Elsevier

Brain Research

Volume 1184, 12 December 2007, Pages 46-56
Brain Research

Research Report
Agonist treatment did not affect association of mu opioid receptors with lipid rafts and cholesterol reduction had opposite effects on the receptor-mediated signaling in rat brain and CHO cells

https://doi.org/10.1016/j.brainres.2007.09.096Get rights and content

Abstract

Lipid rafts are small cholesterol- and glycosphingolipid-enriched membrane subdomains. Here we compared the mu opioid receptor (MOR)–lipid rafts relationship in the rat brain, where neurons have non-caveolae rafts, and in CHO cells stably transfected with HA-rat MOR (CHO-HA-rMOR), which are enriched in caveolae. Membranes of rat caudate putamen (CPu) and thalamus or CHO-HA-rMOR cells were homogenized, sonicated in a detergent-free 0.5 M Na2CO3 buffer and fractionated through sucrose density gradients. Western blot and [3H]diprenorphine binding showed that ∼ 70% of MOR in CHO-HA-rMOR was present in low-density (5–20% sucrose) fractions enriched in cholesterol and/or ganglioside M1 (GM1) (lipid rafts) in plasma membranes, whereas about 70% and 45% of MOR in CPu and thalamus, respectively, were associated with lipid rafts. Incubation with a saturating concentration of etorphine or morphine at 37 °C for 30 min failed to change the MOR location in rafts in CHO-HA-rMOR, indicating that the internalized MOR does not move out of rafts, in contrast to the delta opioid receptor. In vivo, rafts association of MOR in CPu and thalamus was not affected significantly in rats implanted with two 75-mg morphine pellets for 72 h. In addition, cholesterol reduction by methyl-β-cyclodextrin (MCD) disrupted rafts and shifted MOR to higher density fractions in both CHO-HA-rMOR and CPu membranes. However, MCD treatment had opposite impacts on MOR signaling in the two tissues: it attenuated MOR-mediated [35S]GTPγS binding in CPu but enhanced it in CHO-HA-rMOR.

Introduction

The actions of opioid drugs are mediated by at least three types of opioid receptors: mu, delta and kappa (MOR, DOR and KOR, respectively) (Pasternak, 1988). These receptors belong to rhodopsin subfamily of 7-transmembrane receptors (7-TMRs)/G-protein-coupled receptors (GPCRs) (Bockaert and Pin, 1999). They are coupled via pertussis toxin-sensitive Gi/Go proteins to many downstream effectors such as adenylate cyclase, potassium channels, calcium channels, and a mitogen-activated protein kinase pathway [for a review, see Law et al., 2000]. Although opioid drugs have long been used as analgesics, their clinical use is limited by the fear of addiction and side effects such as respiratory depression, decreased gastrointestinal motility, sedation, mood changes, and tolerance (Kieffer, 1999). Morphine acts primarily on the MOR (Matthes et al., 1996) and is among the most widely used and abused drugs.

Lipid rafts, including non-caveolae rafts and caveolae, are small cell membrane domains (10–200 nm), which are thought to serve as platforms for many cellular processes (Pike, 2006). They are enriched in cholesterol and glycosphingolipids (e.g., GM1). Recently, lipid rafts were suggested to be renamed as “membrane rafts,” because it has become clear that proteins play a major role in their formation and contribute to their functions (Pike, 2006). While caveolae are relatively stable because the protein caveolins support the structure, non-caveolae rafts are considered to be unstable, heterogeneous and dynamically produced and degraded (Pike, 2006). Electron micrographs show that caveolae are flask-shaped membrane invaginations at plasma membranes in most differentiated cells (Cohen et al., 2004). One the contrary, morphological identification of non-caveolae rafts has been elusive (Shaw, 2006). The existence of non-caveolae rafts is largely based on their biochemical nature of insolubility in nonionic detergents at low temperature and high buoyancy in density gradients (Brown and Rose, 1992), which does not differentiate non-caveolae rafts from caveolae.

