Elsevier

Neuropharmacology

Volume 44, Issue 4, March 2003, Pages 482-492
Neuropharmacology

μ-opioid receptor activation prevents apoptosis following serum withdrawal in differentiated SH-SY5Y cells and cortical neurons via phosphatidylinositol 3-kinase

https://doi.org/10.1016/S0028-3908(03)00024-8Get rights and content

Abstract

Opioid peptides and alkaloids exert their effects via G protein-coupled receptors (GPCRs). It has been shown that, in addition to trophic factors, some GPCRs are able to activate the phosphatidylinositol 3-kinase/Akt (PI 3-K/Akt) signal transduction pathway, thus leading to cell survival. The aim of this study was to test whether activation of μ-opioid receptors has protective effects on serum withdrawal-induced cell death and to study the possible implication of PI 3-K in this process. In SH-SY5Y neuroblastoma cells fully differentiated by exposure to retinoic acid for five days, the enkephalin derivative selective μ-agonist DAMGO (0.1–2 μM) and the alkaloid morphine (0.1–10 μM) promoted cell survival after serum deprivation (MTT and trypan blue exclusion assays), without inducing cell proliferation. These effects were fully reversed by naloxone, by the selective μ-antagonist β-funaltrexamine (β-FNA) and also by the specific PI 3-K inhibitor LY294002. The two agonists stimulated Akt phosphorylation and the effect was also abolished by β-FNA and by LY294002. In mouse primary cortical neurons, DAMGO reduced the percentage of apoptosis after 6, 12, 24 and 48 h of serum withdrawal; as determined by Hoechst staining. This effect was blocked by β-FNA, by pre-treatment with pertussis toxin and by LY294002. DAMGO also stimulated Akt phosphorylation via PI 3-K in this primary neuronal culture. Together, these results indicate that stimulation of the μ-opioid receptor promotes neuronal survival in a Gi/o-linked, PI 3-K-dependent signaling cascade and suggest that Akt may be a key downstream kinase involved in this anti-apoptotic effect.

Introduction

Endogenous opioid peptides and alkaloids such as morphine mediate their biological effects through membrane-bound heptahelical proteins called opioid receptors. The three main types of receptors for opioid drugs, μ (also known as OP3), δ (OP1) and κ (OP2) receptors (Dhawan et al., 1996) belong to the G protein-coupled receptor (GPCR) family (Connor and Christie, 1999). Agonist activation of GPCRs leads to a physical interaction between the receptor and the GDP-bound G protein heterodimer, which causes the dissociation of the guanine nucleotide and the incorporation of GTP in the G protein α subunit, thus releasing the βγ heterodimer. In turn, GTP-bound α subunits and βγ complexes initiate, independently, a variety of intracellular signaling pathways (Gilman, 1987).

Evidence has accumulated that, in addition to regulate classic effectors such as adenylate cyclase, phosphodiesterases, phospholipases and ion channels, some members of the GPCRs family are able to activate intracellular kinase cascades implicated in cell survival and translational control, such as the mitogen-activated protein kinases (MAPKs) and the phosphatidylinositol 3-kinase (PI 3-K)/protein kinase B (also known as Akt) signalling pathways (for a review see Katada et al., 1999, Lopez-Ilasaca, 1998, Luttrell et al., 1999). In this context, specific Gβγ heterodimers may directly activate PI 3-Ks (Kurosu et al., 1997, Leopoldt et al., 1998, Murga et al., 1998, Murga et al., 2000); whereas Gα subunits have no stimulatory effect (Bommakanti et al., 2000). Activated PI 3-Ks phosphorylate membrane inositol lipids at the D-3 position of the inositol ring, thus generating binding sites for the pleckstrin-homology (PH) domain of Akt. Once in the plasma membrane, Akt is fully activated by phosphorylation at specific residues (Alessi and Cohen, 1998).

It is well documented that when Akt is phosphorylated in response to a wide variety of growth factor stimuli, it plays a key role in promoting cell survival by opposing apoptotic pathways (Dudek et al., 1997, Lawlor and Alessi, 2001). In this context, morphine administration has been shown to reduce naturally occurring neuronal death (Meriney et al., 1985) and a role for endogenous opioid peptides acting as trophic factors regulating neuronal survival and cell proliferation during nervous system development has been proposed (Meriney et al., 1991, Pérez-Navarro et al., 1993, Schmahl et al., 1989). Moreover, the μ selective agonist [d-Ala2, N-Me-Phe4, Gly5-ol]enkephalin (DAMGO) has synergistic effects with nerve growth factor (NGF) on neuronal survival in culture (Sakaguchi et al., 1999) and opioid agonists transiently prevent activation of apoptotic cell death following serum withdrawal in PC12 cells (Dermitzaki et al., 2000). However, no evidence has been reported for the implication of opioid-mediated PI 3-K/Akt activation on these cell survival-promoting effects.

