Differential β-arrestin2 requirements for constitutive and agonist-induced internalization of the CB1 cannabinoid receptor

https://doi.org/10.1016/j.mce.2013.03.013Get rights and content

Highlights

  • CB1R binds β-arrestin2 but not β-arrestin1 upon activation.

  • Agonist-induced internalization of CB1R is β-arrestin2 dependent.

  • Constitutive CB1R internalization does not require β-arrestin2.

  • Agonist-induced and constitutive CB1R endocytosis are distinctly regulated processes.

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.

Introduction

Internalization of G protein-coupled receptors (GPCRs) is an important process in the regulation of receptor function. Although its main function is the modulation of receptor number on the cell surface, thereby adjusting the sensitivity of the cell to external stimuli, it also plays role in the resensitization and signaling of GPCRs (Ferguson, 2001, Hunyady and Catt, 2006, Shenoy and Lefkowitz, 2011). At the molecular level, β-arrestins are key regulatory proteins of receptor internalization, as they can bind to activated GPCRs, as well as to clathrin and the adaptor protein AP-2, thus directing the receptor towards clathrin-mediated endocytosis (Shenoy and Lefkowitz, 2011). β-Arrestins also mediate receptor desensitization, as their binding to the activated GPCRs causes the uncoupling of the receptor from its cognate G protein (Shenoy and Lefkowitz, 2011). Furthermore, they play important roles in the activation of G protein-independent signal transduction pathways, e.g. the activation of MAP kinases, phosphatidylinositol 3 kinase, Akt or the small GTP-ase RhoA (DeWire et al., 2007, Wei et al., 2003).

β-Arrestin1 (β-arr1) and β-arrestin2 (β-arr2) are two ubiquitously expressed isoforms of β-arrestins (Ferguson, 2001). Although β-arrestin binding is a general property of most activated GPCRs, the selectivity and stability of these binding shows receptor specific differences. Namely, class A receptors (e.g. the β2 adrenergic receptor) bind β-arr2 with a higher affinity than β-arr1, and this binding is transient, i.e. it can only be detected at or near the plasma membrane. In contrast, class B GPCRs, such as the AT1 angiotensin receptor, bind both β-arr1 and β-arr2 with relatively high affinity and form stable complexes with β-arrestins, so that β-arrestins remain bound to the receptor after internalization and can be detected on intracellular vesicles (Oakley et al., 2000).

The CB1 cannabinoid receptor (CB1R) belongs to the superfamily of G protein-coupled receptors (GPCRs). The receptor plays role in many important physiological processes, such as learning, thinking, nociception or the regulation of food-intake (Pacher et al., 2006). Activation of presynaptic CB1Rs by postsynaptic endocannabinoid release, which mediates retrograde transmission, is a key regulatory mechanism in the central nervous system, but paracrine activation of CB1Rs with a similar mechanism can also occur in extraneural tissues (Freund et al., 2003, Gyombolai et al., 2012, Sanchez et al., 2001, Turu et al., 2009, Szekeres et al., 2012). The cellular signaling events following CB1R activation are mainly associated with the activation of heterotrimeric Gi/o-proteins and include inhibition of adenylyl cyclases, activation of Kir channels, inhibition of Cav channels and phosphorylation and activation of different subtypes of mitogen-activated protein kinases (MAP kinases) (Turu and Hunyady, 2010). G protein-independent signaling events following CB1R stimulation have also been reported (Sanchez et al., 2001).

Similar to most GPCRs, CB1R internalizes upon agonist stimulation. This has been demonstrated in many cell lines, including CHO (Rinaldi-Carmona et al., 1998), AtT20 (Hsieh et al., 1999, Jin et al., 1999, Roche et al., 1999), F11 (Coutts et al., 2001), neuroblastoma N18TG2 (Keren and Sarne, 2003) and HEK293 (Keren and Sarne, 2003, Leterrier et al., 2004) cells, as well as in hippocampal neurons, which naturally express CB1R (Coutts et al., 2001, Leterrier et al., 2006). According to different studies, this agonist-induced CB1R endocytosis occurs via clathrin- and/or caveolin-mediated pathways in different cell types (Bari et al., 2008, Hsieh et al., 1999, Keren and Sarne, 2003, Wu et al., 2008).

