N-Acetylcysteine causes analgesia in a mouse model of painful diabetic neuropathy

Serena Notartomaso, Pamela Scarselli, Giada Mascio, Francesca Liberatore, Emanuela Mazzon, Santa Mammana, Agnese Gugliandolo, Giorgio Cruccu, Valeria Bruno, Ferdinando Nicoletti, Giuseppe Battaglia, Serena Notartomaso, Pamela Scarselli, Giada Mascio, Francesca Liberatore, Emanuela Mazzon, Santa Mammana, Agnese Gugliandolo, Giorgio Cruccu, Valeria Bruno, Ferdinando Nicoletti, Giuseppe Battaglia

Abstract

N-Acetylcysteine, one of the most prescribed antioxidant drugs, enhances pain threshold in rodents and humans by activating mGlu2 metabotropic glutamate receptors. Here, we assessed the analgesic activity of N-acetylcysteine in the streptozotocin model of painful diabetic neuropathy and examined the effect of N-acetylcysteine on proteins that are involved in mechanisms of nociceptive sensitization. Mice with blood glucose levels ≥250 mg/dl in response to a single intraperitoneal (i.p.) injection of streptozotocin (200 mg/kg) were used for the assessment of mechanical pain thresholds. Systemic treatment with N-acetylcysteine (100 mg/kg, i.p., either single injection or daily injections for seven days) caused analgesia in diabetic mice. N-acetylcysteine-induced analgesia was abrogated by the Sxc− inhibitors, sulfasalazine (8 mg/kg, i.p.), erastin (30 mg/kg, i.p.), and sorafenib (10 mg/kg, i.p.), or by the mGlu2/3 receptor antagonist, LY341495 (1 mg/kg, i.p.). Repeated administrations of N-acetylcysteine in diabetic mice reduced ERK1/2 phosphorylation in the dorsal region of the lumbar spinal cord. The analgesic activity of N-acetylcysteine was occluded by the MEK inhibitor, PD0325901 (25 mg/kg, i.p.), the TRPV1 channel blocker, capsazepine (40 mg/kg, i.p.), or by a cocktail of NMDA and mGlu5 metabotropic glutamate receptor antagonists (memantine, 25 mg/kg, plus MTEP, 5 mg/kg, both i.p.). These findings offer the first demonstration that N-acetylcysteine relieves pain associated with diabetic neuropathy and holds promise for the use of N-acetylcysteine as an add-on drug in diabetic patients.

Keywords: N-acetylcysteine; Neuropathic pain; diabetes; nociceptive sensitization; streptozotocin.

Figures

Figure 1.
Figure 1.
NAC-induced analgesia in the STZ model of painful diabetic neuropathy. Blood glucose levels in mice receiving a single injection of saline or STZ (200 mg/kg, i.p.) are shown in (a), where values are means ± S.E.M. of 7–10 mice. *p (1,13) = 77.224). The effect of a single injection of NAC or pregabalin on mechanical pain thresholds in diabetic mice and non-diabetic control mice are shown in (c), where values are means ± S.E.M. of 7–10 mice. *p < 0.05 (one-way ANOVA + Duncan method applied only to the groups of diabetic mice; F(2,18) = 4.562). The effect of repeated administrations of saline or NAC (100 mg/kg, i.p., once a day for 7 days starting from 21 days after STZ injection) on mechanical pain thresholds are shown in (d and e). In (d), all groups of mice were injected once with saline, LY341495, or sulfasalazine at the end of the chronic treatment with saline or NAC (see Methods). Values are means ± S.E.M. of 6–7 mice per group. p < 0.05 versus the control group receiving repeated injections of saline followed by a single injection of saline (*); or versus the group treated with NAC and a single injection of saline (#) (one-way ANOVA + Duncan method; F(5,35) = 3.364). In (e), all groups of mice were injected once with vehicle, erastin, sorafenib, PD0325901, capsazepine, and mementine+MTEP at the end of the chronic treatment with saline or NAC. Values are means ± S.E.M. of 4–10 mice per group. p < 0.05 versus the control group receiving repeated injections of saline followed by a single injection of vehicle (*); or versus the group treated with NAC and a single injection of vehicle (#); or versus the control group receiving repeated injections of saline followed by a single injection of vehicle ($) (one-way ANOVA + Duncan method; F(11,73) = 7.947). Blood glucose levels in mice receiving a single injection of saline or NAC (100 mg/kg, i.p.) are shown in (f), where values are means ± S.E.M. of 9 mice. In (g), four groups of mice were injected once with vehicle or JNJ47965567 (30 mg/kg) at the end of the chronic treatment with saline or NAC. Values are means ± S.E.M. of 7–10 mice per group. p < 0.05 versus all other groups (one-way ANOVA + Duncan method; F(3,28) = 18.643). NAC: N-acetylcysteine; STZ: streptozotocin.
