Cannabidiol modulates serotonergic transmission and reverses both allodynia and anxiety-like behavior in a model of neuropathic pain

Danilo De Gregorio, Ryan J McLaughlin, Luca Posa, Rafael Ochoa-Sanchez, Justine Enns, Martha Lopez-Canul, Matthew Aboud, Sabatino Maione, Stefano Comai, Gabriella Gobbi, Danilo De Gregorio, Ryan J McLaughlin, Luca Posa, Rafael Ochoa-Sanchez, Justine Enns, Martha Lopez-Canul, Matthew Aboud, Sabatino Maione, Stefano Comai, Gabriella Gobbi

Abstract

Clinical studies indicate that cannabidiol (CBD), the primary nonaddictive component of cannabis that interacts with the serotonin (5-HT)1A receptor, may possess analgesic and anxiolytic effects. However, its effects on 5-HT neuronal activity, as well as its impact on models of neuropathic pain are unknown. First, using in vivo single-unit extracellular recordings in rats, we demonstrated that acute intravenous (i.v.) increasing doses of CBD (0.1-1.0 mg/kg) decreased the firing rate of 5-HT neurons in the dorsal raphe nucleus, which was prevented by administration of the 5-HT1A antagonist WAY 100635 (0.3 mg/kg, i.v.) and the TRPV1 antagonist capsazepine (1 mg/kg, i.v.) but not by the CB1 receptor antagonist AM 251 (1 mg/kg, i.v.). Repeated treatment with CBD (5 mg/kg/day, subcutaneously [s.c.], for 7 days) increased 5-HT firing through desensitization of 5-HT1A receptors. Rats subjected to the spared nerve injury model for 24 days showed decreased 5-HT firing activity, mechanical allodynia, and increased anxiety-like behavior in the elevated plus maze test, open-field test, and novelty-suppressed feeding test. Seven days of treatment with CBD reduced mechanical allodynia, decreased anxiety-like behavior, and normalized 5-HT activity. Antiallodynic effects of CBD were fully prevented by capsazepine (10 mg/kg/day, s.c., for 7 days) and partially prevented by WAY 100635 (2 mg/kg/day, s.c., for 7 days), whereas the anxiolytic effect was blocked only by WAY. Overall, repeated treatment with low-dose CBD induces analgesia predominantly through TRPV1 activation, reduces anxiety through 5-HT1A receptor activation, and rescues impaired 5-HT neurotransmission under neuropathic pain conditions.

Conflict of interest statement

Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article.