The present work was designed first, to test whether activation of the μ-opioid receptor could have protective effects on neuronal survival after trophic factor deprivation and secondly, to study if these effects could be mediated through the PI 3-K/Akt signalling cascade. We first tested these hypotheses in the SH-SY5Y neuroblastoma cell line. This cell line is a subclone derived from the parent SK-N-SH that expresses predominantly μ-opioid receptors and, thus, has been widely employed as a model system for studying the molecular mechanisms of the function of these receptors (Elliott et al., 1997, Zadina et al., 1994). We have previously developed a protocol of differentiation for the SH-SY5Y cell line based on the sequential treatment with retinoic acid (RA) and brain-derived neurotrophic factor (BDNF) in serum-free medium. This procedure yields homogenous populations of non-proliferating cells exhibiting neuronal phenotype and depending on the continuous presence of BDNF for survival (Encinas et al., 2000). Removal of this neurotrophin or selective blockade of the PI 3-K/Akt pathway causes apoptotic cell death (Encinas et al., 1999, Encinas et al., 2000). Then, we tested the potential protective effects of μ-opioid receptor agonists on apoptotic cell death in a primary culture of mouse cortical neurons. It is well established that in these cells the PI 3-K/Akt pathway is necessary and sufficient for serum as well as BDNF-mediated survival (Hetman et al., 1999).

Section snippets

Materials

DAMGO and β-funaltrexamine (β-FNA) were purchased from Tocris Cookson Ltd (Bristol, UK). Morphine HCl was from Unión Química Farmacéutica, S.A.E. (Madrid, Spain). Human recombinant BDNF was from Alomone Laboratories (Jerusalem, Israel). The PI 3-K inhibitor LY294002 and the MAPK and extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor PD 98059 were from Calbiochem-Novabiochem (Läufelfingen, Switzerland). Naloxone HCl, pertussis toxin (PTX), culture media and other reagents were

Effects of μ-opioid agonists on SH-SY5Y survival following serum withdrawal

To study possible survival-promoting effects of μ-agonists, cell death was induced in SH-SY5Y cells by culturing them in serum-free medium. RA-differentiated SH-SY5Y cells in the presence of BDNF (50 ng/ml) were taken as 100% of survival (Encinas et al., 2000). Serum withdrawal for 24 h caused a significant decrease (48%) in the ability to metabolize MTT to formazan (Fig. 1(a)); indicating loss of mitochondrial membrane potential in dying cells (Ankarcrona et al., 1995). The presence of the

Discussion

Evidence has accumulated that, in addition to survival factors, agonists of the GPCR family can activate the PI 3-K/Akt pathway; some of these receptors include the β-adrenergic receptor (Moule et al., 1997), the m1 and m2 muscarinic receptors (Murga et al., 1998), the lysophosphatidic acid (LPA1/VZG-1) receptor (Weiner and Chun, 1999), the CB1 cannabinoid receptor (Gómez del Pulgar et al., 2000), the A2A adenosine receptor (Lee and Chao, 2001) and the μ-opioid receptor (Polakiewicz et al., 1998

Acknowledgements

This study was supported by Spanish Govern ‘Comisión Interministerial de Ciencia y Tecnología’ through the ‘Plan Nacional de Salud y Farmacia’ Grants SAF2000-0164-CO2-02 (G.O.) and SAF2000-0164-CO2-01 (J.X.C.). Research of the J.X.C. group is supported by ‘Generalitat de Catalunya (Distinció per a Joves Investigadors Universitaris and Suport a Grups de Recerca)’. M.F.S. was supported by a predoctoral fellowship from ‘Ministerio de Educación, Cultura y Deporte’ (Spain). We thank I. Sanchez, R.

References (63)

  • Y.N. Hou et al.

    A μ-receptor opioid agonist induces AP-1 and NF-κB transcription factor activity in primary cultures of rat cortical neurons

    Neuroscience Letters

    (1996)
  • T. Katada et al.

    Synergistic activation of a family of phosphoinositide 3-kinase via G-protein coupled and tyrosine kinase-related receptors

    Chemistry and Physics of Lipids

    (1999)
  • M.S. Kim et al.

    Protective effects of morphine in peroxynitrite-induced apoptosis of primary rat neonatal astrocytes: potential involvement of G protein and phosphatidylinositol 3-kinase (PI3 kinase)

    Biochemical Pharmacology

    (2001)
  • F. Kugawa et al.

    Buprenorphine hydrochloride induces apoptosis in NG108-15 nerve cells

    European Journal of Pharmacology

    (1998)
  • H. Kurosu et al.

    Heterodimeric phosphoinositide 3-kinase consisting of p85 and p110β is synergistically activated by the βγ subunits of G proteins and phosphotyrosyl peptide

    Journal of Biological Chemistry

    (1997)
  • D. Leopoldt et al.

    Gβγ stimulates phosphoinositide 3-kinase by direct interaction with two domains of the catalytic p110 subunit

    Journal of Biological Chemistry

    (1998)
  • M. Lopez-Ilasaca

    Signaling from G-protein-coupled receptors to mitogen-activated protein (MAP)-kinase cascades

    Biochemical Pharmacology

    (1998)
  • L.M. Luttrell et al.