Numerous studies suggest that β-arr2 is involved in the regulation of CB1R. Co-expression of both GRK3 and β-arr2 was needed for the proper desensitization of the receptor in Xenopus oocyte (Jin et al., 1999), and dominant negative GRK2 and β-arrestin constructs reduced CB1R desensitization in hippocampal neurons (Kouznetsova et al., 2002). In β-arr2 knockout mice desensitization and downregulation of CB1R were impaired in certain regions of the central nervous system (Nguyen et al., 2012). Recruitment of β-arr2 to the activated CB1R has also been demonstrated (Daigle et al., 2008a). Mutation of amino acids S426 and S430 was shown to inhibit receptor desensitization as well as late phase receptor endocytosis, but not β-arrestin binding (Daigle et al., 2008b, Jin et al., 1999). It has been demonstrated that serine and threonine residues at the C-terminus of CB1R are involved in its β-arr2 binding and agonist-induced endocytosis (Daigle et al., 2008b, Hsieh et al., 1999). In contrast to the various studies that clearly point to an interaction between CB1R and β-arr2 upon agonist stimulation, no direct data have been hitherto presented concerning the β-arr1 binding of CB1R. In some structural studies, the association of β-arr1 with a synthesized CB1R C-terminus has been shown (Bakshi et al., 2007, Singh et al., 2011), however, such binding has not been demonstrated with the intact CB1R in living cells.

Constitutive internalization of CB1R (i.e. spontaneous internalization in the absence of CB1R agonists) has also been detected in hippocampal neurons, as well as in CHO and HEK cells (Leterrier et al., 2004, Leterrier et al., 2006, McDonald et al., 2007a, Turu et al., 2007). It has been suggested that constitutive CB1R internalization is the consequence of its basal activity, since inverse agonist treatment or inhibition of basal activity with a DAG lipase inhibitor (e.g. tetrahydrolipstatin) interfered with this process (Leterrier et al., 2004, Leterrier et al., 2006, Rinaldi-Carmona et al., 1998, Turu et al., 2007). However, other studies have concluded that constitutive internalization occurs independently of receptor activity (McDonald et al., 2007a, McDonald et al., 2007b, Kleyer et al., 2012). The latter statement raises the possibility that constitutive and agonist-induced internalization of CB1R may occur via distinct endocytic mechanisms (McDonald et al., 2007a). However, no evidence has been hitherto presented showing that these two processes are truly different in that they require distinct endocytic machinery to take place.

Our main goal was, considering the important consequences of β-arrestin recruitment in the regulation of cell function, to characterize the β-arrestin binding properties of CB1R and to reveal possible differences in constitutive and agonist-driven CB1R endocytosis by investigating the role of β-arrestins in these processes.

Section snippets

Materials

The cDNAs of the rat vascular CB1R and CB1R-eYFP were provided by Zsolt Lenkei (Centre National de la Recherche Scientificue, Paris). β-Arrestin1, β-arrestin2 and β-arrestin2-eGFP cDNAs were kindly provided by Dr. Marc G. Caron (Duke University, Durham, NC). Molecular biology enzymes were obtained from Fermentas (Vilnius, Lithuania) and Stratagene (La Jolla, CA). pcDNA3.1 vector, coelenterazine h, fetal bovine serum (FBS), OptiMEM, Lipofectamine 2000, and PBS-EDTA were from Invitrogen

β-Arrestin binding properties of CB1R

β-Arrestin binding properties of CB1R were first analyzed using a bioluminescence resonance energy transfer (BRET) based approach. BRET was measured between Renilla luciferase tagged β-arr1 or β-arr2 (β-arr1-Rluc and β-arr2-Rluc) and the mVenus-tagged CB1R (CB1R-mVenus) in transiently transfected HeLa cells. Stimulation using WIN55,212-2 (WIN55, 10 μM), a potent synthetic CB1R agonist, led to an increase in the BRET signal when β-arr2-Rluc was co-expressed with CB1R-mVenus (Fig. 1A) reflecting

