing additional mechanisms through which GPCRs can potentially achieve their diverse biological functions. This has important implications for the use and future development of pharmaceuticals targeting these receptors.
Collapse
MESH Headings
- Adrenergic alpha-1 Receptor Antagonists/pharmacology
- Adrenergic alpha-Agonists/pharmacology
- Allosteric Regulation/physiology
- Animals
- Arrestins/metabolism
- CHO Cells
- Chemokines/metabolism
- Cricetinae
- HEK293 Cells
- Humans
- Inositol Phosphates/metabolism
- Labetalol/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Norepinephrine/pharmacology
- Prazosin/analogs & derivatives
- Prazosin/pharmacology
- Prostate/metabolism
- Protein Structure, Quaternary
- Receptors, Adrenergic, alpha-1/chemistry
- Receptors, Adrenergic, alpha-1/metabolism
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Interleukin-8B/chemistry
- Receptors, Interleukin-8B/metabolism
- beta-Arrestins
Collapse
Affiliation(s)
- Sanam Mustafa
- From the Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia 6009
| | - Heng B. See
- From the Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia 6009
| | - Ruth M. Seeber
- From the Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia 6009
| | - Stephen P. Armstrong
- From the Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia 6009
| | - Carl W. White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052 and
| | - Sabatino Ventura
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052 and
| | - Mohammed Akli Ayoub
- From the Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia 6009
| | - Kevin D. G. Pfleger
- From the Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia 6009
- Dimerix Bioscience Pty Ltd, Nedlands, Perth, Western Australia 6009, Australia
| |
Collapse
|
141
|
Engagement of β-arrestin by transactivated insulin-like growth factor receptor is needed for V2 vasopressin receptor-stimulated ERK1/2 activation. Proc Natl Acad Sci U S A 2012; 109:E1028-37. [PMID: 22493236 DOI: 10.1073/pnas.1112422109] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have been shown to activate the mitogen-activated protein kinases, ERK1/2, through both G protein-dependent and -independent mechanisms. Here, we describe a G protein-independent mechanism that unravels an unanticipated role for β-arrestins. Stimulation of the V2 vasopressin receptor (V2R) in cultured cells or in vivo in rat kidney medullar collecting ducts led to the activation of ERK1/2 through the metalloproteinase-mediated shedding of a factor activating the insulin-like growth factor receptor (IGFR). This process was found to be both Src- and β-arrestin-dependent. Whereas Src was found to act upstream of the metalloproteinase activation and be required for the release of the IGFR-activating factor, β-arrestins were found to act downstream of the IGFR transactivation. Unexpectedly, the engagement of β-arrestins by the IGFR but not by the V2R was needed to promote the vasopressin-stimulated ERK1/2 activation, indicating that a pool of β-arrestins distinct from those β-arrestins recruited to the V2R acts downstream of the receptor tyrosine kinase to activate ERK1/2. Such a dual site of action for β-arrestins helps explain the pleiotropic actions of this scaffolding protein. Given the role that V2R-stimulated ERK1/2 plays in kidney cell proliferation, this transactivation mechanism may have important implications for renal pathophysiology. Still, the role of β-arrestins downstream of a transactivation event is not limited to the V2R, because we observed a similar involvement for an unrelated GPCR (the platelet-activating factor receptor), indicating that it may be a general mechanism shared among GPCRs.
Collapse
|
142
|
Canals M, Scholten DJ, de Munnik S, Han MKL, Smit MJ, Leurs R. Ubiquitination of CXCR7 controls receptor trafficking. PLoS One 2012; 7:e34192. [PMID: 22457824 PMCID: PMC3311620 DOI: 10.1371/journal.pone.0034192] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 02/28/2012] [Indexed: 12/21/2022] Open
Abstract
The chemokine receptor CXCR7 binds CXCL11 and CXCL12 with high affinity, chemokines that were previously thought to bind exclusively to CXCR4 and CXCR3, respectively. Expression of CXCR7 has been associated with cardiac development as well as with tumor growth and progression. Despite having all the canonical features of G protein-coupled receptors (GPCRs), the signalling pathways following CXCR7 activation remain controversial, since unlike typical chemokine receptors, CXCR7 fails to activate Gαi-proteins. CXCR7 has recently been shown to interact with β-arrestins and such interaction has been suggested to be responsible for G protein-independent signals through ERK-1/2 phosphorylation. Signal transduction by CXCR7 is controlled at the membrane by the process of GPCR trafficking. In the present study we investigated the regulatory processes triggered by CXCR7 activation as well as the molecular interactions that participate in such processes. We show that, CXCR7 internalizes and recycles back to the cell surface after agonist exposure, and that internalization is not only β-arrestin-mediated but also dependent on the Serine/Threonine residues at the C-terminus of the receptor. Furthermore we describe, for the first time, the constitutive ubiquitination of CXCR7. Such ubiquitination is a key modification responsible for the correct trafficking of CXCR7 from and to the plasma membrane. Moreover, we found that CXCR7 is reversibly de-ubiquitinated upon treatment with CXCL12. Finally, we have also identified the Lysine residues at the C-terminus of CXCR7 to be essential for receptor cell surface delivery. Together these data demonstrate the differential regulation of CXCR7 compared to the related CXCR3 and CXCR4 receptors, and highlight the importance of understanding the molecular determinants responsible for this process.