Figure 2.
Figure 2.
Expression of proteins targeted by NAC or involved in mechanisms of nociceptive sensitization in the dorsal region of the lumbar spinal cord of diabetic and non-diabetic mice receiving repeated administrations of NAC. Mice were treated i.p. with either saline or NAC for seven days (100 mg/kg) once a day, starting from 21 days following a single injection of saline or STZ. Densitometric values are expressed as % values obtained in the group of STZ mice treated with saline for seven days (this group is common to two–three different immunoblots used for the analysis). Only one representative blot is shown. Densitometric values are means ± S.E.M. of 2–7 mice. p (1,13) = 11.996 for pathological model (control and diabetic mice) and F(1,13) = 4.717 for drug treatment; P-ERK1: F(1,17) = 13.312 for pathological model (control and diabetic mice) and F(1,17) = 17.514 for drug treatment; and P-ERK2: F(1,16) = 6.652 for pathological model (control and diabetic mice) and F(1,16) = 19.678 for drug treatment)). NAC: N-acetylcysteine.
Figure 3.
Figure 3.
NAC-induced analgesia in the CFA model of chronic inflammatory pain. Mechanical pain thresholds in mice seven days following CFA injection in the plantar paw after acute systemic treatment with saline or NAC (25-100 mg/kg, i.p.) are shown in (a), where values are means ± S.E.M. of 6–7 mice per group. p (4,28) = 3.834) versus saline treated-mice (*) or versus CFA-injected mice after saline treatment (#). The effect of repeated administrations of saline or NAC (100 mg/kg, i.p., once a day for seven days starting from seven days after CFA injection) on mechanical pain thresholds is shown in (b). Values are means ± S.E.M. of seven mice per group. *p < 0.05 (Student’s t-test; t = −1.841). NAC: N-acetylcysteine; CFA: Complete Freund’s Adjuvant.
Figure 4.
Figure 4.
Expression of proteins targeted by NAC or involved in mechanisms of nociceptive sensitization in the dorsal region of the ipsilateral lumbar spinal cord or in the plantar paw of CFA-injected mice receiving repeated administrations of NAC. Mice were treated i.p. with either saline or NAC for seven days (100 mg/kg) once a day, starting from seven days following CFA injection in the plantar paw. Densitometric values are means ± S.E.M. of 3–5 mice. *p 

References

    1. Sloan G, Shillo P, Selvarajah D, Wu J, Wilkinson ID, Tracey I, Anand P, Tesfaye S. A new look at painful diabetic neuropathy. Diabetes Res Clin Pract 2018; 144: 177–191.
    1. Attal N, Cruccu G, Baron R, Haanpää M, Hansson P, Jensen TS, Nurmikko T; European Federation of Neurological Societies. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol 2010; 17: 1113–e88.
    1. Jensen TS, Backonja MM, Hernández Jiménez S, Tesfaye S, Valensi P, Ziegler D. New perspectives on the management of diabetic peripheral neuropathic pain. Diab Vasc Dis Res 2006; 3: 108–119.
    1. Tesfaye S, Boulton AJ, Dickenson AH. Mechanisms and management of diabetic painful distal symmetrical polyneuropathy. Diabetes Care 2013; 36: 2456–2465.
    1. Chiechio S, Copani A, Zammataro M, Battaglia G, Gereau RW, IV, Nicoletti F. Transcriptional regulation of type-2 metabotropic glutamate receptors: an epigenetic path to novel treatments for chronic pain. Trends Pharmacol Sci 2010; 31: 153–160.
    1. Zammataro M, Chiechio S, Montana M C, Traficante A, Copani A, Nicoletti F, Gereau RW. mGlu2 metabotropic glutamate receptors restrain inflammatory pain and mediate the analgesic activity of dual mGlu2/mGlu3 receptor agonists. Mol Pain 2011; 7: 1744–8069.