    Regulation of tyrosine kinase cascades by G-protein-coupled receptors

    Current Opinion in Cell Biology

    (1999)
  • A. Mansour et al.

    Opiod-receptor mRNA expression in the rat CNS: anatomical and functional implications

    Trends in Neuroscience

    (1995)
  • T. Mosmann

    Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays

    Journal of Immunological Methods

    (1983)
  • S.K. Moule et al.

    Regulation of protein kinase B and glycogen synthase kinase-3 by insulin and β-adrenergic agonists in rat epididymal fat cells. Activation of protein kinase B by wortmannin-sensitive and -insensitive mechanisms

    Journal of Biological Chemistry

    (1997)
  • C. Murga et al.

    Activation of Akt/protein kinase B by G protein-coupled receptors

    Journal of Biological Chemistry

    (1998)
  • C. Murga et al.

    A novel role for phosphatidylinositol 3-kinase β in signaling from G protein-coupled receptors to Akt

    Journal of Biological Chemistry

    (2000)
  • L.A. Opanashuk et al.

    Opposing actions of the EGF family and opioids: heparin binding-epidermal growth factor (HB-EGF) protects mouse cerebellar neuroblasts against the antiproliferative effect of morphine

    Brain Research

    (1998)
  • E. Pérez-Navarro et al.

    Nerve growth factor and its receptor are differentially modified by chronic naltrexone treatment during rat brain development

    Neuroscience Letters

    (1993)
  • R.D. Polakiewicz et al.

    μ-Opioid receptor activates signaling pathways implicated in cell survival and translational control

    Journal of Biological Chemistry

    (1998)
  • M. Sakaguchi et al.

    Effects of opioids on neuronal survival in culture of embryonic chick dorsal root ganglion neurons

    Neuroscience Letters

    (1999)
  • W. Schmahl et al.

    Long-lasting effects of naltrexone, an opioid antagonist, on cell proliferation in developing rat forebrain

    Brain Research

    (1989)
  • A.E. Takemori et al.

    The irreversible narcotic antagonistic and reversible agonistic properties of the fumaramate methyl ester derivative of naltrexone

    European Journal of Pharmacology

    (1981)
  • C.J. Vlahos et al.

    A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002)

    Journal of Biological Chemistry

    (1994)
  • P. Xie et al.

    Activation of NF-κB by bradykinin through a Gαq- and Gβγ-dependent pathway that involves phosphoinositide 3-kinase and Akt

    Journal of Biological Chemistry

    (2000)
  • Cited by (70)

    • Evaluating the expression pattern of the opioid receptor in pituitary neuroendocrine tumors (PitNET) and the role of morphine and naloxone in the regulation of pituitary cell line growth and apoptosis

      2023, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      To assess the possible involvement of MOR in the morphine-induced effects in GH3 cells, naloxone was selected as a well-documented MOR antagonist and applied in the presence of morphine. Based on preliminary experiments performed in our laboratory and on existing data in the literature, the 10 and 25 µM naloxone were applied to inhibit MOR activity [18]. The concentration of 50 mM morphine and 50 mM naloxone was dissolved in DMSO as a stock solution and stored at − 20 °C.

    • Whether short peptides are good candidates for future neuroprotective therapeutics?

      2021, Peptides
      Citation Excerpt :

      Impressive results were obtained for the synthetic enkephalin derivative with high μ-opioid receptor affinity and selectivity, DAMGO (Tyr-d-Ala-Gly-N-MePhe-Gly-ol). This compound's potential protective effects on apoptotic cell death were tested using non-transfected fully differentiated SH-SY5Y cells (exposed to retinoic acid for five days) and primary culture of mouse cortical neurons [75]. In differentiated SH-SY5Y cells, DAMGO (0.1–2 μM) promoted cell survival after serum deprivation without inducing cell proliferation.

    • The effects of tramadol on cancer stem cells and metabolic changes in colon carcinoma cells lines

      2019, Gene
      Citation Excerpt :

      They concluded that different types of tumor response to morphine may occur regarding apoptosis. On the other hand, Iglesias et al. showed that morphine inhibits apoptosis in neuroblastoma cells (Igleisas et al., 2003). They stated that endogenous opioids may play a neurotrophic role during development in neuronal cells.

    • The proteins interacting with C-terminal of μ receptor are identified by bacterial two-hybrid system from brain cDNA library in morphine-dependent rats

      2015, Life Sciences
      Citation Excerpt :

      Endogenous opioid peptides and alkaloid opioid agonists affect nervous system development by promoting neuronal survival [39,40]. Morphine (0.1–10 μM) also has protective effects on serum withdrawal-induced cell death in non-transfected fully differentiated neuroblastoma SH-SY5Y cells [27]. There are also studies showing that chronic morphine treatment is associated with a remarkable opposite modulation of proteins in rat brain, which involve in the regulation of the programmed cell death [9].

    View all citing articles on Scopus
    View full text