Review
The “Toll” of Opioid-Induced Glial Activation: Improving the Clinical Efficacy of Opioids by Targeting Glia

https://doi.org/10.1016/j.tips.2009.08.002Get rights and content

Glial activation participates in the mediation of pain including neuropathic pain, due to release of neuroexcitatory, proinflammatory products. Glial activation is now known to occur in response to opioids as well. Opioid-induced glial activation opposes opioid analgesia and enhances opioid tolerance, dependence, reward and respiratory depression. Such effects can occur, not via classical opioid receptors, but rather via non-stereoselective activation of toll-like receptor 4 (TLR4), a recently recognized key glial receptor participating in neuropathic pain as well. This discovery identifies a means for separating the beneficial actions of opioids (opioid receptor mediated) from the unwanted side-effects (TLR4/glial mediated) by pharmacologically targeting TLR4. Such a drug should be a stand-alone therapeutic for treating neuropathic pain as well. Excitingly, with newly-established clinical trials of two glial modulators for treating neuropathic pain and improving the utility of opioids, translation from rats-to-humans now begins with the promise of improved clinical pain control.

Introduction

Normally, a painful stimulus is perceived via a chain of events beginning with the activation of “pain”-receptive sensory nerve fibers. The resultant action potentials relay information of potential or actual tissue injury to pain transmission neurons in the spinal cord dorsal horn. These, in turn, send the information to multiple sites within the brain where various aspects of the pain experience (sensation, analysis of meaning, emotional reactions, etc) are analyzed and responded to. However, pain processing is not a passive process but rather is under powerful modulatory control. Pain messages can be suppressed by drugs like morphine, relayed unaltered, or amplified under conditions such as chronic pain. When chronic pain develops as a result of peripheral nerve injury, for example, these conditions have typically been attributed to a variety of neuronal changes, including altered excitability of sensory neurons, alterations in which neurotransmitters are synthesized and released by various sensory neurons, alterations in pain transmission neuron excitability via multiple changes in receptor and ion channel functions, and so on [1].

Intriguingly, powerful modulatory control exists not only for pain, but also for an organism's responses to opioids, such as morphine. Opioids not only suppress pain, they also activate endogenous counter-regulatory mechanisms that, for example, actively oppose opioid-induced pain suppression, enhance analgesic tolerance wherein repeated opioids lose their ability to suppress pain, and enhance dependence wherein organisms require continued opioid exposure to stave off drug withdrawal. These modulatory controls have again been attributed to a variety if neuronal mechanisms, including release of endogenous anti-opioid peptides such as cholecystokinin, internalization and/or desensitization of opioid receptors, alterations in opioid receptor signaling cascades, and so on 2, 3.

While modulatory control systems regulating pain and opioid actions have been thought to involve separate mechanisms, the present paper will review recent evidence that suggests that these two phenomena are closely related and involve overlapping mechanisms. This development has been predicted from prior studies focused on the neuronal bases of chronic pain and opioid tolerance, where striking commonalities such as upregulation of NMDA function transcend what initially appeared to be quite different phenomena [4]. The present review will extend the commonalities between chronic pain and various opioid effects to include a non-neuronal component (glial cells, especially microglia and astrocytes) and a distinctly non-traditional mechanism, that being activation of the innate immune receptor expressed by glia, called toll like receptor 4 (TLR4).

Concepts of chronic pain and opioid actions have evolved in recent years with the realization that alterations in neuronal functions fail to capture all of the critical mechanisms involved. Recognition of a role for microglia and astrocytes in these processes first occurred for pain in the early 1990 s, but the involvement of glia in modulating opioid actions was not discovered until a decade later. Indeed, the growing recognition of striking similarities in mechanisms underlying chronic pain and opioid tolerance directly led to the discovery of glial involvement in modulating opioid actions. Since glia were so convincingly important in chronic pain, it became a natural question whether they regulated the actions of opioids as well.

The goals of this article are to explore how glial activation impacts both pain and opioid actions. Pain will be considered first, including how glial activation increases neuronal excitability and how glial activation occurs under conditions leading to chronic pain. Included in this latter topic will be a discussion of TLR4 as a key glial activation receptor for the initiation and maintenance of chronic pain via TLR4-induced release of neuroexcitatory products such as proinflammatory cytokines. TLR4 will also be discussed as having a unique role in regulating the actions of opioids, as opioids have now been found to activate TLR4 on glia, in addition to their classical actions occurring through neuronal opioid receptors. Glial activation by opioids is an important phenomenon to understand, as glial activation opposes opioid analgesia and enhances opioid tolerance, dependence, reward and other negative side effects such as respiratory depression. Lastly, the implications of such glial activation will be considered for drug development aimed at improving both clinical pain control and clinical efficacy of opioids.

