Elsevier

Cellular Signalling

Volume 27, Issue 3, March 2015, Pages 716-726
Cellular Signalling

Cannabinoid receptor interacting protein (CRIP1a) attenuates CB1R signaling in neuronal cells

https://doi.org/10.1016/j.cellsig.2014.11.006Get rights and content

Highlights

  • Cannabinoid receptor interacting protein 1a (CRIP1a) decreases CB1R at the plasma membrane surface without altering total CB1R mRNA or protein levels in a neuronal cell model.

  • CRIP1a regulates both constitutive and agonist-stimulated extracellular signal-regulated kinase phosphorylation in a neuronal cell model.

  • CRIP1a regulates inhibition of cyclic AMP accumulation.

  • CRIP1a switches the subtype preference for Gi/o proteins.

  • These are the first demonstrations of cellular functional responses modulated by the abundance of CRIP1a in a neuronal cell.

Abstract

CB1 cannabinoid receptors (CB1R) are one of the most abundantly expressed G protein coupled receptors (GPCR) in the CNS and regulate diverse neuronal functions. The identification of GPCR interacting proteins has provided additional insight into the fine-tuning and regulation of numerous GPCRs. The cannabinoid receptor interacting protein 1a (CRIP1a) binds to the distal carboxy terminus of CB1R, and has been shown to alter CB1R-mediated neuronal function [1]. The mechanisms by which CRIP1a regulates CB1R activity have not yet been identified; therefore the focus of this investigation is to examine the cellular effects of CRIP1a on CB1R signaling using neuronal N18TG2 cells stably transfected with CRIP1a over-expressing and CRIP1a knockdown constructs. Modulation of endogenous CRIP1a expression did not alter total levels of CB1R, ERK, or forskolin-activated adenylyl cyclase activity. When compared to WT cells, CRIP1a over-expression reduced basal phosphoERK levels, whereas depletion of CRIP1a augmented basal phosphoERK levels. Stimulation of phosphoERK by the CB1R agonists WIN55212-2, CP55940 or methanandamide was unaltered in CRIP1a over-expressing clones compared with WT. However, CRIP1a knockdown clones exhibited enhanced ERK phosphorylation efficacy in response to CP55940. In addition, CRIP1a knockdown clones displayed a leftward shift in CP55940-mediated inhibition of forskolin-stimulated cAMP accumulation. CB1R-mediated Gi3 and Go activation by CP99540 was attenuated by CRIP1a over-expression, but robustly enhanced in cells depleted of CRIP1a. Conversely, CP55940-mediated Gi1 and Gi2 activation was significant enhanced in cells over-expressing CRIP1a, but not in cells deficient of CRIP1a. These studies suggest a mechanism by which endogenous levels of CRIP1a modulate CB1R-mediated signal transduction by facilitating a Gi/o protein subtype preference for Gi1 and Gi2, accompanied by an overall suppression of G-protein-mediated signaling in neuronal cells.

Introduction

The CB1 cannabinoid receptor (CB1R) belongs to the class A rhodopsin-like G protein coupled receptor (GPCR) family. CB1Rs display highest expression in the nervous systems [2], [3], [4], where they have been implicated in numerous physiological processes, including but not limited to energy balance, neuroprotection, pain, and cellular differentiation and proliferation [5]. Based on the location and function of CB1Rs in the CNS, it is no surprise that CB1Rs provide a potentially promising therapeutic target for a diverse number of diseases and disorders [6]; however, the clinical utility and success of CB1R therapeutic agents has been impeded as a result of untoward side-effect profiles.

CB1R signaling is mediated by pertussis toxin-sensitive Gi/o proteins, and leads to inhibition of adenylyl cyclase (AC), regulation of ion channels, induction of immediate early gene expression, and activation of members of the mitogen-activated protein kinase (MAPK) family including extra-cellular regulated kinase 1/2 (ERK1/2) [7]. Studies using peptides mimicking specific regions of CB1R's C-terminus or intracellular loop 3 have demonstrated a preference in binding of specific G proteins to different regions of CB1R. Gαi1 and Gαi2 have been reported to interact with the third intracellular loop of CB1R [8], [9], whereas Gαi3 and Gαo primarily interact with the juxtamembrane C-tail domain [8] of CB1R. Additionally, specificity in G protein activation appears to occur in a ligand-dependent manner [10], suggesting that upon binding, CB1R ligands can induce differences in receptor conformational changes, which can lead to the coupling and activation of specific G protein subtypes.

