Elsevier

Neurotoxicology and Teratology

Volume 86, July–August 2021, 107002
Neurotoxicology and Teratology

Endogenous and exogenous opioid effects on oligodendrocyte biology and developmental brain myelination

https://doi.org/10.1016/j.ntt.2021.107002Get rights and content

Highlights

  • High levels of opioid receptors and ligands in the developing brain suggest maturational functions still poorly understood.

  • Opioid use rates underscore the need for identifying those functions and their roles in normal and pathological conditions.

  • Findings support significant opioid effects on developmental oligodendrocyte differentiation and brain myelination.

  • Exogenous opioids may alter the timing of developmental brain myelination disrupting its coordination with axonal outgrowth.

Abstract

The elevated presence of opioid receptors and their ligands throughout the developing brain points to the existence of maturational functions of the endogenous opioid system that still remain poorly understood. The alarmingly increasing rates of opioid use and abuse underscore the urgent need for clear identification of those functions and the cellular bases and molecular mechanisms underlying their physiological roles under normal and pathological conditions. This review is focused on current knowledge on the direct and indirect regulatory roles that opioids may have on oligodendrocyte development and their generation of myelin, a complex insulating membrane that not only facilitates rapid impulse conduction but also participates in mechanisms of brain plasticity and adaptation. Information is examined in relation to the importance of endogenous opioid function, as well as direct and indirect effects of opioid analogues, which like methadone and buprenorphine are used in medication-assisted therapies for opioid addiction during pregnancy and pharmacotherapy in neonatal abstinence syndrome. Potential opioid effects are also discussed regarding late myelination of the brain prefrontal cortex in adolescents and young adults. Such knowledge is fundamental for the design of safer pharmacological interventions for opioid abuse, minimizing deleterious effects in the developing nervous system.

Introduction

Opioid abuse and misuse continues to represent a problem of major epidemic proportions. This is particularly alarming when considering the large number of babies exposed to opioids during pregnancy, and in addition, the significant percentage of these infants that require opioids after birth for the pharmacological treatment of neonatal abstinence syndrome (NAS) (Honein et al., 2019). Newborns affected by NAS exhibit different symptoms of variable magnitude that may include central nervous system dysfunction reflected in tremors and seizures, inconsolable crying, excessive irritability, poor sleep, and elevated muscle tone; as well as autonomic nervous system effects resulting in various digestive and respiratory problems and altered temperature regulation (Finnegan et al., 1975; Desai et al., 2015; Hoegerman et al., 1990; Jones et al., 2016; Gomez-Pomar and Finnegan, 2018). Opioids and their metabolites have the capacity of crossing the placenta (Nekhayeva et al., 2005; Krishna et al., 1997; Malek and Mattison, 2011) and blood-brain barrier (Bostrom et al., 2008; Bourasset et al., 2003). Thus, NAS symptoms are logically considered to be the result of abrupt discontinuation of maternal opioid supply after birth. Current successful and necessary medication-assisted therapies for opioid addiction during pregnancy involve the administration of the synthetic long-lasting opioid analogue and full mu-opioid receptor agonist methadone; and more recently, buprenorphine, a partial mu-opioid receptor agonist and kappa-opioid receptor antagonist that not only successfully prevents the maternal abuse of opioids but also exhibits higher efficacy than methadone in reducing the incidence and severity of NAS (Fischer et al., 2006; Lejeune et al., 2006; Welle-Strand et al., 2013; Minozzi et al., 2013; Jones et al., 2010). However, an increasing number of reports suggest that some of these opioid-based therapeutic approaches may also exert neurodevelopmental effects. While much is known about opioids and their role in pain regulation, the high expression levels of different opioid receptors and their endogenous opioid ligands throughout the developing brain point to the existence of maturational functions that still remain poorly understood. This raises the question of whether interference with the endogenous opioid system by exogenous opioids, including those used in pharmacotherapy treatments, could also alter important developmental brain processes. A recent large prospective study in which potential effects of other drug co-exposures and compounding factors were carefully controlled and periodically monitored, concluded that gestational opioid exposure sufficient to result in NAS also increased the proportion of neonates with reduced head circumference (Towers et al., 2019). Notably, the great majority of those infants were born from mothers that were maintained under methadone or buprenorphine treatment. The mechanisms behind these effects remain poorly understood but different findings suggest the possibility of opioid actions on different neural cell types. For example, animal models showed that perinatal methadone exposure alters the function of dopaminergic, noradrenergic and serotonergic neurons in the neonatal and early postnatal period (Rech et al., 1980). Furthermore, while human effects are difficult to evaluate, studies using cultured human cortical organoids indicated methadone suppressive actions on neuronal function and maturation (Wu et al., 2020a). Different lines of evidence also point to potential opioid roles on various glial cell populations (Hauser and Knapp, 2017). This review is focused on the neurodevelopmental effects that opioids may have on brain oligodendrocytes and their synthesis of myelin, the remarkably complex multilamellar structure that not only facilitates the rapid “saltatory conduction” of nerve impulses (Castelfranco and Hartline, 2015) but is also now recognized as a crucial player in brain plasticity and in active bidirectional neuron-glial communications (Fields, 2008; Hasan et al., 2019; Xiao et al., 2016). As such, oligodendrocyte generation and myelin formation are among the most critical and vulnerable processes that take place during brain development.