    1. Davidson S, Golden JP, Copits BA, Ray PR, Vogt SK, Valtcheva MV, Schmidt RE, Ghetti A, Price TJ, Gereau RW IV. Group II mGluRs suppress hyperexcitability in mouse and human nociceptors. Pain 2016; 157: 2081–2088.
    1. De Grandis D, Minardi C. Acetyl-L-carnitine (levacecarnine) in the treatment of diabetic neuropathy. A long-term, randomised, double-blind, placebo-controlled study. Drugs R&D 2002; 3: 223–231.
    1. Sima AAF, Calvani M, Mehra M, Amato A; Acetyl-L-Carnitine Study Group. Acetyl-L-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: an analysis of two randomized placebo-controlled trials. Diabetes Care 2005; 28: 89–94.
    1. Sima AA. Acetyl-L-carnitine in diabetic polyneuropathy: experimental and clinical data. CNS Drugs 2007; 21 Suppl 1: 13–23.
    1. Evans JD, Jacobs TF, Evans EW. Role of acetyl-L-carnitine in the treatment of diabetic peripheral neuropathy. Ann Pharmacother 2008; 42: 1686–1691.
    1. Chiechio S, Caricasole A, Barletta E, Storto M, Catania MV, Copani A, Vertechy M, Nicolai R, Calvani M, Melchiorri D, Nicoletti F. L-Acetylcarnitine induces analgesia by selectively up-regulating mGlu2 metabotropic glutamate receptors. Mol Pharmacol 2002; 61: 989–996.
    1. Chiechio S, Copani A, De Petris L, Morales ME, Nicoletti F, Gereau 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: 1744–8069.
    1. Notartomaso S, Mascio G, Bernabucci M, Zappulla C, Scarselli P, Cannella M, Imbriglio T, Gradini R, Battaglia G, Bruno V, Nicoletti F. Analgesia induced by the epigenetic drug, L-acetylcarnitine, outlasts the end of treatment in mouse models of chronic inflammatory and neuropathic pain. Mol Pain 2017; 13: 1744806917697009.
    1. Rogers JJ, Heard K. Does age matter? Comparing case fatality rates for selected poisonings reported to U.S. poison centers. Clin Toxicol (Phila) 2007; 45: 705–708.
    1. Heard KJ. Acetylcysteine for acetaminophen poisoning. N Engl J Med 2008; 359: 285–292.
    1. Millea PJ. N-acetylcysteine: multiple clinical applications. Am Fam Physician 2009; 80: 265–269.
    1. Anderson SM, Park ZH, Patel RV. Intravenous N-acetylcysteine in the prevention of contrast media-induced nephropathy. Ann Pharmacother 2011; 45: 101–107.
    1. Dodd S, Dean O, Copolov DL, Malhi GS, Berk M. N-acetylcysteine for antioxidant therapy: pharmacology and clinical utility. Expert Opin Biol Ther 2008; 8: 1955–1962.
    1. Rushworth GF, Megson IL. Existing and potential therapeutic uses for N-acetylcysteine: the need for conversion to intracellular glutathione for antioxidant benefits. Pharmacol Ther 2014; 141: 150–159.
    1. Elbini Dhouib I, Jallouli M, Annabi A, Gharbi N, Elfazaa S, Lasram MM. A minireview on N-acetylcysteine: an old drug with new approaches. Life Sci 2016; 151: 359–363.
    1. Kalivas PW. The glutamate homeostasis hypothesis of addiction. Nat Rev Neurosci 2009; 10: 561–572.
    1. Nicoletti F, Bockaert J, Collingridge GL, Conn PJ, Ferraguti F, Schoepp DD, Wroblewski JT, Pin JP. Metabotropic glutamate receptors: from the workbench to the bedside. Neuropharmacology 2011; 60: 1017–1041.
    1. Olive MF, Cleva RM, Kalivas PW, Malcolm RJ. Glutamatergic medications for the treatment of drug and behavioral addictions. Pharmacol Biochem Behav 2012; 100: 801–810.
    1. Schmaal L, Veltman DJ, Nederveen A, van den Brink W, Goudriaan AE. N-acetylcysteine normalizes glutamate levels in cocaine-dependent patients: a randomized crossover magnetic resonance spectroscopy study. Neuropsychopharmacology 2012; 37: 2143–2152.
    1. Post RM, Kalivas P. Bipolar disorder and substance misuse: pathological and therapeutic implications of their comorbidity and cross-sensitisation. Br J Psychiatry 2013; 20: 172–176.
