Skip to main content

N-acetyl-cysteine, a drug that enhances the endogenous activation of group-II metabotropic glutamate receptors, inhibits nociceptive transmission in humans

Abstract

Background

Emerging research seeking novel analgesic drugs focuses on agents targeting group-II metabotropic glutamate receptors (mGlu2 and mGlu3 receptors). N-Acetylcysteine (NAC) enhances the endogenous activation of mGlu2/3 receptors by activating the glial glutamate:cystine membrane exchanger. Here, we examined whether NAC inhibits nociceptive responses in humans and animals. We tested the effect of oral NAC (1.2 g) on thermal-pain thresholds and laser-evoked potentials in 10 healthy volunteers, according to a crossover, double-blind, placebo-controlled design, and the effect of NAC (100 mg/kg, i.p.) on the tail-flick response evoked by radiant heat stimulation in mice.

Results

In healthy subjects, NAC treatment left thermal-pain thresholds unchanged, but significantly reduced pain ratings to laser stimuli and amplitudes of laser-evoked potentials. NAC induced significantly greater changes in these measures than placebo. In the tail-flick test, NAC strongly reduced the nocifensive reflex response to radiant heat. The action of NAC was abolished by the preferential mGlu2/3 receptor antagonist, LY341495 (1 mg/kg, i.p.).

Conclusions

Our findings show for the first time that NAC inhibits nociceptive transmission in humans, and does the same in mice by activating mGlu2/3 receptors. These data lay the groundwork for investigating the therapeutic potential of NAC in patients with chronic pain.

Background

Despite recent advances, only few patients suffering from chronic pain achieve acceptable pain relief with the currently available analgesic drugs [1].

Potential targets for novel analgesic drugs include group-II metabotropic glutamate receptor subtypes (mGlu2 and mGlu3 receptors) [2,3]. mGlu2 and mGlu3 receptors localized in the spinal cord and other regions of the nociceptive system negatively regulate glutamate release by reducing cAMP formation and inhibiting voltage-sensitive calcium channels in presynaptic terminals [2,4]. Immune gold labelling has shown that both receptors are preferentially (albeit not exclusively) localized in the preterminal axonal region, and are therefore inaccessible to synaptic glutamate. A growing body of evidence suggests that presynaptic mGlu2 and mGlu3 receptors are activated by the glutamate released from astrocytes via the glutamate:cystine antiporter (Sxc-) [5,6]. A drug that activates Sxc-, and might therefore be used to reinforce the endogenous activation of mGlu2/3 receptors, is N-acetylcysteine (NAC) [7]. We have shown recently that NAC induces analgesia in animal models of inflammatory and neuropathic pain [8]. NAC-induced analgesia was abolished by genetic deletion of mGlu2 receptors or by co-treatment with the preferential mGlu2/3 receptor antagonist, LY341495 [8]. The study of nociceptive transmission in healthy volunteers is an essential step towards the clinical research of NAC in patients with chronic pain.

In this study we examined whether oral NAC was able to modulate nociceptive transmission in healthy volunteers. Using a double-blind, placebo-controlled design, we tested NAC-induced changes in quantitative sensory testing and laser-evoked potentials, two techniques that international guidelines indicate as “reference standards” for assessing the nociceptive system and testing analgesic efficacy [9-11]. As an experimental counterpart in animals, we also examined changes induced by intraperitoneal-injected NAC on the tail-flick test elicited by radiant heat in mice, the animal model most closely corresponding to laser stimulation in humans.

Results

NAC-induced changes in nociceptive transmission in humans

No subjects reported adverse events after receiving NAC or placebo. The NAC and placebo sessions yielded comparable baseline values for quantitative sensory testing and laser-evoked potential variables (P > 0.1; Table 1). Baseline values were within the normal ranges established in our laboratory.

Table 1 Effect of oral NAC (1.2 g) and placebo on nociceptive transmission in ten healthy volunteers

Oral NAC (1.2 g) significantly attenuated pain evoked by laser stimuli, and the N1-and N2P2 laser-evoked potential amplitudes (laser pain ratings: p < 0.01; N1 component: p < 0.02; N2-P2 complex: p < 0.01) (Figure 1), but left thermal and pain perceptive thresholds unchanged. Placebo left all variables unchanged (Table 1).