Section snippets

Developmental oligodendrocyte generation and brain myelin formation as direct targets of endogenous and exogenous opioids

Oligodendrocytes are generated from bipolar highly proliferative and migratory progenitor cells that experience several distinct stages of differentiation prior to their transformation into quiescent and morphologically complex multipolar cells capable of myelin formation (Baumann and Pham-Dinh, 2001). Importantly, each of these mature oligodendrocytes has the remarkable capacity of generating multiple extensive membrane extensions that contact numerous neurons and concentrically wrap around

Oligodendrocyte maturation and developmental myelination as secondary targets of opioid effects

The previous section of this review focused on physiological functions of endogenous opioid systems in oligodendrocyte development and the consequences of their direct interference by exogenous opioids. However, as a logical reflection of the multiple roles and integrative functions of oligodendrocytes and myelin, important consideration should also be given to potential indirect effects mediated by primary opioid actions on other diverse cell targets. Understanding of such secondary effects

Discussion

The findings described in this review point to significant opioid modulatory effects on perinatal oligodendrocyte differentiation and brain myelination. Cell culture studies and animal models of perinatal opioid exposure suggest that a complex balance between opposing effects of the mu-opioid- and nociceptin receptor activities control the precise timing of oligodendrocyte maturation and myelinating activity (Eschenroeder et al., 2012; Mohamed et al., 2020). Such balance may play and important

Declaration of Competing Interest

The authors state no conflict of interest.

Acknowledgements

Some of the studies reported in this review were supported by grants R21DA027099-01 from the National Institutes of Health, RG 15012891 from the National Multiple Sclerosis Society, and a sub-award from NIH CTSA grant UL1TR00058 from the Virginia Commonwealth University (VCU) Center for Clinical and Translational Research.

References (131)

  • J.M. Fuster

    The prefrontal cortex--an update: time is of the essence

    Neuron

    (2001)
  • H.Z. Guo et al.

    The effect of prenatal exposure to methadone on neurotransmitters in neonatal rats

    Dev. Brain Res.

    (1990)
  • T. Ishibashi et al.

    Astrocytes promote myelination in response to electrical impulses

    Neuron

    (2006)
  • L.L. Jantzie et al.

    Prenatal opioid exposure: the next neonatal neuroinflammatory disease

    Brain Behav. Immun.

    (2020)
  • P.E. Knapp et al.

    mu-Opioid receptor activation enhances DNA synthesis in immature oligodendrocytes

    Brain Res.

    (1996)
  • C. Lejeune et al.