    1. McClure EA, Gipson CD, Malcolm RJ, Kalivas PW, Gray KM. Potential role of N-acetylcysteine in the management of substance use disorders. CNS Drugs 2014; 28: 95–106.
    1. Deepmala Slattery J, Kumar N, Delhey L, Berk M, Dean O, Spielholz C, Frye R. Clinical trials of N-acetylcysteine in psychiatry and neurology: a systematic review. Neurosci Biobehav Rev 2015; 55: 294–321.
    1. Fernandes BS, Dean OM, Dodd S, Malhi GS, Berk M. N-Acetylcysteine in depressive symptoms and functionality: a systematic review and meta-analysis. J Clin Psychiatry 2016; 77: e457–e466.
    1. Nocito Echevarria MA, Andrade Reis T, Ruffo Capatti G, Siciliano Soares V, da Silveira DX, Fidalgo TM. N-acetylcysteine for treating cocaine addiction – a systematic review. Psychiatry Res 2017; 251: 197–203.
    1. Tomko RL, Gilmore AK, Gray KM. The role of depressive symptoms in treatment of adolescent cannabis use disorder with N-Acetylcysteine. Addict Behav 2018; 85: 26–30.
    1. Bernabucci M, Notartomaso S, Zappulla C, Fazio F, Cannella M, Motolese M, Battaglia G, Bruno V, Gradini R, Nicoletti F. N-Acetyl-cysteine causes analgesia by reinforcing the endogenous activation of type-2 metabotropic glutamate receptors. Mol Pain 2012; 8: 1744–8069.
    1. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 1994; 53: 55–63.
    1. Smilkstein MJ, Knapp GL, Kulig KW, Rumack BH. Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. Analysis of the national multicenter study (1976 to 1985). N Engl J Med 1988; 319: 1557–1562.
    1. Borgström L, Kågedal B, Paulsen O. Pharmacokinetics of N-acetylcysteine in man. Eur J Clin Pharmacol 1986; 31: 217–222.
    1. Olsson B, Johansson M, Gabrielsson J, Bolme P. Pharmacokinetics and bioavailability of reduced and oxidized N-acetylcysteine. Eur J Clin Pharmacol 1988; 34: 77–82.
    1. Knackstedt LA, LaRowe S, Mardikian P, Malcolm R, Upadhyaya H, Hedden S, Markou A, Kalivas PW. The role of cystine-glutamate exchange in nicotine dependence in rats and humans. Biol Psychiatry 2009; 65: 841–845.
    1. Moussawi K, Pacchioni A, Moran M, Olive MF, Gass JT, Lavin A, Kalivas PW. N-Acetylcysteine reverses cocaine-induced metaplasticity. Nat Neurosci 2009; 12: 182–189.
    1. Reissner KJ, Gipson CD, Tran PK, Knackstedt LA, Scofield MD, Kalivas PW. Glutamate transporter GLT-1 mediates N-acetylcysteine inhibition of cocaine reinstatement. Addict Biol 2015; 20: 316–323.
    1. Islam MS. Animal models of diabetic neuropathy: progress since 1960s. J Diabetes Res 2013; 2013: 149452.
    1. Gao F, Zheng ZM. Animal models of diabetic neuropathic pain. Exp Clin Endocrinol Diabetes 2014; 122: 100–106.
    1. Jolivalt CG, Frizzi KE, Guernsey L, Marquez A, Ochoa J, Rodriguez M, Calcutt NA. Peripheral neuropathy in mouse models of diabetes. Curr Protoc Mouse Biol 2016; 6: 223–255.
    1. Gout PW, Buckley AR, Simms CR, Bruchovsky N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the x(c)- cystine transporter: a new action for an old drug. Leukemia 2001; 15: 1633–1640.
    1. Dahlmanns M, Yakubov E, Chen D, Sehm T, Rauh M, Savaskan N, Wrosch JK. Chemotherapeutic xCT inhibitors sorafenib and erastin unraveled with the synaptic optogenetic function analysis tool. Cell Death Discov 2017; 3: 1–11.
    1. Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet 2007; 369: 1641–1657.
    1. Fallahi P, Ferrari SM, Santini F, Corrado A, Materazzi G, Ulisse S, Miccoli P, Antonelli A. Sorafenib and thyroid cancer. BioDrugs 2013; 27: 615–628.