Figure 1
figure 1

Oral NAC inhibits nociceptive transmission in healthy volunteers. A: Laser-evoked potential (LEP) recordings during placebo and N-acetyl-cysteine (NAC) sessions in a representative subject. Black: pre-drug recordings. Grey: post-drug recordings. Each trace is the mean of 20 trials. Horizontal calibration: 100 ms; vertical calibration: 20 μV. B: Mean pre-drug (black) and post-drug (grey) values for N1, N2-P2 amplitude LEP components and laser pain ratings during placebo and NAC sessions. Whereas placebo was ineffective, NAC significantly reduced all LEP components and laser pain ratings. Values are means + S.E.M. of 10 determinations. *p < 0.02; **p < 0.01.

NAC-induced changes in tail-flick latencies in mice

A single injection of NAC (100 mg/kg, i.p.; 30 min before the test) substantially increased tail-flick latencies as compared with saline injection in mice (p=0.018) (Figure 2). Pretreatment for 15 min with the preferential mGlu2/3 receptor antagonist, LY341495 (1 mg/kg, i.p.), which was inactive on its own, abolished NAC-induced analgesia (p=0.017) (Figure 2).

Figure 2
figure 2

Acute injection of NAC inhibits radiant heat-induced nocifensive behavior in mice by activating mGlu2/3 receptors. Percentage of the maximum possible effect (%MPE) in the four tail-flick conditions: i) saline followed by saline; (ii) saline followed by N-acetyl-cysteine (NAC) (100 mg/kg); (iii) LY341495 (1 mg/kg) followed by saline; and (iv) LY341495 followed by NAC. NAC injection increased tail-flick latencies. Values are means + S.E.M. of 6–8 determinations. *p < 0.05 vs. all other values, F(3,22)=6.38, p=0.003.

Discussion

Our study, showing that oral NAC reduced laser pain ratings and laser-evoked potential amplitudes, provides the previously unavailable evidence that NAC inhibits nociceptive transmission in healthy humans. These findings in humans received confirmation from the NAC-induced tail-flick suppression in mice.

Precisely where in the nervous system NAC acts to induce analgesia in the nociceptive transmission pathway remains unclear. Although mGlu2/3 receptors lie at various levels across the nociceptive pathway [4], we hypothesize that it does so by inhibiting neurotransmitter release at the synapse between first-order and second-order nociceptive neurons in the dorsal horn [12]. This hypothesis is consistent with our data of laser-evoked potentials showing that NAC concomitantly reduced the N1 component (generated in the SII area), and the N2-P2 complex (generated in the insular and cingulate cortex) [13], which is indicative of a lower action of the drug within the nociceptive pathway. The hypothesis that NAC induces its analgesic effect predominantly in the dorsal horn is also supported by data of the formalin test in mice. NAC specifically reduced nocifensive behavior in the second phase of the formalin test [8], which reflects the development of nociceptive sensitization in the dorsal horn of the spinal cord [14].

Whereas NAC reduced the laser pain ratings and laser-evoked potential amplitudes, it left the thermal-pain thresholds-assessed by the quantitative sensory testing-unchanged. This finding is not surprising because analgesic agents, such as opioids and monoaminergic drugs, have more pronounced effects on high-intensity pain (e.g., laser-evoked pain) than on thermal-pain thresholds [11,15,16].

Unexpectedly in our study we did not find any placebo effect on laser-evoked potentials. This finding is not in line with some studies demonstrating that placebo affects laser-evoked potentials [17]. There are the following potential explanations: (i) in our experiments the subjects received information on the aim of the study; (ii) our subjects knew they had the chance to receive placebo; and (iii) all subjects were well-trained neurophysiologists and neurologists of our Department, and this might have minimized the placebo effect.

As expected in none of our subjects did NAC cause adverse events. NAC, commonly used as a mucolytic agent, is an extremely safe drug. The standard dose (600 mg/day) induces negligible adverse effects [18]. Even when given at high I.V. doses in the treatment of acetaminophen poisoning, its adverse effects are usually mild and easily managed [19]. Hence, NAC might be an ideal and safe adjuvant drug for treating patients with chronic pain undergoing multiple treatments for pain and the related comorbidities.

