Regular article
Opioid-induced proliferation through the MAPK pathway in cultures of adult hippocampal progenitors

https://doi.org/10.1016/S1044-7431(03)00061-7Get rights and content

Abstract

Administration of opioid agonists or antagonists has been reported to regulate proliferation or survival of neural progenitors in vivo. Here we report that β-endorphin and selective mu-opioid receptor (MOR) and delta-opioid receptor (DOR) agonists stimulate proliferation of isolated rat adult hippocampal progenitors (AHPs). The AHPs were found to express DORs and MORs, but not kappa-opioid receptors. Incubation with β-endorphin for 48 h increased the number of AHPs found in mitosis, the total DNA content, and the expression of proliferating cell nuclear antigen. This proliferative effect from β-endorphin on AHPs was antagonized by naloxone. The β-endorphin-induced proliferation was mediated through phosphorylation of extracellular signal-regulated kinases 1 and 2 and dependent on phosphatidylinositol 3-kinase and both intra- and extracellular calcium. These data suggest a role for the opioid system in the regulation of proliferation in progenitors from the adult hippocampus.

Introduction

Neurogenesis has been shown to occur in several species, including humans Altman and Das 1965, Nottebohm 1985, Stanfield and Trice 1988, Kempermann et al 1997, Eriksson et al 1998. The birth of new cells in the dentate gyrus of the hippocampus, a brain region important for learning and memory, is decreased by stress (Gould et al., 1998) and age (Cameron and McKay, 1999) and increased by hippocampal-dependent learning (Gould et al., 1999), environmental enrichment (Nilsson et al., 1999), and voluntary exercise van Praag et al 1999, van Praag et al 2000. Chronic exposure to morphine, widely used for treatment of pain, has been shown to result in impaired memory Saha et al 1991, Li et al 2001 and also to decrease neurogenesis and the survival of progenitors in the dentate gyrus of the adult rat hippocampus (Eisch et al., 2000). Several reports suggest a negative effect in vivo of morphine on DNA synthesis in the neonatal and adult brain Kornblum et al 1987, Hauser et al 1994, Eisch et al 2000 and a positive effect of opioid antagonists on this process Zagon and McLaughlin 1986, Schmahl et al 1989, Hauser et al 1989, Seatriz and Hammer 1993. By contrast, in an early in vivo study, DNA synthesis has been seen to increase after acute administration of morphine to adult rats (Miller et al., 1982).

In vitro, both proliferation of cerebellar granule neuron precursors (Hauser et al., 2000) and astrocytes (Hauser et al., 1996) were decreased after exposure to morphine, whereas the opioid antagonist naloxone has been shown to decrease DNA synthesis of O4-immunoreactive oligodendrocyte progenitors in glial cultures (Hauser et al., 1993). The inhibitory effect of naloxone was not found in cultures enriched with oligodendrocyte progenitors and fewer astrocytes. Furthermore, the mu-opioid receptor (MOR) agonist PL017 increased DNA synthesis in immature O4-immunoreactive oligodendrocytes in vitro (Knapp et al., 1998). In this study, opposite effects of opiates on proliferation were seen in the immature O4-immunoreactive oligodendrocytes compared with mature myelin basic protein (MBP)-immunoreactive oligodendrocytes in which proliferation was down-regulated. Furthermore, in cultured astrocytes the increase or decrease in proliferation is depending on extracellular levels of calcium (Hauser et al., 1996). This suggests a complex regulation of proliferation in neural cell types. The response from opioids on proliferation seems to depend on the specific cell type studied and whether the cell is at an immature or a mature stage. Other influencing factors include the type of surrounding cells, the calcium level, what opioid receptor subtypes are expressed on the cells, and possibly several other unknown factors.

All three opioid receptor subtypes, delta-opioid receptors (DORs), MORs, and kappa-opioid receptors (KORs), have been found in the dentate gyrus of the adult rat hippocampus at both the protein and the mRNA levels Mansour et al 1987, McLean et al 1987, Delfs et al 1994. β-Endorphin, an endogenous opioid peptide derived from the proopiomelanocortin gene, is expressed in both the embryonal (Angelogianni et al., 2000) and the adult Barna et al 1997, Johansson et al 1997 rat hippocampus.

To better understand whether endogenous opioids have direct effects on the proliferation of adult neural progenitors, we used adult hippocampal progenitors (AHPs) isolated from rat hippocampus Gage et al 1995, Palmer et al 1999. These cells are capable of generating neurons as well as glia Palmer et al 1997, Takahashi et al 1999, Aberg et al 2001 and have been transplanted back to the adult hippocampus, where they retain the capacity to become new granule cell layer neurons Gage et al 1995, van Praag et al 2002.