    1. Ferrari SM, Politti U, Spisni R, Materazzi G, Baldini E, Ulisse S, Miccoli P, Antonelli A, Fallahi P. Sorafenib in the treatment of thyroid cancer. Expert Rev Anticancer Ther 2015; 15: 863–874.
    1. Connell LC, Harding JJ, Abou-Alfa GK. Advanced hepatocellular cancer: the current state of future research. Curr Treat Options Oncol 2016; 17: 43.
    1. Corrado A, Ferrari SM, Politti U, Mazzi V, Miccoli M, Materazzi G, Antonelli A, Ulisse S, Fallahi P, Miccoli P. Aggressive thyroid cancer: targeted therapy with sorafenib. Minerva Endocrinol 2017; 42: 64–76.
    1. Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, Wolpaw AJ, Smukste I, Peltier JM, Boniface JJ, Smith R, Lessnick SL, Sahasrabudhe S, Stockwell BR. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 2007; 447: 864–868.
    1. Schoepp DD, Jane DE, Monn JA. Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology 1999; 38: 1431–1476.
    1. Zhuo M. Ionotropic glutamate receptors contribute to pain transmission and chronic pain. Neuropharmacology 2017; 112: 228–234.
    1. Chen SR, Zhang J, Chen H, Pan HL. Streptozotocin-induced diabetic neuropathic pain is associated with potentiated calcium-permeable AMPA receptor activity in the spinal cord. J Pharmacol Exp Ther 2019; 371: 242–249.
    1. Ismail CAN, Suppian R, Abd Aziz CB, Haris K, Long I. Increased nociceptive responses in streptozotocin-induced diabetic rats and the related expression of spinal NR2B subunit of N-methyl-D-aspartate receptors. Diabetes Metab J 2019; 43: 222–235.
    1. Müller WE, Mutschler E, Riederer P. Noncompetitive NMDA receptor antagonists with fast open-channel blocking kinetics and strong voltage-dependency as potential therapeutic agents for Alzheimer’s dementia. Pharmacopsychiatry 1995; 28: 113–124.
    1. Anderson JJ, Rao SP, Rowe B, Giracello DR, Holtz G, Chapman DF, Tehrani L, Bradbury MJ, Cosford ND, Varney MA. [3H]Methoxymethyl-3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine binding to metabotropic glutamate receptor subtype 5 in rodent brain: in vitro and in vivo characterization. J Pharmacol Exp Ther 2002; 303: 1044–1051.
    1. Chen SR, Samoriski G, Pan HL. Antinociceptive effects of chronic administration of uncompetitive NMDA receptor antagonists in a rat model of diabetic neuropathic pain. Neuropharmacology 2009; 57: 121–126.
    1. Font J, López-Cano M, Notartomaso S, Scarselli P, Di Pietro P, Bresolí-Obach R, Battaglia G, Malhaire F, Rovira X, Catena J, Giraldo J, Pin JP, Fernández-Dueñas V, Goudet C, Nonell S, Nicoletti F, Llebaria A, Ciruela F. Optical control of pain in vivo with a photoactive mGlu(5) receptor negative allosteric modulator. Elife 2017; 6: e23545.
    1. Pereira V, Goudet C. Emerging trends in pain modulation by metabotropic glutamate receptors. Front Mol Neurosci 2018; 11: 464.
    1. Ma W, Quirion R. Partial sciatic nerve ligation induces increase in the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in astrocytes in the lumbar spinal dorsal horn and the gracile nucleus. Pain 2002; 99: 175–184.
    1. Ma W, Quirion R. The ERK/MAPK pathway, as a target for the treatment of neuropathic pain. Expert Opin Ther Targets 2005; 9: 699–713.
    1. Ciruela A, Dixon AK, Bramwell S, Gonzalez MI, Pinnock RD, Lee K. Identification of MEK1 as a novel target for the treatment of neuropathic pain. Br J Pharmacol 2003; 138: 751–756.
    1. Song JJ, Lee YJ. Cross-talk between JIP3 and JIP1 during glucose deprivation: SEK1-JNK2 and Akt1 act as mediators. J Biol Chem 2005; 280: 26845–26855.
    1. Zhuang ZY, Gerner P, Woolf CJ, Ji RR. ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain 2005; 114: 149–159.
    1. Crown ED, Ye Z, Johnson KM, Xu GY, McAdoo DJ, Hulsebosch CE. Increases in the activated forms of ERK 1/2, p38 MAPK, and CREB are correlated with the expression of at-level mechanical allodynia following spinal cord injury. Exp Neurol 2006; 199: 397–407.