Using a radiant heat experimental pain model--similar to that used in humans--our experiments showed that tail-flick responses had a longer latency in NAC-treated mice than in saline-treated mice. The tail-flick delay was sensitive to the LY341495-induced mGlu2/3 receptor block. These findings are consistent with the analgesic effect of NAC reported in other experimental pain models (e.g. the formalin test, the complete Freund’s adjuvant chronic inflammatory pain model, and in the chronic constriction injury neuropathic pain model) [8], and show that mGlu2/3 receptors mediate the NAC-induced changes in nociceptive transmission in mice.

Conclusions

Our study, showing that NAC reduces laser-induced pain and laser-evoked brain potentials in humans and delays the tail-flick response in mice indicates that NAC inhibits nociceptive transmission. Accordingly, a previous study directly addressing the efficacy of NAC in patients with complex regional pain syndrome type I, has raised the possibility that NAC might be potentially useful for relieving pain [20]. Hence, because NAC is a safe drug, it deserves to be tested in further adequately sized clinical trials in patients with pain.

Methods

Human experiments

Ten healthy volunteers (4 M, 6 F; 23–38 years) participated in the study. The Institutional Review Board approved all procedures and all subjects gave their written informed consent.

The Institutional Review Board of the Policlinico Umberto I – Department of Neurology and Psychiatry, Sapienza University approved all procedures and all subjects gave their written informed consent.

Quantitative sensory testing of thermal-pain sensitivity

For quantitative sensory testing we used a thermode (ATS, PATHWAY, Medoc, Israel). The computer-driven PATHWAY system contains a metal contact plate (contact area 30 × 30 mm) equipped with an external Peltier element that cools and heats the plate to target levels. The baseline temperature of 32°C reached target temperature at a ramp rate of 1°C/s. Quantitative sensory variables were tested on the right hand dorsum. We tested subjects’ thermal-pain perceptive thresholds: cold detection threshold (CDT), warm detection threshold (WDT), cold pain threshold (CPT) and heat pain threshold (HPT).

Laser-evoked potentials

We used a Nd: YAP laser stimulator under fiber–optic guidance (Electronic Engineering, Florence, Italy). Laser stimuli were set to induce a clear painful pinprick (intensity 119.4-150 mJ/mm2; duration 5 ms; diameter 4 mm) and directed to the right hand dorsum. The laser beam was shifted after each stimulus and the interstimulus interval was varied pseudo-randomly (10–15 s). Subjects, wearing protective goggles, rested comfortably on a medical examination table, keeping their eyes open. The different laser evoked potential components were recorded through disk electrodes from the scalp: T3 referenced to Fz for recording the early lateralized N1 component, and Cz referenced to the nose, for recording the late vertex N2-P2 complex. Electro-oculographic recordings monitored possible eye movements or blinks. For each session, two series of 10–15 artefact-free trials were averaged off line. We measured the peak latencies and amplitudes of the lateralized N1 and the vertex N2-P2 complex. These methods adhered to the recommendations given by the International Federation of Clinical Neurophysiology [21]. In all sessions the subjects were asked to rate the pain evoked by laser stimuli on a 0–10 numeric rating scale (NRS) (0=no sensation, 10=worst possible pain).

Experimental procedure

We conducted a double-blind, placebo-controlled, crossover study. All subjects underwent two separate sessions, one with oral N-acetylcysteine (NAC) (effervescent tablets, Ratiopharm, 1.2 g) and the other with oral placebo (Multicentrum, multivitamin supplement, in effervescent tablets). The two sessions were randomly alternated among subjects. Two investigators, both unaware of the type of session (drug or placebo), recorded the thermal-pain perceptive thresholds and laser evoked potential measures. Each session comprised two recording blocks: before oral NAC or placebo (pre-drug), and 60 min after NAC or placebo (post-drug). As primary outcome variables, we selected the warm and cold detection thresholds, the cold and heat pain thresholds; N1 component and N2-P2 laser-evoked potential amplitudes and the numerical rating scale for the perceived pain intensity during the laser evoked potential recordings.

Animal experiments

All experiments were conducted according to the European (86/609/EEC) and Italian (D: Lgs. 116/92) guidelines for animal care. The experimental protocol was approved by the Italian Ministry of Health. All efforts were made to minimize animal suffering and the number of animals used.

Drugs

NAC was purchased from Sigma Aldrich (Milano, Italy). (2S)-2-Amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl) propanoic acid (LY341495) was purchased from Tocris Cookson (Avonmouth, Bristol, UK).

