Elsevier

NeuroToxicology

Volume 28, Issue 4, July 2007, Pages 813-818
NeuroToxicology

Oral administration of dextromethorphan does not produce neuronal vacuolation in the rat brain

https://doi.org/10.1016/j.neuro.2007.03.009Get rights and content

Abstract

Dextromethorphan is a widely used antitussive agent, also showing increased recreational abuse. Dextromethorphan and its metabolite dextrorphan are non-competitive antagonists at the N-methyl-d-aspartate (NMDA) receptor ion channel. Single doses of some NMDA receptor antagonists produce neuropathologic changes in neurons of the retrosplenial/posterior cingulate cortices (RS/PC), characterized by vacuolation or neurodegeneration. To determine whether dextromethorphan produces these characteristic lesions, dextromethorphan was administered orally either as a single dose of 120 mg/kg to female rats, or daily for 30 days at doses of 5–400 mg/(kg day) to male rats and 5–120 mg/(kg day) to female rats. Brains were examined microscopically for evidence of neuronal vacuolation (4–6 h postdose) and neurodegeneration (∼24 or 48 h postdose). Administration of dextromethorphan at 120 mg/(kg day) in females, and at ≥150 mg/(kg day) in males produced marked behavioral changes, indicative of neurologic effects. Mortality occurred at the highest doses administered. There were no detectable neuropathologic changes following single or repeated oral administration of dextromethorphan at any dose. Administration of MK-801 (9 mg/kg) produced both cytoplasmic vacuolation and neuronal degeneration in neurons of the RS/PC cortex. Thus characteristic neuropathologic changes found with more potent NMDA receptor antagonists do not occur following single or repeated oral administration of dextromethorphan.

Introduction

Dextromethorphan (3 methoxy-17-methylmorphinan monohydrate) is the d-isomer of levorphanol, a codeine analog. Dextromethorphan is used worldwide as an antitussive agent in cough and cold medications in at least 125 products. Because of its ready accessibility, recreational abuse of dextromethorphan has been increasing in recent years, although accurate statistics are not available.

Dextromethorphan and its primary metabolite dextrorphan act as channel blockers at the N-methyl-d-aspartate (NMDA) receptor and are generally classified as non-competitive, low affinity antagonists (Franklin and Murray, 1992, Roth et al., 1996). Dextromethorphan is rapidly demethylated in vivo to dextrorphan which has higher NMDA receptor binding affinity than dextromethorphan (Sun and Wessinger, 2004). Therefore dextrorphan likely contributes to the in vivo activity of dextromethorphan. Neither dextromethorphan nor dextrorphan has significant in vivo opioid activity.

Several NMDA receptor antagonists have been associated with a specific neuropathologic lesion following acute exposure in rats. The lesion (occasionally referred to as the “Olney lesion”) is characterized by neuronal vacuolation in the retrosplenial/posterior cingulated cortices (RS/PC) (Olney et al., 1989). The effect has been described for both non-competitive antagonists (ketamine, tiletamine, phencyclidine and MK-801) and the competitive antagonists (2-amino, 5-phosphonopentanoate [AP5], 3-(2-carboxypiperazin-4-yl)propyl-l-phosphonoic acid [CPP]) (Olney et al., 1989, Fix, 1994, Fix et al., 1995a, Fix et al., 1996, Fix et al., 2000, Auer and Coulter, 1994). The time-and dose-dependent nature of the lesion has been most extensively studied using the prototypical NMDA antagonist, MK-801. At low single doses neuronal vacuolation is readily reversible and limited to the RS/PC. Vacuolation is observed most prominently at 4–8 h after dosing, but is not apparent by 24 h (Olney et al., 1989). The vacuolar changes are also not sustained with repeated dosing (up to 4 days) of MK-801, suggesting the rapid development of cellular tolerance (Olney et al., 1989). At higher doses, irreversible neuronal necrosis occurs with associated reactive gliosis (Fix et al., 1995b), and disseminated changes involving additional corticolimbic structures (Horváth et al., 1997, Wozniak et al., 1998) have been described.