Many 7-TMRs/GPCRs have been shown to locate in and regulated by lipid rafts (Pike, 2003, Chini and Parenti, 2004, Ostrom and Insel, 2004), including opioid receptors (Head et al., 2005, Xu et al., 2006, Zhao et al., 2006, Patel et al., 2006, Huang et al., 2007). For example, MOR expressed in HEK293 cells was found by Zhao et al. (2006) to be mainly localized in lipid rafts domains of plasma membranes and this localization was required for MOR-mediated adenylyl cyclase superactivation. Our recent findings demonstrated that the kappa and delta opioid receptor (KOR and DOR, respectively) expressed in CHO cells were mostly associated with and regulated by lipid rafts (Xu et al., 2006, Huang et al., 2007). We have also demonstrated that the acute treatment with a full agonist move some DORs out of lipid rafts in both CHO cells and NG108-15 cells (Huang et al., 2007). However, most of those studies were performed in caveolae-rich non-neuronal cells, while numerous GPCRs, including opioid receptors, are present in neurons in the brain, which were demonstrated to be devoid of caveolae and deficient in its structural protein(s), the caveolins (Cameron et al., 1997).

By the use of an anti-MOR antibody (anti-μC), we have recently found that the MOR associated with lipid rafts at different levels in rat caudate putamen (CPu) and thalamus, with cholesterol and ganglioside M1 as markers for lipid rafts (Huang et al., in press). In this study, we determined to what extent MOR in CHO-HA-rMOR was localized in lipid rafts. In addition, we investigated rafts association of MOR in cells following acute treatment of CHO-HA-rMOR with agonists and in CPu and thalamus after chronic morphine treatment of rats in vivo. Moreover, effects of disrupting rafts by cholesterol reduction on MOR–G protein coupling in the two systems were examined.

Section snippets

MOR in rat brain CPu and rMOR expressed in CHO cells are mainly localized in the lipid rafts from plasma membranes

A modified version of our published procedure was used to prepare lipid rafts (Xu et al., 2006, Huang et al., 2007), which was originally described by Song et al. (1996). Prepared by this method, non-caveolae rafts cannot be distinguished from caveolae.

MOR is present at high levels in several regions in the rat brain, including caudate putamen (CPu), where MORs are located in neurons (Mansour et al., 1988). CPu membranes and CHO-HA-rMOR cells were sonicated on ice in a 500 mM sodium carbonate

Discussion

Neurons in the rat brain have non-caveolae rafts (Cameron et al., 1997), while CHO cells have abundant caveolae (Navratil et al., 2003). In this study, we found that (1) ∼ 70% of MOR was located in the rafts of plasma membranes in both rat CPu and CHO-HA-rMOR cells; (2) MOR–rafts association was not changed by incubation of CHO-HA-rMOR cells with agonists; (3) chronic treatment of rats in vivo with morphine did not change distribution of MOR in lipid rafts in CPu and thalamus, which, to the best

Materials

[3H]diprenorphine (58 Ci/mmol) and [35S]guanosine 5-(γ-thio)triphosphate (GTPγS) (1250 Ci/mmol) were purchased from Perkin-Elmer Co. (Boston, MA). Morphine and etorphine were provided by the Drug Supply System of National Institute on Drug Abuse (NIDA). Naloxone, DAMGO, sodium carbonate, 2-morpholinoethanesulfonic acid (MES), glycerol, ethylenediamine tetraacetic acid (EDTA), ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA), dithiothreitol (DTT), PMSF, GDP, GTPγS, and

Acknowledgments

This work was supported by NIH grants DA04745 and DA17302 and supported in part by the Pennsylvania Department of Health. S.-I. Y. and P.L.-G.C. acknowledge the support from AHA (0255082N) and ACS (PRF-38205-AC-7).