Figures

Figure 1.
Figure 1.
Acute and repeated CBD administration on firing activity of DRN 5-HT neurons in naive rats. (A) Representation of coronal sections of the rat brain with the photomicrograph of the recording site in the DRN. Central gray dorsal (CGD); central gray lateral ventral (CGLV); aqueduct (Aq). The white arrow indicates the site of the electrode recording labeled with pontamine sky blue dye. (B) The typical spike waveform of 5-HT neuron. (C) Acute intravenous (i.v.) CBD administration decreases firing rate of DRN 5-HT neurons (n = 9), prevented by previous i.v. injection of WAY 100635 (WAY, n = 4) and CPZ (n = 4) but not by AM 251 (n = 4). Each point of the line represents mean ± SEM expressed as percentage of baseline before injections of veh, CBD, or antagonists. White arrow indicates the injection of veh, WAY, CPZ, or AM 251, and black arrow indicates the beginning of cumulative CBD injection. (D) WAY, CPZ, and AM 251 do not affect the basal 5-HT firing rate activity. (E-H) Representative firing rate histograms showing the acute response of 5-HT neurons to CBD alone (E) and with previous injection of WAY (F), CPZ (G), and AM 251 (H). Black arrows indicate sequence of a single injection of antagonists and of increasing doses of CBD. The cumulative doses are indicated on top of each arrow. Two-way ANOVA for RM followed by Bonferroni post hoc comparisons. *P < 0.05 and ***P < 0.001 vs veh; P < 0.001 vs CBD. (I) Mean firing DRN 5-HT activity of naive rats treated with veh (65 recorded neurons in 4 rats) or with CBD (5 mg/kg/day, subcutaneously [s.c.], for 7 days) (77 neurons recorded in 4 rats). Each bar represents mean ± SEM and each point represents a single neuron recorded in each group. The Student unpaired 2-tailed t test. ***P < 0.001 vs veh. (J) Simple linear regression analysis showing relationship between degree of suppression of 5-HT firing activity in the DRN and the dose of LSD administered i.v. in naive rats treated for 7 days with vehicle or CBD (5.0 mg/kg/day, s.c.). ANOVA, analysis of variance; CBD, cannabidiol; CPZ, capsazepine; DRN, dorsal raphe nucleus; N.S., not significant; RM, repeated measures.
Figure 2.
Figure 2.
Repeated CBD treatment increases paw withdrawal threshold and prevents anxiety-like behavior in the OFT, EPMT, and NSFT after SNI surgery. (A) Scheme illustrating the timing of SNI induction, behavioral and electrophysiological experiments. (B) Mechanical hypersensitivity was measured with von Frey filaments at day 15 (D0), 19 (D5), and 23 (D7) after SNI surgery. Sham rats treated with veh (n = 6) or CBD (5 mg/kg/day, subcutaneously [s.c.], for 7 days, n = 6) and SNI rats treated with veh (n = 9) and CBD (5 mg/kg/day, s.c., for 7 days, n = 9) were tested. Each point of the line represents mean ± SEM expressed as mean of 50% paw withdrawal threshold (g). Three-way ANOVA followed by Bonferroni post hoc comparisons. ***P < 0.001 vs sham/veh; ###P < 0.001 vs SNI/veh at D7. (C) Horizontal movement traces in the OFT of sham and SNI rats treated with veh or CBD (5 mg/kg/day, s.c., for 7 days). (D-F) Distance travelled (cm) (D), time spent (s) in the central area (E), and number of entries in the central area (F) of the OFT. Sham rats treated with veh (n = 9) or CBD (5 mg/kg/day, s.c., for 7 days, n = 9) and SNI rats treated with veh (n = 9) and CBD (5 mg/kg/day, s.c., for 7 days, n = 9) were tested. Each bar represents mean ± SEM. In D-F, each point represents data from an individual rat. Two-way ANOVA followed by Bonferroni post hoc comparisons. *P < 0.05 and ***P < 0.001 vs sham/veh; ###P < 0.001 vs SNI/veh. (G) Time of immobility (s) in the FST. (H and I) Percentage of time (%) (H) spent in the open arms in the EPMT and head dipping duration (s) (I). Each bar represents mean ± SEM. (J) Horizontal movement traces in the EPMT of sham and SNI rats treated with veh or CBD (5 mg/kg/day, s.c., for 7 days). (K and L) Latency to feed (s) in the novel (K) and familiar (L) environments in the NSFT. Each symbol represents mean ± SEM. Sham rats treated with veh (n = 8) or CBD (5 mg/kg/day, s.c., for 7 days, n = 9) and SNI rats treated with veh (n = 9) and CBD (5 mg/kg/day, s.c., for 7 days, n = 9) were tested. In G-I, each point represents data from an individual rat. Two-way ANOVA followed by Bonferroni post hoc comparisons. **P < 0.01 vs sham/veh., #P < 0.05, and ##P < 0.01 vs SNI/veh. ANOVA, analysis of variance; CBD, cannabidiol; EPMT, elevated plus maze test; FST, forced swim test; N.S., not significant; NSFT, novelty-suppressed feeding test; OFT, open-field test; SNI, spared nerve injury.
Figure 3.
Figure 3.