Tail-flick test

We used adult male C57BL/6 J mice (body weight 25–28 g) purchased from Charles River (Calco, Italy). All mice were housed 5 per cage, under a standard 12/12 h light/dark cycle with food and water ad libitum. Testing took place from 9:00 a.m. to 11 a.m. on two consecutive days. Four groups of 6–8 mice received two intraperitoneal (i.p.) injections separated by a 15-min interval, as follows: (i) saline followed by saline; (ii) saline followed by NAC (100 mg/kg); (iii) LY341495 (1 mg/kg) followed by saline; and (iv) LY341495 followed by NAC. In the tail-flick test, mice were loosely wrapped in a velvet towel and placed on the tail-flick apparatus (Tail Flick model DS 20 Socrel Apelex, France). A light beam was focused on the tail approximately 1 to 3 cm from the base, and to minimize tissue damage the latency to a vigorous radiant-heat-induced tail flick was measured with a 10-second cut-off time. Tail-flick latencies at baseline (BL) were measured 3 times per mouse at 2-min intervals, and 5 min before the first i.p. injection. The study included only mice whose mean baseline reaction times ranged between 3 and 5 sec. Latencies were measured again 30 min after the second i.p. injection (test latency, TL). Results were expressed as a percentage of the maximum possible effect (%MPE) using the following formula: %MPE=100_{(TL–mean BL)/(Cut-off time–mean BL)}.

Statistical analysis

Because several variables for human subjects had a non-normal distribution, all comparisons in humans were analyzed for significance with the Wilcoxon signed-rank test. Data for tail-flick in mice were tested with a one-way analysis of variance (ANOVA) followed by Dunnett’s t test to isolate the differences. P values <0.05 were considered as statistically significant.

Abbreviations

mGlu:

Metabotropic glutamate receptor

Sxc-:

Cystine/glutamate antiporter

NAC:

N-Acetylcysteine

CDT:

Cold detection threshold

WDT:

Warm detection threshold

CPT:

Cold pain threshold

HPT:

Heat pain threshold

References

  1. Hansson PT, Attal N, Baron R, Cruccu G. Toward a definition of pharmacoresistant neuropathic pain. Eur J Pain. 2009;13:439–40.

    Article  PubMed  Google Scholar 

  2. Chiechio S, Copani A, De Petris L, Morales ME, Nicoletti F, Gereau 4th RW. Transcriptional regulation of metabotropic glutamate receptor 2/3 expression by the NF-kappaB pathway in primary dorsal root ganglia neurons: a possible mechanism for the analgesic effect of L-acetylcarnitine. Mol Pain. 2006;2:20.

    Article  PubMed Central  PubMed  Google Scholar 

  3. Montana MC, Gereau RW. Metabotropic glutamate receptors as targets for analgesia: antagonism, activation, and allosteric modulation. Curr Pharm Biotechnol. 2011;12:1681–8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Nicoletti F, Bockaert J, Collingridge GL, Conn PJ, Ferraguti F, Schoepp DD, et al. Metabotropic glutamate receptors: from the workbench to the bedside. Neuropharmacology. 2011;60:1017–41.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Baker DA, Xi ZX, Shen H, Swanson CJ, Kalivas PW. The origin and neuronal function of in vivo nonsynaptic glutamate. J Neurosci. 2002;22:9134–41.

    CAS  PubMed  Google Scholar 

  6. Moran MM, McFarland K, Melendez RI, Kalivas PW, Seamans JK. Cystine/glutamate exchange regulates metabotropic glutamate receptor presynaptic inhibition of excitatory transmission and vulnerability to cocaine seeking. J Neurosci. 2005;25:6389–93.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. Bridges R, Lutgen V, Lobner D, Baker DA. Thinking outside the cleft to understand synaptic activity: contribution of the cystine-glutamate antiporter (System xc-) to normal and pathological glutamatergic signaling. Pharmacol Rev. 2012;64:780–802.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. Bernabucci M, Notartomaso S, Zappulla C, Fazio F, Cannella M, Motolese M, et al. N-Acetyl-cysteine causes analgesia by reinforcing the endogenous activation of type-2 metabotropic glutamate receptors. Mol Pain. 2012;8:77.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Haanpää M, Attal N, Backonja M, Baron R, Bennett M, Bouhassira D, et al. NeuPSIG guidelines on neuropathic pain assessment. Pain. 2011;152:14–27.