    Prospective multicenter observational study of 260 infants born to 259 opiate-dependent mothers on methadone or high-dose buprenophine substitution

    Drug Alcohol Depend.

    (2006)
  • J. Liu et al.

    Epigenetic control of oligodendrocyte development: adding new players to old keepers

    Curr. Opin. Neurobiol.

    (2016)
  • I. Lizasoain et al.

    Buprenorphine: bell-shaped dose-response curve for its antagonist effects

    Gen. Pharmacol.

    (1991)
  • K. Mahon et al.

    A role for white matter abnormalities in the pathophysiology of bipolar disorder

    Neurosci. Biobehav. Rev.

    (2010)
  • S. Merighi et al.

    Morphine mediates a proinflammatory phenotype via mu-opioid receptor-PKCvarepsilon-Akt-ERK1/2 signaling pathway in activated microglial cells

    Biochem. Pharmacol.

    (2013)
  • V.J. Monnelly et al.

    Prenatal methadone exposure is associated with altered neonatal brain development

    Neuroimage Clin.

    (2018)
  • B.M. Morrison et al.

    Oligodendroglia: metabolic supporters of axons

    Trends Cell Biol.

    (2013)
  • I.A. Nekhayeva et al.

    Bidirectional transfer of methadone across human placenta

    Biochem. Pharmacol.

    (2005)
  • E. Nygaard et al.

    Neuroanatomical characteristics of youths with prenatal opioid and poly-drug exposure

    Neurotoxicol. Teratol.

    (2018)
  • D. Qiu et al.

    Diffusion tensor imaging of normal white matter maturation from late childhood to young adulthood: voxel-wise evaluation of mean diffusivity, fractional anisotropy, radial and axial diffusivities, and correlation with reading development

    Neuroimage

    (2008)
  • M.N. Rasband et al.

    Developmental clustering of ion channels at and near the node of Ranvier

    Dev. Biol.

    (2001)
  • K. Rekik et al.

    Activation of nociceptin/orphanin FQ receptors inhibits contextual fear memory reconsolidation

    Neuropharmacology

    (2017)
  • W.D. Richardson et al.

    A role for platelet-derived growth factor in normal gliogenesis in the central nervous system

    Cell

    (1988)
  • S.E. Robinson et al.

    Prenatal exposure to methadone delays the development of striatal cholinergic neurons

    Brain Res. Dev. Brain Res.

    (1993)
  • S.E. Robinson et al.

    Postnatal methadone exposure does not prevent prenatal methadone-induced changes in striatal cholinergic neurons

    Brain Res. Dev. Brain Res.

    (1996)
  • D. Avey et al.

    Single-cell RNA-seq uncovers a robust transcriptional response to morphine by Glia

    Cell Rep.

    (2018)
  • N. Barnea-Goraly et al.

    White matter development during childhood and adolescence: a cross-sectional diffusion tensor imaging study

    Cereb. Cortex

    (2005)
  • B.A. Barres et al.

    A crucial role for neurotrophin-3 in oligodendrocyte development

    Nature

    (1994)
  • N. Baumann et al.

    Biology of oligodendrocyte and myelin in the mammalian central nervous system

    Physiol. Rev.

    (2001)
  • S.L. Bengtsson et al.

    Extensive piano practicing has regionally specific effects on white matter development

    Nat. Neurosci.

    (2005)
  • E. Bostrom et al.

    Blood-brain barrier transport helps to explain discrepancies in in vivo potency between oxycodone and morphine

    Anesthesiology

    (2008)
  • F. Bourasset et al.

    Evidence for an active transport of morphine-6-beta-d-glucuronide but not P-glycoprotein-mediated at the blood-brain barrier

    J. Neurochem.

    (2003)
  • P. Brambilla et al.

    White matter connectivity in bipolar disorder

    Int Rev Psychiatry

    (2009)
  • K.D. Buono et al.

    Leukemia inhibitory factor is essential for subventricular zone neural stem cell and progenitor homeostasis as revealed by a novel flow cytometric analysis

    Dev. Neurosci.