Discussion

Our data show that, based on its β-arrestin binding characteristics, CB1R can be classified as a class A GPCR. The relatively low affinity of the binding between CB1R and β-arr2 is indicated by the low steepness of slope of the BRET saturation curve between the two molecules upon CB1R stimulation, and by the lack of detectable β-arrestin2 in late endosomes after CB1R internalization in confocal experiments. Furthermore, we found in our BRET and confocal measurements that β-arr1 recruitment to

Acknowledgements

This research was supported by Hungarian Science Foundation (OTKA NK-100883), TAMOP-4.2.1.B-09/1/KMR-2010-0001 and a Marie Curie International Outgoing Fellowship within the 7th European Community Framework Programme (PIOF-GA-2009-253628).

References (61)

  • J.G. Krupnick et al.

    Modulation of the arrestin–clathrin interaction in cells. Characterization of beta-arrestin dominant-negative mutants

    J. Biol. Chem.

    (1997)
  • C. Leterrier et al.

    Constitutive endocytic cycle of the CB1 cannabinoid receptor

    J. Biol. Chem.

    (2004)
  • S. Marullo et al.

    Resonance energy transfer approaches in molecular pharmacology and beyond

    Trends Pharmacol. Sci.

    (2007)
  • N.A. McDonald et al.

    Generation and functional characterization of fluorescent, N-terminally tagged CB1 receptor chimeras for live-cell imaging

    Mol. Cell. Neurosci.

    (2007)
  • P.T. Nguyen et al.

    Beta-Arrestin2 regulates cannabinoid CB(1) receptor signaling and adaptation in a central nervous system region-dependent manner

    Biol. Psychiatry

    (2012)
  • R.H. Oakley et al.

    Differential affinities of visual arrestin, beta arrestin1, and beta arrestin2 for G protein-coupled receptors delineate two major classes of receptors

    J. Biol. Chem.

    (2000)
  • R.H. Oakley et al.

    Molecular determinants underlying the formation of stable intracellular G protein-coupled receptor-beta-arrestin complexes after receptor endocytosis

    J. Biol. Chem.

    (2001)
  • M.M. Paing et al.

    Beta-Arrestins regulate protease-activated receptor-1 desensitization but not internalization or down-regulation

    J. Biol. Chem.

    (2002)
  • J.L. Parent et al.

    Role of the differentially spliced carboxyl terminus in thromboxane A2 receptor trafficking: identification of a distinct motif for tonic internalization

    J. Biol. Chem.

    (2001)
  • S.K. Shenoy et al.

    Beta-Arrestin-mediated receptor trafficking and signal transduction

    Trends Pharmacol. Sci.

    (2011)
  • M. Szekeres et al.

    Angiotensin II induces vascular endocannabinoid release, which attenuates its vasoconstrictor effect via CB1 cannabinoid receptors

    J. Biol. Chem.

    (2012)
  • A. Tohgo et al.

    The stability of the G protein-coupled receptor-beta-arrestin interaction determines the mechanism and functional consequence of ERK activation

    J. Biol. Chem.

    (2003)
  • G. Turu et al.

    Differential beta-arrestin binding of AT1 and AT2 angiotensin receptors

    FEBS Lett.

    (2006)
  • G. Turu et al.

    The role of diacylglycerol lipase in constitutive and angiotensin AT1 receptor-stimulated cannabinoid CB1 receptor activity

    J. Biol. Chem.

    (2007)
  • G. Turu et al.

    Paracrine transactivation of the CB1 cannabinoid receptor by AT1 angiotensin and other Gq/11 protein-coupled receptors

    J. Biol. Chem.

    (2009)
  • C.M. Vines et al.

    N-formyl peptide receptors internalize but do not recycle in the absence of arrestins

    J. Biol. Chem.

    (2003)
  • R.J. Ward et al.

    Hetero-multimerization of the cannabinoid CB1 receptor and the orexin OX1 receptor generates a unique complex in which both protomers are regulated by orexin A

    J. Biol. Chem.

    (2011)
  • J. Zhang et al.

    Dynamin and beta-arrestin reveal distinct mechanisms for G protein-coupled receptor internalization

    J. Biol. Chem.

    (1996)
  • M. Zhao et al.