The GPCR C-terminal tail is a major site for protein-protein interactions, and although G protein binding is a key component in GPCR signaling, it is now well appreciated that other modulatory proteins are involved in receptor activity-dependent and G protein selective signaling [11], [12]. The cannabinoid receptor interacting protein (CRIP1a), which binds to the distal C-terminal tail of CB1R, was initially characterized for its ability to reverse CB1R-mediated tonic inhibition of Ca2 + channels in superior cervical ganglion neurons [1]. Studies using a cell culture model of glutamate neurotoxicity in primary neuronal cortical neurons, showed that lentiviral over-expression of CRIP1a reversed CB1R-mediated neuroprotection from an agonist- to antagonist-driven mechanism [13]. However, the underlying mechanism responsible for this modification of CB1R ligand-mediated neuroprotection is unknown.

To explore the emerging roles of CRIP1a in regulating CB1R, our laboratory developed CRIP1a gain and loss of function transgenic neuronal clones, and reported preliminary observations of alterations in agonist-promoted CB1R activation and internalization by CRIP1a [14], [15]. In the present study, we determined the effect of CRIP1a on CB1R signaling and the associated downstream consequences on cellular function. Both CRIP1a over-expression and RNA interference-induced CRIP1a knockdown were examined in stably-transfected clones of the N18TG2 neuronal cell line, which endogenously expresses both CB1R and CRIP1a. The focus was on CB1R- Gαi/o-mediated inhibition of cAMP production and Gβγ-mediated MAPK activation. Herein we demonstrate that CRIP1a functions as a negative modulator of CB1R cellular signaling, as depletion of CRIP1a increased the interaction with Gi3 and Go subtypes, increased the potency of CB1R agonists to inhibit forskolin-stimulated cAMP accumulation, and enhanced the efficacy of CB1R agonist-stimulated ERK phosphorylation. These studies suggest a role for CRIP1a in CB1R signaling and modulation of agonist-mediated G protein coupling.

Section snippets

Cell culture and generation of stable neuronal CRIP1a transgenic clones

N18TG2 neuroblastoma cells and stable clones were cultured and maintained in complete media containing Dulbecco's Modified Eagle's Medium (DMEM):Ham's F-12 (1:1) with GlutaMax, sodium bicarbonate, and pyridoxine-HCl, supplemented with penicillin (100 units/ml) and streptomycin (100 μg/ml) (Gibco Life Technologies, Gaithersburg, MD, USA) and 10% heat-inactivated bovine serum (JRH Biosciences, Lenexa, KS, USA), and incubated at 37 °C in a humidified atmosphere containing 95% air and 5% carbon

CRIP1a influences CB1R cell surface equilibrium but not mRNA or total protein levels

In order to better understand the cellular mechanisms involved in the regulation of CB1R by CRIP1a, we developed stable neuronal transgenic CRIP1a over-expressing (XS) and knockdown (KD) clones in the N18TG2 cell line. Based on their CRIP1a mRNA and protein expression, two different CRIP1a XS and KD clones were selected for the present investigation. CRIP1a XS clones 1 and 5 express CRIP1a:CB1R mRNA levels that are 12:1 (XS 1) and 7:1 (XS 5), compared with a 1:7 ratio in WT cells (Fig. 1A).

Discussion

In addition to direct stimulation of the receptor by agonists, signaling by CB1R can be further modified by accessory proteins, such as β-arrestin, G protein Associated Sorting Proteins (GASP), and CRIP1a [11], [12], [15]. CRIP1a was initially characterized for its ability to bind to a segment of the distal C-terminal of the CB1R [1]. Following that report, studies of retinal circuitry showed that CRIP1a could be found in amacrine cells, and in certain cone (but not rod) terminals [33]. The CB1

Conclusions

The knowledge gained regarding the effects of CRIP1a on CB1R signaling demonstrates how CB1R function and activity can be fine-tuned by accessory proteins. We determined that reduction in CRIP1a protein levels increased CB1R agonist-stimulated Gi3/o protein activation and promoted inhibition of forskolin-stimulated cAMP accumulation in neuroblastoma cells. Depletion of CRIP1a enhanced CB1R-mediated maximal ERK phosphorylation, a process that was abolished by blocking CB1R internalization. This

Acknowledgements

This work was supported by US Public Health Services grants: R01-DA03690 (ACH), R21-DA025321 (ACH and DES), K01-DA024763 (CEB), P50-DA006634 (ACH, CEB, KE), K12-GM102773 (KE), T32-DA00724 and F31-DA032215 (LCB). The authors declare no conflicts of interest.

References (40)

  • B. Stauffer et al.

    Neurosci. Lett.

    (2011)
  • J.L. Wacker et al.