Section snippets

Role of glial activation in pain enhancement

The conclusion that the activation of microglia and astrocytes are critical to pain enhancement arose from 3 independent lines of study [5]: (a) cell culture studies showing that spinal cord is one of the rare CNS sites where astrocytes are activated in response to substance P, providing the first evidence that spinal cord glia are responsive to “pain” neurotransmitters; (b) anatomy studies that recognized that microglia and astrocytes each upregulate their expression of so-called activation

Beyond pathological pain: glial modulation of opioid actions

In the past decade, a series of discoveries have revised our views of the pharmacological actions of opioids. Since 2001, several laboratories have reported that glia become activated in response to opioids and this glial activation leads to the release of proinflammatory products, including proinflammatory cytokines 27, 28, 29. In vivo, opioid-induced glial activation has been inferred from (Table 2): (a) morphine-induced upregulation of microglial and astrocytic activation markers 30, 31, (b)

TLR4 and glial activation: implications for drug development

Gaining insights into glial modulation of pain and opioid effects in humans has been constrained by the challenges of assaying glial activation and glial products within the CNS. In rodents, positron emission tomography allows analyses of glial activation through visualizing uptake of labeled fluoroacetate (metabolic inhibitor specific to the Krebs cycle in glia) [50] or labeled ligands of the translocator protein (TSPO; formerly known as the peripheral benzodiazepine receptor), the

Concluding remarks

As reviewed above, while glia in their basal state play important roles in maintaining the health and normal functioning of the nervous system, their inappropriate proinflammatory activation concurrent with chronic pain pathologies and opioid administration can dramatically amplify pain and detrimentally alter the actions of opioids. While proinflammatory responses by glia can be important for inducing resolution of CNS immune challenges, neuroprotection, and repair, under conditions of chronic

Acknowledgements

International Association for the Study of Pain International Collaborative grant, American Australian Association Merck Company Foundation Fellowship, National Health and Medical Research Council CJ Martin Fellowship (ID 465423;M.R.H.) and NIH Grants DA015642, DA017670, DA024044, and DE017782. This work was partially supported by the by the NIH Intramural Research Programs of the National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism. We thank Avigen for

References (115)

  • L.R. Watkins

    Glia: novel counter-regulators of opioid analgesia

    Trends Neurosci

    (2005)
  • Y. Cui

    Activation of p38 mitogen-activated protein kinase in spinal microglia mediates morphine antinociceptive tolerance

    Brain Res

    (2006)
  • M.R. Hutchinson

    Reduction of opioid withdrawal and potentiation of acute opioid analgesia by systemic AV411 (ibudilast)

    Brain Behav Immun

    (2009)
  • Y. Cui

    A novel role of minocycline: attenuating morphine antinociceptive tolerance by inhibition of p38 MAPK in the activated spinal microglia

    Brain Behav Immun

    (2008)
  • Y. Shavit

    Interleukin-1 antagonizes morphine analgesia and underlies morphine tolerance

    Pain

    (2005)
  • H.E. Wu

    Stereoselective action of (+)-morphine over (-)-morphine in attenuating the (-)-morphine-produced antinociception via the naloxone-sensitive sigma receptor in the mouse

    Eur J Pharmacol

    (2007)
  • H.E. Wu

    dextro- and levo-morphine attenuate opioid delta and kappa receptor agonist produced analgesia in mu-opioid receptor knockout mice

    Eur J Pharmacol

    (2006)
  • A. Juni

    Nociception increases during opioid infusion in opioid receptor triple knock-out mice

    Neuroscience

    (2007)
  • Y. Li

    Morphine promotes apoptosis via TLR2, and this is negatively regulated by beta-arrestin 2

    Biochem Biophys Res Commun

    (2009)
  • M.M. Backonja

    Altered cytokine levels in the blood and cerebrospinal fluid of chronic pain patients

    J Neuroimmunol

    (2008)
  • L. Del Valle

    Spinal cord histopathological alterations in a patient with longstanding complex regional pain syndrome

    Brain Behav Immun

    (2009)
  • G.M. Alexander

    Changes in immune and glial markers in the CSF of patients with Complex Regional Pain Syndrome

    Brain Behav Immun

    (2007)
  • M. Weber

    Increased polysialic acid neural cell adhesion molecule expression in human hippocampus of heroin addicts