Collapse
Affiliation(s)
- Meritxell Canals
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
143
|
Law IKM, Murphy JE, Bakirtzi K, Bunnett NW, Pothoulakis C. Neurotensin-induced proinflammatory signaling in human colonocytes is regulated by β-arrestins and endothelin-converting enzyme-1-dependent endocytosis and resensitization of neurotensin receptor 1. J Biol Chem 2012; 287:15066-75. [PMID: 22416137 DOI: 10.1074/jbc.m111.327262] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The neuropeptide/hormone neurotensin (NT) mediates intestinal inflammation and cell proliferation by binding of its high affinity receptor, neurotensin receptor-1 (NTR1). NT stimulates IL-8 expression in NCM460 human colonic epithelial cells by both MAP kinase- and NF-κB-dependent pathways. Although the mechanism of NTR1 endocytosis has been studied, the relationship between NTR1 intracellular trafficking and inflammatory signaling remains to be elucidated. In the present study, we show that in NCM460 cells exposed to NT, β-arrestin-1 (βARR1), and β-arrestin-2 (βARR2) translocate to early endosomes together with NTR1. Endothelin-converting enzyme-1 (ECE-1) degrades NT in acidic conditions, and its activity is crucial for NTR1 recycling. Pretreatment of NCM460 cells with the ECE-1 inhibitor SM19712 or gene silencing of βARR1 or βARR2 inhibits NT-stimulated ERK1/2 and JNK phosphorylation, NF-κB p65 nuclear translocation and phosphorylation, and IL-8 secretion. Furthermore, NT-induced cell proliferation, but not IL-8 transcription, is attenuated by the JNK inhibitor, JNK(AII). Thus, NTR1 internalization and recycling in human colonic epithelial cells involves βARRs and ECE-1, respectively. Our results also indicate that βARRs and ECE-1-dependent recycling regulate MAP kinase and NF-κB signaling as well as cell proliferation in human colonocytes in response to NT.
Collapse
Affiliation(s)
- Ivy Ka Man Law
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095-7019, USA
| | | | | | | | | |
Collapse
|
144
|
Kilpatrick LE, Briddon SJ, Holliday ND. Fluorescence correlation spectroscopy, combined with bimolecular fluorescence complementation, reveals the effects of β-arrestin complexes and endocytic targeting on the membrane mobility of neuropeptide Y receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1068-81. [PMID: 22487268 PMCID: PMC3793875 DOI: 10.1016/j.bbamcr.2012.03.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 01/22/2023]
Abstract
Fluorescence correlation spectroscopy (FCS) and photon counting histogram (PCH) analysis are powerful ways to study mobility and stoichiometry of G protein coupled receptor complexes, within microdomains of single living cells. However, relating these properties to molecular mechanisms can be challenging. We investigated the influence of β-arrestin adaptors and endocytosis mechanisms on plasma membrane diffusion and particle brightness of GFP-tagged neuropeptide Y (NPY) receptors. A novel GFP-based bimolecular fluorescence complementation (BiFC) system also identified Y1 receptor-β-arrestin complexes. Diffusion co-efficients (D) for Y1 and Y2-GFP receptors in HEK293 cell plasma membranes were 2.22 and 2.15 × 10− 9 cm2 s− 1 respectively. At a concentration which promoted only Y1 receptor endocytosis, NPY treatment reduced Y1-GFP motility (D 1.48 × 10− 9 cm2 s− 1), but did not alter diffusion characteristics of the Y2-GFP receptor. Agonist induced changes in Y1 receptor motility were inhibited by mutations (6A) which prevented β-arrestin recruitment and internalisation; conversely they became apparent in a Y2 receptor mutant with increased β-arrestin affinity. NPY treatment also increased Y1 receptor-GFP particle brightness, changes which indicated receptor clustering, and which were abolished by the 6A mutation. The importance of β-arrestin recruitment for these effects was illustrated by reduced lateral mobility (D 1.20–1.33 × 10− 9 cm2 s− 1) of Y1 receptor-β-arrestin BiFC complexes. Thus NPY-induced changes in Y receptor motility and brightness reflect early events surrounding arrestin dependent endocytosis at the plasma membrane, results supported by a novel combined BiFC/FCS approach to detect the underlying receptor-β-arrestin signalling complex.