In this study we wanted to determine whether any opioid receptor subtype exists on the AHPs and, if so, whether they mediate effects from the endogenous opioid peptide β-endorphin or specific opioid receptor agonists. Total DNA content and the expression of the cell-cycle markers, phosphorylated histone H3 (phospho-histone H3), and proliferating cell nuclear antigen (PCNA), were used to determine opioid regulation of AHP proliferation. Previous studies have shown that stimulation of opioid receptors increases the level of free intracellular calcium ([Ca2+]i) Stiene-Martin et al 1998, Thorlin et al 1998, a signal transducer known to be involved in regulation of proliferation (Hauser et al., 1996). Opioids also increase phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2; also called mitogen-activated protein kinase (MAPK)) Gutstein et al 1997, Kramer and Simon 2000, Belcheva et al 2001. Inhibited phosphorylation of MAPK has previously been shown to block proliferation of neural progenitor cells, suggesting that this pathway is important for proliferation of neural progenitors (Learish et al., 2000). Therefore, we also wanted to study whether β-endorphin regulates phosphorylation of ERK1/2 and the levels of [Ca2+]i in the AHPs.

Section snippets

Visualization of DORs and MORs but not KORs on adult hippocampal progenitors in vitro

Immunohistochemically, MORs (Fig. 1A) and DORs (Fig. 1B) are visualized on AHPs in vitro. No immunoreactivity for KORs was found (Fig. 1C). Similarly, no immunoreactivity for MORs or DORs was seen when antisera against these receptors were preincubated with control peptides (Figs. 1D and E) or when normal rabbit serum was used instead of antiserum against any opioid receptor (Fig. 1F). Immunoblotting, using total protein prepared from cultured AHPs, showed the expected molecular weights at 45

Discussion

Opioid receptors have been found in neurons, astrocytes, and oligodendrocytes Eriksson et al 1990, Knapp et al 1998, Stiene-Martin et al 1998, Stiene-Martin et al 2001, Thorlin et al 1999, Persson et al 2000. Activation of these receptors plays different roles in these cells, such as inhibition of transmitter release (Nicol et al., 1996), inhibition of induced cyclic adenosine monophosphate (cAMP) levels (Eriksson et al., 1992), and release of [Ca2+]i Stiene-Martin et al 1998, Thorlin et al 1998

Cell culture

The isolation of AHPs has previously been described (Palmer et al., 1997). The clonal population was received at passage 4 as a gift from Dr. Fred Gage (Laboratory of Genetics, The Salk Institute, La Jolla, CA). In brief, hippocampi were bilaterally dissected from adult (>3 months) female Fischer 344 rats. Thereafter, tissue was washed in cold sterile Dulbecco’s phosphate-buffered saline, mechanically dissociated, and treated with enzymes (0.01% papain, 0.1% neutral protease). The cells were

Acknowledgements

We thank Kjell Pettersson of the Institute of Statistics, Göteborg University, for help with the statistics. The work was supported by grants from the Swedish Medical Research Council (Project 12X-12535), the Medical Faculty of Göteborg University, the Swedish Society of Medicine, the Swedish Society for Medical Research, John och Brit Wennerström’s Foundation for Neurological Research, Edit Jacobsson’s foundation, and Tore Nilson’s foundation.

References (74)

  • P.S. Eriksson et al.

    Opiate receptors in neuronal primary cultures

    Neuropharmacology

    (1990)
  • D. Guerra et al.

    Neuropsychological performance in opiate addicts after rapid detoxification

    Drug Alcohol Depend.

    (1987)
  • K.F. Hauser et al.

    Morphine inhibits Purkinje cell survival and dendritic differentiation in organotypic cultures of the mouse cerebellum

    Exp. Neurol.

    (1994)
  • K.F. Hauser et al.

    mu-Opioid receptor-induced Ca2+ mobilization and astroglial developmentmorphine inhibits DNA synthesis and stimulates cellular hypertrophy through a Ca(2+)-dependent mechanism

    Brain Res.

    (1996)
  • M. Heilig

    Cerebrospinal fluid neuropeptides in Alzheimer’s disease and vascular dementia

    Biol. Psychiatry

    (1995)
  • P. Johansson et al.

    Anabolic androgenic steroids increase beta-endorphin levels in the ventral tegmental area in the male rat brain

    Neurosci. Res.

    (1997)
  • H.I. Kornblum et al.

    Effects of morphine on DNA synthesis in neonatal rat brain

    Brain Res.

    (1987)
  • H.K. Kramer et al.

    mu and delta-opioid receptor agonists induce mitogen-activated protein kinase (MAPK) activation in the absence of receptor internalization

    Neuropharmacology

    (2000)
  • R.D. Learish et al.

    Inhibition of mitogen-activated protein kinase kinase blocks proliferation of neural progenitor cells

    Brain Res. Dev. Brain Res.

    (2000)
  • Z. Li et al.