    Article  PubMed  Google Scholar 

  10. Truini A, Vergari M, Biasiotta A, La Cesa S, Gabriele M, Di Stefano G, et al. Transcutaneous spinal direct current stimulation inhibits nociceptive spinal pathway conduction and increases pain tolerance in humans. Eur J Pain. 2011;15:1023–7.

    Article  CAS  PubMed  Google Scholar 

  11. Truini A, Panuccio G, Galeotti F, Maluccio MR, Sartucci F, Avoli M, et al. Laser-evoked potentials as a tool for assessing the efficacy of antinociceptive drugs. Eur J Pain. 2010;14:222–5.

    Article  CAS  PubMed  Google Scholar 

  12. Carlton SM, Hargett GL, Coggeshall RE. Localization of metabotropic glutamate receptors 2/3 on primary afferent axons in the rat. Neuroscience. 2001;105:957–69.

    Article  CAS  PubMed  Google Scholar 

  13. Garcia-Larrea L, Frot M, Valeriani M. Brain generators of laser-evoked potentials: from dipoles to functional significance. Neurophysiol Clin. 2003;33:279–92.

    Article  CAS  PubMed  Google Scholar 

  14. Zammataro M, Chiechio S, Montana MC, Traficante A, Copani A, Nicoletti F, et al. mGlu2 metabotropic glutamate receptors restrain inflammatory pain and mediate the analgesic activity of dual mGlu2/mGlu3 receptor agonists. Mol Pain. 2011;7:6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Poulsen L, Arendt-Nielsen L, Brøsen K, Nielsen KK, Gram LF, Sindrup SH. The hypoalgesic effect of imipramine in different human experimental pain models. Pain. 1995;60:287–93.

    Article  CAS  PubMed  Google Scholar 

  16. Skarke C, Darimont J, Schmidt H, Geisslinger G, Lötsch J. Analgesic effects of morphine and morphine-6-glucuronide in a transcutaneous electrical pain model in healthy volunteers. Clin Pharmacol Ther. 2003;73:107–21.

    Article  CAS  PubMed  Google Scholar 

  17. Colloca L, Tinazzi M, Recchia S, Le Pera D, Fiaschi A, Benedetti F, et al. Learning potentiates neurophysiological and behavioral placebo analgesic responses. Pain. 2008;139:306–14.

    Article  PubMed  Google Scholar 

  18. Treweeke AT, Winterburn TJ, Mackenzie I, Barrett F, Barr C, Rushworth GF, et al. Megson IL N-Acetylcysteine inhibits platelet-monocyte conjugation in patients with type 2 diabetes with depleted intraplatelet glutathione: a randomised controlled trial. Diabetologia. 2012;55:2920–8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Schmidt LE, Dalhoff K. Risk factors in the development of adverse reactions to N-acetylcysteine in patients with paracetamol poisoning. Br J Clin Pharmacol. 2001;51:87–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  20. Perez RS, Zuurmond WW, Bezemer PD, Kuik DJ, van Loenen AC, de Lange JJ, et al. The treatment of complex regional pain syndrome type I with free radical scavengers: a randomized controlled study. Pain. 2003;102:297–307.

    Article  CAS  PubMed  Google Scholar 

  21. Cruccu G, Aminoff MJ, Curio G, Guerit JM, Kakigi R, Mauguiere F, et al. Recommendations for the clinical use of somatosensory-evoked potentials. Clin Neurophysiol. 2008;119:1705–19.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgment

This work was supported by the Italian Ministry of Health (project code: RF-2011-02352582).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ferdinando Nicoletti.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

AT, SP, EP, GDS: performed human experiments and analyzed data; SN, RL, GB: performed animal experiments and analyzed data; AT, GC, FN: designed experiments, supervised research and wrote the manuscript. All authors read and approved the final manuscript.

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Truini, A., Piroso, S., Pasquale, E. et al. N-acetyl-cysteine, a drug that enhances the endogenous activation of group-II metabotropic glutamate receptors, inhibits nociceptive transmission in humans. Mol Pain 11, 14 (2015). https://doi.org/10.1186/s12990-015-0009-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12990-015-0009-2

Keywords