Collapse
Affiliation(s)
- Laura E Kilpatrick
- Cell Signaling Research Group, School of Biomedical Sciences, University of Nottingham, the Medical School, Queen's Medical Centre, Nottingham, UK
| | | | | |
Collapse
|
145
|
Cheng SB, Filardo EJ. Trans-Golgi Network (TGN) as a regulatory node for β1-adrenergic receptor (β1AR) down-modulation and recycling. J Biol Chem 2012; 287:14178-91. [PMID: 22378779 DOI: 10.1074/jbc.m111.323782] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Receptor down-modulation is the key mechanism by which G protein-coupled receptors (GPCRs) prevent excessive receptor signaling in response to agonist stimulation. Recently, the trans-Golgi network (TGN) has been implicated as a key checkpoint for receptor endocytosis and degradation. Here, we investigated the involvement of the TGN in down-modulation of β1-adrenergic receptor in response to persistent isoprotenerol stimulation. Immunofluorescent staining showed that ~50% of endocytosed β1AR colocalized with TGN-46 at 5 h. Disruption of the TGN by brefeldin A (BFA) led to the robust accumulation of endocytosed β1AR in Rab11(+) recycling endosomes, inhibited β1AR entry into LAMP1(+) lysosomes, and as a result enhanced β1AR recycling to the plasma membrane. The lysosomotropic agent, chloroquine, arrested the majority of endocytosed β1AR in the TGN by 4 h. Immunoblot analysis showed that either disruption of the TGN or blockage of the lysosome prevented β1AR degradation. Co-expression of GFP-arrestin-3 in β1AR cells increased the endocytosis of β1AR and facilitated its entry to the TGN but inhibited recycling to the plasma membrane. Arrestin-3-induced inhibition of β1AR recycling was reversed by BFA treatment, whereas chloroquine induced the accumulation of arrestin-3 with β1AR in the TGN. These results demonstrate for the first time that the TGN acts as a checkpoint for both the recycling and down-regulation of β1AR and that arrestin-3 not only mediates β1AR endocytosis but also its recycling through the TGN.
Collapse
Affiliation(s)
- Shi-Bin Cheng
- Division of Hematology and Oncology, Rhode Island Hospital and Brown University, Providence, Rhode Island 02903, USA
| | | |
Collapse
|
146
|
Punn A, Chen J, Delidaki M, Tang J, Liapakis G, Lehnert H, Levine MA, Grammatopoulos DK. Mapping structural determinants within third intracellular loop that direct signaling specificity of type 1 corticotropin-releasing hormone receptor. J Biol Chem 2012; 287:8974-85. [PMID: 22247544 PMCID: PMC3308756 DOI: 10.1074/jbc.m111.272161] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The type 1 corticotropin-releasing hormone receptor (CRH-R1) influences biological responses important for adaptation to stressful stimuli, through activation of multiple downstream effectors. The structural motifs within CRH-R1 that mediate G protein activation and signaling selectivity are unknown. The aim of this study was to gain insights about important structural determinants within the third intracellular loop (IC3) of the human CRH-R1α important for cAMP and ERK1/2 pathways activation and selectivity. We investigated the role of the juxtamembrane regions of IC3 by mutating amino acid cassettes or specific residues to alanine. Although simultaneous tandem alanine mutations of both juxtamembrane regions Arg292-Met295 and Lys311-Lys314 reduced ligand binding and impaired signaling, all other mutant receptors retained high affinity binding, indistinguishable from wild-type receptor. Agonist-activated receptors with tandem mutations at the proximal or distal terminal segments enhanced activation of adenylyl cyclase by 50–75% and diminished activation of inositol trisphosphate and ERK1/2 by 60–80%. Single Ala mutations identified Arg292, Lys297, Arg310, Lys311, and Lys314 as important residues for the enhanced activation of adenylyl cyclase, partly due to reduced inhibition of adenylyl cyclase activity by pertussis toxin-sensitive G proteins. In contrast, mutation of Arg299 reduced receptor signaling activity and cAMP response. Basic as well as aliphatic amino acids within both juxtamembrane regions were identified as important for ERK1/2 phosphorylation through activation of pertussis toxin-sensitive G proteins as well as Gq proteins. These data uncovered unexpected roles for key amino acids within the highly conserved hydrophobic N- and C-terminal microdomains of IC3 in the coordination of CRH-R1 signaling activity.