    Reversal of morphine-induced memory impairment in mice by withdrawal in Morris water mazepossible involvement of cholinergic system

    Pharmacol. Biochem. Behav.

    (2001)
  • M.J. Marinissen et al.

    G-protein-coupled receptors and signaling networksemerging paradigms

    Trends Pharmacol. Sci.

    (2001)
  • A.M. Mathieu-Kia

    Mu-, delta- and kappa-opioid receptor populations are differentially altered in distinct areas of postmortem brains of Alzheimer’s disease patients

    Brain Res.

    (2001)
  • M.J. Millan et al.

    The endocrinology of the opioids

    Int. Rev. Neurobiol.

    (1985)
  • B. Nicol et al.

    mu- and kappa-opioids inhibit K+ evoked glutamate release from rat cerebrocortical slices

    Neurosci. Lett.

    (1996)
  • M.A. Nugent et al.

    Fibroblast growth factor-2

    Int. J. Biochem. Cell Biol.

    (2000)
  • T.D. Palmer et al.

    The adult rat hippocampus contains primordial neural stem cells

    Mol. Cell. Neurosci.

    (1997)
  • W. Schmahl et al.

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

    Brain Res.

    (1989)
  • J.V. Seatriz et al.

    Effects of opiates on neuronal development in the rat cerebral cortex

    Brain Res. Bull.

    (1993)
  • A. Stiene-Martin et al.

    Morphine alters astrocyte growth in primary cultures of mouse glial cellsevidence for a direct effect of opiates on neural maturation

    Brain Res. Dev. Brain Res.

    (1991)
  • P. Taupin et al.

    FGF-2-responsive neural stem cell proliferation requires CCg, a novel autocrine/paracrine cofactor

    Neuron

    (2000)
  • T. Thorlin et al.

    Delta-opioid receptors on astroglial cells in primary culturemobilization of intracellular free calcium via a pertussis sensitive G protein

    Neuropharmacology

    (1998)
  • N. Trapaidze et al.

    Opioid receptor endocytosis and activation of MAP kinase pathway

    Brain Res. Mol. Brain Res.

    (2000)
  • P. Tryoen-Toth et al.

    Regulation of kappa-opioid receptor mRNA level by cyclic AMP and growth factors in cultured rat glial cells

    Brain Res. Mol. Brain Res.

    (1998)
  • D. Uberti et al.

    p53 is dispensable for apoptosis but controls neurogenesis of mouse dentate gyrus cells following gamma-irradiation

    Brain Res. Mol. Brain Res.

    (2001)
  • I.S. Zagon et al.

    Opioid antagonist-induced modulation of cerebral and hippocampal developmenthistological and morphometric studies

    Brain Res.

    (1986)
  • J. Altman et al.

    Autoradiographic and histological evidence of postnatal hippocampal neurogenesis in rats

    J. Comp. Neurol.

    (1965)
  • P. Angelogianni et al.

    Ontogenesis of proopiomelanocortin and its processing to beta-endorphin by the fetal and neonatal rat brain

    Neuroendocrinology

    (2000)
  • Cited by (73)

    • Receptors: Opioid receptors

      2021, Encyclopedia of Biological Chemistry: Third Edition
    • Morphine increases macrophages at the lesion site following spinal cord injury: Protective effects of minocycline

      2019, Brain, Behavior, and Immunity
      Citation Excerpt :

      Studies suggest that morphine may induce microglial migration via a mechanism involving the ionotropic purinergic receptor P2X4, which has also been implicated in aberrant glial activation and pathological pain (Biber et al., 2011; Horvath and DeLeo, 2009; Inoue et al., 2004; Tsuda et al., 2003). Alternatively, opioid-induced proliferation has been observed in hippocampal neural progenitors, vascular endothelial cells, and peripheral blood mononuclear cells (Hutchinson et al., 2004; Leo et al., 2009; Persson et al., 2003). It is possible that the increase in microglia observed in our study could also be due to increased proliferation stimulated by morphine treatment.

    • Effect of delta opioid receptor activation on spatial cognition and neurogenesis in cerebral ischemic rats

      2016, Neuroscience Letters
      Citation Excerpt :

      Many studies have focused on the mechanisms of adult neurogenesis and how interventions designed to modulate these processes could enhance endogenous neurogenesis following ischemia [9]. Persson et al., showed that DOR agonists stimulate proliferation of adult hippocampal progenitors in vitro [24], and DOR antagonists decrease proliferation in the same situation [25]. In our present study, we also found that ischemic injury indeed increased BrdU positive cells in the CA1 and DG regions of the hippocampus in rats 7 days after global ischemia, while DADLE further increased BrdU positive cells in both regions compared to I/R rats, and NAL blocked DADLE’s effects.

    View all citing articles on Scopus
    View full text