    1. Ji R-R, Gereau RW, Malcangio M, Strichartz GR. MAP kinase and pain. Brain Res Rev 2009; 60: 135–148.
    1. Matsuoka Y, Yang J. Selective inhibition of extracellular signal-regulated kinases 1/2 blocks nerve growth factor to brain-derived neurotrophic factor signaling and suppresses the development of and reverses already established pain behavior in rats. Neuroscience 2012; 206: 224–236.
    1. Uttam S, Wong C, Amorim IS, Jafarnejad SM, Tansley SN, Yang J, Prager Khoutorsky M, Mogil JS, Gkogkas CG, Khoutorsky A. Translational profiling of dorsal root ganglia and spinal cord in a mouse model of neuropathic pain. Neurobiol Pain 2018; 4: 35–44.
    1. Papale A, Morella IM, Indrigo MT, Bernardi RE, Marrone L, Marchisella F, Brancale A, Spanagel R, Brambilla R, Fasano S. Impairment of cocaine-mediated behaviours in mice by clinically relevant Ras-ERK inhibitors. Elife 2016; 5: e17111.
    1. Choi JW, In JH, Kim YS, Kang YJ, Lim YG, Cho SM, Shin EY, Joo JD. Low dose ketamine reduces the induction of ERK1/2 and CREB signaling protein in a neuropathic pain model of rats. Korean J Anesthesiol 2009; 57: 210–216.
    1. Landry RP, Martinez E, DeLeo JA, Romero-Sandoval EA. Spinal cannabinoid receptor type 2 agonist reduces mechanical allodynia and induces mitogen-activated protein kinase phosphatases in a rat model of neuropathic pain. J Pain 2012; 13: 836–848.
    1. Lin JP, Chen CQ, Huang LE, Li NN, Yang Y, Zhu SM, Yao YX. Dexmedetomidine attenuates neuropathic pain by inhibiting P2X7R expression and ERK phosphorylation in rats. Exp Neurobiol 2018; 27: 267–276.
    1. Hu H-J, Carrasquillo Y, Karim F, Jung WE, Nerbonne JM, Schwarz TL, Gereau RW IV. The kv4.2 potassium channel subunit is required for pain plasticity. Neuron 2006; 50: 89–100.
    1. Hu H-J, Alter BJ, Carrasquillo Y, Qiu C-S, Gereau RW IV. Metabotropic glutamate receptor 5 modulates nociceptive plasticity via extracellular signal-regulated kinase-Kv4.2 signaling in spinal cord dorsal horn neurons. J Neurosci 2007; 27: 13181–13191.
    1. Hu H-J, Gereau RW. Metabotropic glutamate receptor 5 regulates excitability and Kv4.2-containing K+ channels primarily in excitatory neurons of the spinal dorsal horn. J Neurophysiol 2011; 105: 3010–3021.
    1. Kuan YH, Shyu BC. Nociceptive transmission and modulation via P2X receptors in central pain syndrome. Mol Brain 2016; 9: 58.
    1. Donnelly-Roberts DL, Jarvis MF. Discovery of P2X7 receptor-selective antagonists offers new insights into P2X7 receptor function and indicates a role in chronic pain states. Br J Pharmacol 2007; 151: 571–579.
    1. Trang T, Beggs S, Salter MW. ATP receptors gate microglia signaling in neuropathic pain. Exp Neurol 2012; 234: 354–361.
    1. Burnstock G. Purinergic mechanisms and pain. Adv Pharmacol 2016; 75: 91–137.
    1. Ursu D, Ebert P, Langron E, Ruble C, Munsie L, Zou W, Fijal B, Qian YW, McNearney TA, Mogg A, Grubisha O, Merchant K, Sher E. Gain and loss of function of P2X7 receptors: mechanisms, pharmacology and relevance to diabetic neuropathic pain. Mol Pain 2014; 10: 1744–8069.
    1. Bhattacharya A, Wang Q, Ao H, Shoblock JR, Lord B, Aluisio L, Fraser I, Nepomuceno D, Neff RA, Welty N, Lovenberg TW, Bonaventure P, Wickenden AD, Letavic MA. Pharmacological characterization of a novel centrally permeable P2X7 receptor antagonist: JNJ-47965567. Br J Pharmacol 2013; 170: 624–640.