Collapse
Affiliation(s)
- Anu Punn
- Department of Endocrinology and Metabolism, Clinical Sciences Research Institute, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Conway BR, Demarest KT. The Use of Biosensors to Study GPCR Function: Applications for High-Content Screening. ACTA ACUST UNITED AC 2011. [DOI: 10.3109/10606820214641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
148
|
Rebois RV, Hébert TE. Protein Complexes Involved in Heptahelical Receptor-Mediated Signal Transduction. ACTA ACUST UNITED AC 2011. [DOI: 10.3109/10606820308243] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
149
|
Abstract
Somatostatin (SS) and dopamine (DA) receptors have been highlighted as two critical regulators in the negative control of hormonal secretion in a wide group of human endocrine tumors. Both families of receptors belong to the superfamily of G protein-coupled receptors and share a number of structural and functional characteristics. Because of the generally reported high expression of somatostatin receptors (SSTRs) in neuroendocrine tumors (NET), somatostatin analogs (SSA) have a pronounced role in the medical therapy for this class of tumors, especially pituitary adenomas and well-differentiated gastroenteropancreatic NET (GEP NET). Moreover, NET express not only SSTR but also frequently dopamine receptors (DRs), and DA agonists targeting the D(2) receptor (D(2)) have been demonstrated to be effective in controlling hormone secretion and cell proliferation in in vivo and in vitro studies. The treatment with SSAs combined with DA agonists has already been demonstrated efficacious in a subgroup of patients with GH-secreting pituitary adenomas and few reported cases of carcinoids. The recent availability of new selective and universal SSA and DA agonists, as well as the chimeric SS/DA compounds, may shed new light on the potential role of SSTR and D(2) as combined targets for biotherapy in NET. This review provides an overview of the latest studies evaluating the expression of SSTR and DR in NET, focusing on their co-expression and the possible clinical implications of such co-expression. Moreover, the most recent insights in SSTR and D(2) pathophysiology and the future perspectives for treatment with SSA, DA agonists, and SS/DA chimeric compounds are discussed.
Collapse
Affiliation(s)
- Federico Gatto
- Division of Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Room Ee530b, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | |
Collapse
|
150
|
Busnelli M, Saulière A, Manning M, Bouvier M, Galés C, Chini B. Functional selective oxytocin-derived agonists discriminate between individual G protein family subtypes. J Biol Chem 2011; 287:3617-29. [PMID: 22069312 DOI: 10.1074/jbc.m111.277178] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We used a bioluminescence resonance energy transfer biosensor to screen for functional selective ligands of the human oxytocin (OT) receptor. We demonstrated that OT promoted the direct engagement and activation of G(q) and all the G(i/o) subtypes at the OT receptor. Other peptidic analogues, chosen because of specific substitutions in key OT structural/functional residues, all showed biased activation of G protein subtypes. No ligand, except OT, activated G(oA) or G(oB), and, with only one exception, all of the peptides that activated G(q) also activated G(i2) and G(i3) but not G(i1), G(oA), or G(oB), indicating a strong bias toward these subunits. Two peptides (DNalOVT and atosiban) activated only G(i1) or G(i3), failed to recruit β-arrestins, and did not induce receptor internalization, providing the first clear examples of ligands differentiating individual G(i/o) family members. Both analogs inhibited cell proliferation, showing that a single G(i) subtype-mediated pathway is sufficient to prompt this physiological response. These analogs represent unique tools for examining the contribution of G(i/o) members in complex biological responses and open the way to the development of drugs with peculiar selectivity profiles. This is of particular relevance because OT has been shown to improve symptoms in neurodevelopmental and psychiatric disorders characterized by abnormal social behaviors, such as autism. Functional selective ligands, activating a specific G protein signaling pathway, may possess a higher efficacy and specificity on OT-based therapeutics.
Collapse
Affiliation(s)
- Marta Busnelli
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Via Vanvitelli 32, Milan 20143, Italy
| | | | | | | | | | | |
Collapse
|