The neuropathologic effects of dextromethorphan and/or dextrorphan have not been well characterized. Therefore, the studies reported here were conducted to determine if oral administration of dextromethorphan to rats produces these characteristic findings after single or repeated administration at doses up to a maximally tolerated dose. Oral administration was used in these studies because this is the route of human therapeutic use and most common route of recreational abuse.

Section snippets

Animals

Crl:CD® (SD)IGS BR rats, obtained from Charles River Laboratories, Raleigh NC were used for all studies. The animals were 32–33 days old at receipt and were acclimated for 11–12 days prior to dosing. Animals were housed individually in room temperature of 72 ± 4 °F (22 ± 22.6 °C) with a relative humidity of 30–70% and 12:12 h light:dark cycle. LabDiet 5002 was available ad libitum throughout the study. For the repeat-dose studies, body weights were recorded twice weekly. Food consumption was measured

Results

There were no dextromethorphan-related deaths or adverse behavioral effects at any dose in the single-dose or first repeat-dose study. In the second repeat-dose study, four deaths occurred in males given 400 mg/(kg day), and one death occurred in a female rat given 120 mg/(kg day). Deaths occurred on days 1, 5, 18, 25 and 29. Behavioral changes were observed 1–2 h after dosing in all groups (Table 1). The most common finding was impaired equilibrium which occurred in all males at all doses, and

Discussion

Single or repeated administration of dextromethorphan to rats (400 mg/(kg/day) in males; 120 mg/(kg/day) in females) for up to 30 days produced mortality and behavioral changes, most notably impaired equilibrium and hypoactivity, but no evidence of neuronal vacuolation or neurodegeneration in the RS/PC cortices or any other brain region examined. These results show that oral administration of high doses of dextromethorphan (which also leads to high dextrorphan exposure) does not produce

DISCLOSURES

These studies were paid for by Endo Pharmaceuticals Inc. under the direction of R.D. Carliss and D. Shuey as employees of Endo Pharmaceuticals Inc.

References (25)

  • N.B. Farber et al.

    Receptor mechanisms and circuitry underlying NMDA antagonist neurotoxicity

    Mol Psychiatry

    (2002)
  • A.S. Fix

    Pathological effects of MK-801 in the rat posterior cingulate/retrosplenial cortex

    Psychopharm Bull

    (1994)
  • Cited by (17)

    • (2R,6R)-hydroxynorketamine acts through GluA1-induced synaptic plasticity to alleviate PTSD-like effects in rat models

      2022, Neurobiology of Stress
      Citation Excerpt :

      Biol Psychiatry 2012; 72(4): 331–338)," 2016; Highland et al., 2019; P. Zanos, 2017). While ketamine's side effects are the result of its antagonism of the NMDA receptor (NMDAR), some studies have proposed that NMDAR inhibition itself may not be what mediates ketamine's antidepressant effects, and, moreover, that (2R,6R)-HNK does not inhibit NMDAR (Carliss et al., 2007; Z. Zanos et al., 2018). ( 2R,6R)-HNK has been demonstrated to reverse chronic stress-induced negative affect by repairing damaged α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-dependent glutamatergic transmission.

    • D-Ribose-LCysteine attenuates manganese-induced cognitive and motor deficit, oxidative damage, and reactive microglia activation

      2022, Environmental Toxicology and Pharmacology
      Citation Excerpt :

      Compromised behaviours may result from damage to neuronal structures in various brain regions (Ijomone et al., 2012, 2018; Kim et al., 2008; Mori et al., 2001; Santiago et al., 2010). Features that characterize the degenerating process in neurons include; prominent eosinophilic cytoplasm with or without shrunken nuclei; pyknotic nuclei; neuronal swelling and vacuolation within the cytoplasm (Carliss et al., 2007; Garman, 2011). The findings from the histomorphological investigations (Figs. 5 and 6) revealed that RibCys attenuated cytoarchitectural distortions in the striatum and motor cortex induced by Mn neurotoxicity, corroborating our earlier findings that Ribcys preserved neuronal ultrastructure by mitigating damage to the mitochondrial cristae, neuronal nuclei and myelin integrity in the course of Mn intoxication (Akingbade et al., 2021).

    View all citing articles on Scopus
    1

    Tel.: +1 419 289 8700; fax: +1 419 289 3650.

    2

    Tel.: +1 610 558 9800; fax: +1 484 840 4291.

    View full text