    1. Bevan S, Hothi S, Hughes G, James IF, Rang HP, Shah K, Walpole CS, Yeats JC. Capsazepine: a competitive antagonist of the sensory neurone excitant capsaicin. Br J Pharmacol 1992; 107: 544–552.
    1. Rashid MH, Inoue M, Kondo S, Kawashima T, Bakoshi S, Ueda H. Novel expression of vanilloid receptor 1 on capsaicin-insensitive fibers accounts for the analgesic effect of capsaicin cream in neuropathic pain. J Pharmacol Exp Ther 2003; 304: 940–948.
    1. Walker KM, Urban L, Medhurst SJ, Patel S, Panesar M, Fox AJ, McIntyre P. The VR1 antagonist capsazepine reverses mechanical hyperalgesia in models of inflammatory and neuropathic pain. J Pharmacol Exp Ther 2003; 304: 56–62.
    1. Sałat K, Filipek B. Antinociceptive activity of transient receptor potential channel TRPV1, TRPA1, and TRPM8 antagonists in neurogenic and neuropathic pain models in mice. J Zhejiang Univ Sci B 2015; 16: 167–178.
    1. Menéndez L, Juárez L, García E, García-Suárez O, Hidalgo A, Baamonde A. Analgesic effects of capsazepine and resiniferatoxin on bone cancer pain in mice. Neurosci Lett 2006; 393: 70–73.
    1. Mrozkova P, Spicarova D, Palecek J. Hypersensitivity induced by activation of spinal cord PAR2 receptors is partially mediated by TRPV1 receptors. PLoS One 2016; 11: e0163991.
    1. Kalynovska N, Adamek P, Palecek J. TRPV1 receptors contribute topaclitaxel-induced c-Fos expression in spinal cord dorsal horn neurons. Physiol Res 2017; 66: 549–552.
    1. Chakraborty S, Elvezio V, Kaczocha M, Rebecchi M, Puopolo M. Presynaptic inhibition of transient receptor potential vanilloid type 1 (TRPV1) receptors by noradrenaline in nociceptive neurons. J Physiol (Lond) 2017; 595: 2639–2660.
    1. Immke DC, Gavva NR. The TRPV1 receptor and nociception. Semin Cell Dev Biol 2006; 17: 582–591.
    1. Cortright DN, Krause JE, Broom DC. TRP channels and pain. Biochim Biophys Acta 2007; 1772: 978–988.
    1. Spicarová D, Palecek J. The role of spinal cord vanilloid (TRPV1) receptors in pain modulation. Physiol Res 2008; 57: S69–S77.
    1. Yang F, Guo J, Sun WL, Liu FY, Cai J, Xing GG, Wan Y. The induction of long-term potentiation in spinal dorsal horn after peripheral nociceptive stimulation and contribution of spinal TRPV1 in rats. Neuroscience 2014; 269: 59–66.
    1. Chen WH, Tzen JT, Hsieh CL, Chen YH, Lin TJ, Chen SY, Lin YW. Attenuation of TRPV1 and TRPV4 expression and function in mouse inflammatory pain models using electroacupuncture. Evid Based Complement Alternat Med 2012; 2012: 636848.
    1. Wang B, Aw TY, Stokes KY. The protection conferred against ischemia-reperfusion injury in the diabetic brain by N-acetylcysteine is associated with decreased dicarbonyl stress. Free Radic Biol Med 2016; 96: 89–98.
    1. Liao CY, Chung CH, Wu CC, Lin FH, Tsao CH, Wang CC, Chien WC. Protective effect of N-acetylcysteine on progression to end-stage renal disease: necessity for prospective clinical trial. Eur J Intern Med 2017; 44: 67–73.
    1. Yang CT, Meng FH, Chen L, Li X, Cen LJ, Wen YH, Li CC, Zhang H. Inhibition of methylglyoxal-induced AGEs/RAGE expression contributes to dermal protection by N-Acetyl-L-Cysteine. Cell Physiol Biochem 2017; 41: 742–754.
    1. Dludla PV, Nkambule BB, Dias SC, Johnson R. Cardioprotective potential of N-acetyl cysteine against hyperglycaemia-induced oxidative damage: a protocol for a systematic review. Syst Rev 2017; 6: 96.

Source: PubMed

3
Sottoscrivi