    (2012)
  • E.M. Byrnes

    Transgenerational consequences of adolescent morphine exposure in female rats: effects on anxiety-like behaviors and morphine sensitization in adult offspring

    Psychopharmacology

    (2005)
  • A.M. Castelfranco et al.

    The evolution of vertebrate and invertebrate myelin: a theoretical computational study

    J. Comput. Neurosci.

    (2015)
  • C.C. Chao et al.

    Kappa opioid receptors in human microglia downregulate human immunodeficiency virus 1 expression

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

    (1996)
  • C.Y. Chen et al.

    Heroin-induced spongiform leukoencephalopathy: value of diffusion MR imaging

    J. Comput. Assist. Tomogr.

    (2000)
  • L.J. Chew et al.

    Oligodendroglial alterations and the role of microglia in white matter injury: relevance to schizophrenia

    Dev. Neurosci.

    (2013)
  • R.P. Coelho et al.

    Neurotrophin-3 targets the translational initiation machinery in oligodendrocytes

    Glia

    (2009)
  • E. Conradt et al.

    Prenatal opioid exposure: neurodevelopmental consequences and future research priorities

    Pediatrics

    (2019)
  • S.C. Deoni et al.

    White matter maturation profiles through early childhood predict general cognitive ability

    Brain Struct. Funct.

    (2016)
  • R.J. Desai et al.

    Exposure to prescription opioid analgesics in utero and risk of neonatal abstinence syndrome: population based cohort study

    BMJ

    (2015)
  • S.D. D’Souza et al.

    Ciliary neurotrophic factor selectively protects human oligodendrocytes from tumor necrosis factor-mediated injury

    J. Neurosci. Res.

    (1996)
  • J.E. Dum et al.

    In vivo receptor binding of the opiate partial agonist, buprenorphine, correlated with its agonistic and antagonistic actions

    Br. J. Pharmacol.

    (1981)
  • Cited by (11)

    • Effects of prenatal opioid exposure on synaptic adaptations and behaviors across development

      2023, Neuropharmacology
      Citation Excerpt :

      These studies suggest that adolescent ethanol exposure results in long-lasting neurobehavioral dysfunction into aging. Although there are some review articles on the effects of exposure to opioids across specific developmental ages (such as prenatal or adolescent opioid exposures) (Grecco and Atwood, 2020; Velasco et al., 2021; Windisch and Kreek, 2020; Rahman et al., 2022; Boggess and Risher, 2022), there are limited comparative reviews on sex- and age-dependent effects of opioids on synaptic plasticity and opioid-related behavioral deficits (Conradt et al., 2018). There is emerging evidence for the interaction of sex on POE outcomes in clinical studies.

    • Prolonged NoGo P3 latency as a possible neurobehavioral correlate of aggressive and antisocial behaviors: A Go/NoGo ERP study

      2022, Biological Psychology
      Citation Excerpt :

      Decreased rates of myelination during adolescence have also been associated with an increased expression of psychopathology later in life (Vanes et al., 2020). Substance use and impulse control problems, both closely related to aggressive and antisocial behaviors (Alcorn et al., 2013; Krueger, Markon, Patrick, Benning, & Kramer, 2007), may also play a role here, since the use of alcohol (Bjork, Grant, & Hommer, 2003; McQueeny et al., 2009), opioids (Velasco, Mohamed, & Sato-Bigbee, 2021), cannabis (Bava et al., 2009), and cocaine (Bartzokis et al., 2002) is toxic to the vulnerable myelination process, and may in turn exacerbate already existing impulse control problems and contribute to poor outcomes in individuals prone to substance abuse (Bartzokis, 2005). While speculative, it is possible that aggressive and antisocial individuals exhibit aberrant myelination, which in turn could manifest as decreased NoGo P3 latency.

    View all citing articles on Scopus
    1

    Present address: Department of Neurological Surgery, University of California San Francisco, CA 94158, USA.

    View full text