    Arrestin regulates MAPK activation and prevents NADPH oxidase-dependent death of cells expressing CXCR2

    J. Biol. Chem.

    (2004)
  • A.A. Coutts et al.

    Agonist-induced internalization and trafficking of cannabinoid CB1 receptors in hippocampal neurons

    J. Neurosci.

    (2001)
  • Cited by (39)

    • G protein-coupled receptor (GPCR)-dependent transduction

      2022, Allosteric Modulation of G Protein-Coupled Receptors
    • Pharmacological selection of cannabinoid receptor effectors: Signalling, allosteric modulation and bias

      2021, Neuropharmacology
      Citation Excerpt :

      This is especially obvious between low arrestin-recruiting ligands such as Δ9-THC, and high-efficacy ligands such as AMB-FUBINACA (Finlay et al., 2019, discussed below). CB1 has been observed to internalise rapidly from the cell surface in response to stimulation, with rates varying between primary cultures and heterologous cell lines (Coutts et al., 2001; Daigle et al., 2008b; Grimsey et al., 2010; Gyombolai et al., 2013; Rozenfeld and Devi, 2008; Zhu et al., 2019). Following endocytosis, CB1 has been suggested to be returned (recycled) to the cell surface (Leterrier et al., 2004).

    • Heterologous phosphorylation–induced formation of a stability lock permits regulation of inactive receptors by β-arrestins

      2018, Journal of Biological Chemistry
      Citation Excerpt :

      β-Arrestin2-GFP, β-arrestin2 in pCMV5 vector, and dominant–negative (K44A-mutant) dynamin2A were kind gifts of Dr. Marc G. Caron, Dr. Stephen S. Ferguson, and Dr. Kazuhisa Nakayama, respectively. β-Arrestin2–Venus, β-arrestin2–Cerulean, Venus–Rab5, Super Renilla luciferase-tagged β2AR (β2AR-Sluc), EGFR, Cameleon D3 Ca2+ BRET biosensor, β2AR–Cerulean, V2R–Sluc, M3AChR, and α1AAR constructs were described previously (69–74). NES-BirA was produced by cloning the biotin ligase BirA from pcDNA3.1 MCS-BirA(R118G)–HA (obtained from Abcam (Cambridge, UK)) with a nuclear export signal (PubMed PMID 20972448, QVQAGELQGQLVDVH) attached to its N terminus and was placed in pEYFP-N1 vector between AgeI and NotI restriction sites.

    • Angiotensin type 1A receptor regulates β-arrestin binding of the β<inf>2</inf>-adrenergic receptor via heterodimerization

      2017, Molecular and Cellular Endocrinology
      Citation Excerpt :

      These findings reveal a possible new physiological crosstalk mechanism between AT1R and β2AR. The AT1R, AT1R-DRY/AAY (Gaborik et al., 2003), AT1R-Δ319 (Hunyady et al., 1994), β2AR, β2AR-Sluc, untagged 5HT2CR-VGV (I156V, N158G, I160V), 5HT2CR-VGV-Sluc, PM-mRFP (mRFP fused to plasma membrane target sequence of Lyn) (Toth et al., 2012), AT1R-Rluc (Szakadati et al., 2015), AT1R-Venus, β-arrestin1-Venus, β-arrestin2-Venus (Gyombolai et al., 2013), β-arrestin2-Rluc, β2AR-Venus (Turu et al., 2006), Cameleon D3 Ca2+-BRET sensor (Gulyas et al., 2015), EPAC cAMP-BRET sensor (Erdelyi et al., 2014) and L10-Venus (Venus fused to plasma membrane target sequence of Lck) (Toth et al., 2016) constructs were previously described. 5HT2CR-Venus was generated by replacing the Super Renilla luciferase (Sluc) tag to monomeric Venus (Venus) in the 5HT2CR-Sluc construct.

    • Signaling and Regulation of the Cannabinoid CB1 Receptor

      2017, Handbook of Cannabis and Related Pathologies: Biology, Pharmacology, Diagnosis, and Treatment
    View all citing articles on Scopus
    View full text