    J. Biol. Chem.

    (2008)
  • J.P. Meschler et al.

    Biochem. Pharmacol.

    (2000)
  • C. Leterrier et al.

    J. Biochem.

    (2004)
  • J. Marazzi et al.

    J. Immunol. Methods

    (2011)
  • D. Sarnataro et al.

    FEBS Lett.

    (2005)
  • M. Bari et al.

    J. Biol. Chem.

    (2005)
  • M. Bari et al.

    Neuropharmacology

    (2008)
  • S. Mukhopadhyay et al.

    Chem. Phys. Lipids

    (2002)
  • M. McVey et al.

    J. Biol. Chem.

    (1999)
  • G.D. Dalton et al.

    Cell. Signal.

    (2013)
  • J.L. Niehaus et al.

    Mol. Pharmacol.

    (2007)
  • S.M. Eggan et al.

    Cereb. Cortex

    (2007)
  • M. Herkenham et al.

    Brain Res.

    (2001)
  • L.J. Sim et al.

    J. Neurosci.

    (1996)
  • R.G. Pertwee

    Int. J. Obes. (Lond.)

    (2006)
  • K.A. Seely et al.

    Mol. Interv.

    (2011)
  • A.C. Howlett

    Cannabinoid receptor signaling

    Handbook of Experimental Pharmacology

    (2005)
  • S. Mukhopadhyay et al.

    Mol. Pharmacol.

    (2000)
  • S. Mukhopadhyay et al.

    Eur. J. Biochem.

    (2001)
  • Cited by (32)

    • Cannabinoid receptor CB<inf>1</inf> and CB<inf>2</inf> interacting proteins: Techniques, progress and perspectives

      2021, Methods in Cell Biology
      Citation Excerpt :

      CRIP1a did not influence WIN55,212-2-induced inhibition of voltage-gated Ca2 + channels but reduced constitutive CB1-mediated inhibition without influencing inverse agonist affinity nor overall CB1 expression (surface expression was not specifically measured), whereas CRIP1b had no detectable effect on the parameters measured (Niehaus et al., 2007). In most subsequent reports CRIP1a has tended to suppress CB1 activity, reducing agonist-induced G protein coupling, phospho-ERK stimulation, constitutive internalization and agonist-induced internalization (Blume, Eldeeb, Bass, Selley, & Howlett, 2015; Blume et al., 2016, 2017; Mascia et al., 2017; Smith et al., 2015), though effects were not always consistent between studies which may to some degree involve ligand-dependence (Blume et al., 2016), or otherwise likely implies context-dependence. One study in hippocampal neurons instead found that CB1 agonist-induced G protein activation was enhanced (Guggenhuber et al., 2016), though whether this could have been a consequence of lower constitutive activity is not clear.

    • In silico interaction analysis of cannabinoid receptor interacting protein 1b (CRIP1b) – CB1 cannabinoid receptor

      2017, Journal of Molecular Graphics and Modelling
      Citation Excerpt :

      Exogenous CRIP1a reversed CB1 receptor-mediated tonic inhibition of Ca2+ currents, whereas CRIP1b could not [11]. CRIP1a-knockdown clones in the model neuroblastoma N18TG2 cells exhibited enhanced ERK1/2 phosphorylation efficacy in response to CP55940 and displayed a leftward shift in CP55940-mediated inhibition of forskolin-stimulated cAMP accumulation [12,13]. CB1 receptor-mediated Gi3 and Go activation by CP55940 was attenuated by CRIP1a over-expression, but robustly enhanced in CRIP1a-knockdown clones.

    • Mouse Neuroblastoma CB<inf>1</inf> Cannabinoid Receptor-Stimulated [<sup>35</sup>S]GTPɣS Binding: Total and Antibody-Targeted Gα Protein-Specific Scintillation Proximity Assays

      2017, Methods in Enzymology
      Citation Excerpt :

      This procedure can mitigate concerns related to activation or inhibition of multiple G proteins and reduces the variability in sensitivity by different G proteins (Milligan, 2003; Strange, 2010). This assay has since been used to identify specific CB1 receptor–G protein interactions (Blume, Eldeeb, Bass, Selley, & Howlett, 2015; Diez-Alarcia et al., 2016; Eldeeb et al., 2016; Erdozain, Diez-Alarcia, Meana, & Callado, 2012). Notes: Various types of serum, including fetal bovine serum, have been compared for growth of N18TG2 cells, and the heat-inactivated bovine serum or heat-inactivated bovine calf serum provided the greatest viability and cellular signaling in cyclic AMP accumulation assays.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text