    Neuroscience

    (2006)
  • Q. Si

    Differential regulation of microglial activation by propentofylline via cAMP signaling

    Brain Res

    (1998)
  • R.X. Zhang

    Interleukin 1beta facilitates bone cancer pain in rats by enhancing NMDA receptor NR-1 subunit phosphorylation

    Neuroscience

    (2008)
  • S.M. Sweitzer

    Propentofylline attenuates vincristine-induced peripheral neuropathy in the rat

    Neurosci Lett

    (2006)
  • A. Ledeboer

    Intrathecal interleukin-10 gene therapy attenuates paclitaxel-induced mechanical allodynia and proinflammatory cytokine expression in dorsal root ganglia in rats

    Brain Behav Immun

    (2007)
  • J.P. Cata

    The effects of thalidomide and minocycline on taxol-induced hyperalgesia in rats

    Brain Res

    (2008)
  • G.T. Whiteside

    A role for cannabinoid receptors, but not endogenous opioids, in the antinociceptive activity of the CB2-selective agonist, GW405833

    Eur J Pharmacol

    (2005)
  • E. Sloane

    Anti-inflammatory cytokine gene therapy decreases sensory and motor dysfunction in experimental Multiple Sclerosis: MOG-EAE behavioral and anatomical symptom treatment with cytokine gene therapy

    Brain Behav Immun

    (2009)
  • A. Ledeboer

    Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation

    Pain

    (2005)
  • G. Wolf

    Interleukin-1 signaling is required for induction and maintenance of postoperative incisional pain: genetic and pharmacological studies in mice

    Brain Behav Immun

    (2008)
  • A.K. Clark

    Role of spinal microglia in rat models of peripheral nerve injury and inflammation

    Eur J Pain

    (2007)
  • J. Mika

    Minocycline and pentoxifylline attenuate allodynia and hyperalgesia and potentiate the effects of morphine in rat and mouse models of neuropathic pain

    Eur J Pharmacol

    (2007)
  • S.S. Padi et al.

    Minocycline prevents the development of neuropathic pain, but not acute pain: possible anti-inflammatory and antioxidant mechanisms

    Eur J Pharmacol

    (2008)
  • F. Marchand

    Effects of Etanercept and Minocycline in a rat model of spinal cord injury

    Eur J Pain

    (2009)
  • Y.S. Gwak

    Propentofylline attenuates allodynia, glial activation and modulates GABAergic tone after spinal cord injury in the rat

    Pain

    (2008)
  • C.S. Lin

    Chronic intrathecal infusion of minocycline prevents the development of spinal-nerve ligation-induced pain in rats

    Reg Anesth Pain Med

    (2007)
  • J. Liu

    Pentoxifylline attenuates the development of hyperalgesia in a rat model of neuropathic pain

    Neurosci Lett

    (2007)
  • S. Wang

    Regulation of the trigeminal NR1 subunit expression induced by inflammation of the temporomandibular joint region in rats

    Pain

    (2009)
  • T. Wei

    Pentoxifylline attenuates nociceptive sensitization and cytokine expression in a tibia fracture rat model of complex regional pain syndrome

    Eur J Pain

    (2009)
  • P. Song et al.

    The involvement of glial cells in the development of morphine tolerance

    Neurosci Res

    (2001)
  • J. Mika

    Attenuation of morphine tolerance by minocycline and pentoxifylline in naive and neuropathic mice

    Brain Behav Immun

    (2009)
  • E.D. Milligan et al.

    Pathological and protective roles of glia in chronic pain

    Nat Rev Neurosci

    (2009)
  • H. Ueda et al.

    Mechanisms underlying morphine analgesic tolerance and dependence

    Front Biosci

    (2009)
  • L.R. Watkins

    Potentiation of opiate analgesia and apparent reversal of morphine tolerance by proglumide

    Science

    (1984)
  • L.R. Watkins et al.

    Glia and pain: past, present and future

  • F. Wei

    Supraspinal glial-neuronal interactions contribute to descending pain facilitation

    J Neurosci

    (2008)
  • D. Davalos

    ATP mediates rapid microglial response to local brain injury in vivo

    Nat Neurosci

    (2005)
  • A.M. Binshtok

    Nociceptors are interleukin-1beta sensors

    J Neurosci

    (2008)
  • Cited by (322)

    • Temporomandibular Joint Surgery

      2023, Journal of Oral and Maxillofacial Surgery
    View all citing articles on Scopus
    View full text