References (38)

  • L.J. Pike

    Lipid rafts: bringing order to chaos

    J. Lipid Res.

    (2003)
  • L.J. Pike

    Rafts defined: a report on the Keystone Symposium on Lipid Rafts and Cell Function

    J. Lipid Res.

    (2006)
  • V.O. Rybin et al.

    Differential targeting of beta-adrenergic receptor subtypes and adenylyl cyclase to cardiomyocyte caveolae. A mechanism to functionally regulate the cAMP signaling pathway

    J. Biol. Chem.

    (2000)
  • P. Sharma et al.

    Nanoscale organization of multiple GPI-anchored proteins in living cell membranes

    Cell

    (2004)
  • K.S. Song et al.

    Co-purification and direct interaction of Ras with caveolin, an integral membrane protein of caveolae microdomains. Detergent-free purification of caveolae microdomains

    J. Biol. Chem.

    (1996)
  • J. Bockaert et al.

    Molecular tinkering of G protein-coupled receptors: an evolutionary success

    EMBO J.

    (1999)
  • P.L. Cameron et al.

    Identification of caveolin and caveolin-related proteins in the brain

    J. Neurosci.

    (1997)
  • B. Chini et al.

    G-protein coupled receptors in lipid rafts and caveolae: how, when and why do they go there?

    J. Mol. Endocrinol.

    (2004)
  • M. Chun et al.

    Signal transduction of a G protein-coupled receptor in caveolae: colocalization of endothelin and its receptor with caveolin

    Proc. Natl. Acad. Sci. U. S. A.

    (1994)
  • Cited by (35)

    • Determination of δ-opioid receptor molecules mobility in living cells plasma membrane by novel method of FRAP analysis

      2019, Biochimica et Biophysica Acta - Biomembranes
      Citation Excerpt :

      We demonstrated the applicability of our method and its ability to make highly accurate determinations of the diffusion coefficient (D) and the mobile fraction (Mf) of PM proteins by using HEK293 cells that transiently express δ-OR-eYFP as a model. Considering the importance of cholesterol and membrane domains for the maintenance of an intact PM structure and for OR function [12–18], we analyzed the effects of cholesterol depletion and replenishment on δ-OR mobility. To our best knowledge, this is the first reported analysis of the lateral mobility of δ-OR in the PM of live cells.

    • Differential effect of membrane cholesterol removal on μ- and δ-opioid receptors. A parallel comparison of acute and chronic signaling to adenylyl cyclase

      2009, Journal of Biological Chemistry
      Citation Excerpt :

      This was reflected by a 4-fold increase in EC50 from 16.8 ± 9.3 nm in control cells to 73.0 ± 26.5 nm in MβCD-treated cells, although variability precluded significance (p = 0.116). Even though this difference in G protein activation between μ- and δ-opioid receptors in response to cholesterol removal is small, it does disagree with previous reports using heterologous systems that show an effect of MβCD on δ-opioid agonist-stimulated [35S]GTPγS binding (10, 24, 25). This could be due to cell types utilized or methods of data analysis because the basal level of [35S]GTPγS binding does change.

    • Differential modulation of μ- and δ-opioid receptor agonists by endogenous RGS4 protein in SH-SY5Y cells

      2009, Journal of Biological Chemistry
      Citation Excerpt :

      Antibodies were from the indicated sources: anti-phospho-p44/42 MAPK (ERK1/2) and anti-p44/42 MAPK (ERK1/2) (Cell Signaling Technology, Beverly, MA); anti-FLAG M1 and anti-β-actin (Sigma); anti-δ-opioid receptor, anti-mouse, anti-rabbit, anti-hemagglutinin (HA), anti-HA antibody-conjugated agarose beads, and Protein A/G plus agarose (Santa Cruz Biotechnology, Inc., Santa Cruz, CA). Anti-μ-opioid receptor antibody was as described previously (28), and U1079 RGS4 antiserum was a kind gift from Dr. Stephen Gold (Merck). SuperSignal West Pico chemiluminescent substrate was from Pierce.

    View all citing articles on Scopus
    View full text