TRPV1 antagonism blocked the antinociceptive, but not the anxiolytic effects, of repeated CBD administration in SNI rats. (A) Scheme illustrating the timing of SNI induction, behavioral and electrophysiological experiments. (B) Mechanical hypersensitivity was measured with von Frey filaments at day 15 (D0), 19 (D5), and 23 (D7) after SNI surgery. Spared nerve injury rats treated with veh (n = 9), CBD alone (5 mg/kg/day, subcutaneously [s.c.], for 7 days, n = 9), or in combination with CPZ (10 mg/kg/day, s.c., for 7 days, 10 minutes before CBD, n = 8) and CPZ alone (10 mg/kg/day, s.c., for 7 days, n = 7) were tested. Each point of the line represents mean ± SEM expressed as mean of 50% paw withdrawal threshold (g). Two-way ANOVA for repeated measures followed by Bonferroni post hoc comparisons. ***P < 0.001 vs SNI/veh; ###P < 0.001 vs SNI/CPZ + CBD at D7. (C) Horizontal movement traces in the OFT of SNI rats treated with veh, CBD alone (5 mg/kg/day, s.c., for 7 days), or in combination with CPZ (10 mg/kg/day, s.c., for 7 days, 10 minutes before CBD) and CPZ alone (10 mg/kg/day, s.c., for 7 days). (D-F) Distance travelled (cm) (D), time spent (s) in the central area (E), and number of entries in the central area (F) of the OFT. Spared nerve injury rats treated with veh (n = 9), CBD alone (5 mg/kg/day, s.c., for 7 days, n = 9), or in combination with CPZ (10 mg/kg/day, s.c., for 7 days, 10 minutes before CBD, n = 8) and CPZ alone (2 mg/kg, s.c., for 7 days, n = 7) were tested. Each bar represents mean ± SEM. In D-F, each point represents data from an individual rat. One-way ANOVA followed by Bonferroni post hoc comparisons. *P < 0.05 and **P < 0.01 vs SNI/veh. (G) Percentage of time (%) spent in the open arms in the EPMT. Each bar represents mean ± SEM. (H) Horizontal movement traces in the EPMT of SNI rats treated with veh (n = 9), CBD alone (5 mg/kg/day, s.c., for 7 days, n = 9), or in combination with CPZ (10 mg/kg/day, s.c., for 7 days, 10 minutes before CBD, n = 8) and CPZ alone (10 mg/kg/day, s.c., n = 7) were tested. (I and J) Latency to feed (s) in the novel (I) and familiar (J) environments in the NSFT. Each symbol represents mean ± SEM. In G, each point represents data from an individual rat. One-way ANOVA followed by Bonferroni post hoc comparisons. *P < 0.05, **P < 0.01, and ***P < 0.001 vs SNI/veh. ANOVA, analysis of variance; CBD, cannabidiol; CPZ, capsazepine; EPMT, elevated plus maze test; N.S., not significant; NSFT, novelty-suppressed feeding test; OFT, open-field test; SNI, spared nerve injury.
Figure 4.
Figure 4.
5-HT1A antagonism partially prevented antinociception and completely blocked the anxiolytic properties of repeated CBD administration in SNI rats. (A) Scheme illustrating the timing of SNI induction, behavioral and electrophysiological experiments. (B) Mechanical hypersensitivity was measured with von Frey filaments at day 15 (D0), 19 (D5), and 23 (D7) after SNI surgery. Spared nerve injury rats treated with veh (n = 9), CBD alone (5 mg/kg/day, subcutaneously [s.c.], for 7 days, n = 9), or in combination with WAY (2 mg/kg/day, s.c., for 7 days, 10 minutes before CBD, n = 8) and WAY alone (2 mg/kg/day, s.c., for 7 days, n = 7) were tested. Each point of the line represents mean ± SEM expressed as mean of 50% paw withdrawal threshold (g). Two-way ANOVA for repeated measures followed by Bonferroni post hoc comparisons. ***P < 0.001 vs SNI/veh; ##P < 0.01 vs SNI/WAY + CBD at D7. (C) Horizontal movement traces in the OFT of SNI rats treated with veh, CBD alone (5 mg/kg/day, s.c., for 7 days), or in combination with CPZ (10 mg/kg/day, s.c., for 7 days, 10 minutes before CBD) and WAY alone (2 mg/kg/day, s.c., for 7 days). (D-F) Distance travelled (cm) (D), time spent (s) in the central area (E), and number of entries in the central area (F) of the OFT. Spared nerve injury rats treated with veh (n = 9), CBD alone (5 mg/kg/day, s.c., for 7 days, n = 9), or in combination with WAY (2 mg/kg/day, s.c., for 7 days, 10 minutes before CBD, n = 8) and WAY alone (2 mg/kg, s.c., for 7 days, n = 7) were tested. Each bar represents mean ± SEM. In D-F, each point represents data from an individual rat. One-way ANOVA followed by Bonferroni post hoc comparisons. **P < 0.01 vs SNI/veh; #P < 0.05; and ##P < 0.01 vs SNI/CBD. (G) Percentage of time (%) spent in the open arms in the EPMT. Each bar represents mean ± SEM. (H) Horizontal movement traces in the EPMT of SNI rats treated with veh (n = 9), CBD alone (5 mg/kg/day, s.c., for 7 days, n = 9), or in combination with WAY (2 mg/kg/day, s.c., for 7 days, 10 minutes before CBD, n = 8) and WAY alone (2 mg/kg/day, s.c., n = 7) were tested. (I and J) Latency to feed (s) in the novel (I) and familiar (J) environments in the NSFT. Each symbol represents mean ± SEM. In G, each point represents data from an individual rat. One-way ANOVA followed by Bonferroni post hoc comparisons. **P < 0.01 vs SNI/veh; #P < 0.05 vs SNI/CBD. ANOVA, analysis of variance; CBD, cannabidiol; CPZ, capsazepine; EPMT, elevated plus maze test; N.S., not significant; NSFT, novelty-suppressed feeding test; OFT, open-field test; SNI, spared nerve injury.
Figure 5.
Figure 5.
Repeated CBD treatment prevents SNI-induced alterations in DRN 5-HT neuronal activity. (A) Mean DRN 5-HT firing activity (spikes/s). (B) Representative firing rate histograms of the mean DRN 5-HT firing rate activity recorded in sham and SNI rats treated with veh or CBD (5.0 mg/kg/day, for 7 days, subcutaneously [s.c.]). (C) Mean DRN 5-HT COV (%). Each bar represents mean ± SEM. Two-way ANOVA followed by Bonferroni post hoc comparisons. (D) Representative rate histograms recorded from a DRN neuron excited (top) or not (bottom) by mechanical pinch to the operated hind paw. (E) Contingency interleaved bars showing percentage (%) of DRN 5-HT neurons excited or not by mechanical paw pinch stimulation. (F) Contingency stacked bars showing percentage (%) of bursting and nonbursting DRN 5-HT neurons (the χ2 test). (G) Mean number of DRN 5-HT neuron recorded per electrode descent. Sham rats treated with veh (n = 5) or CBD (5 mg/kg/day, s.c., for 7 days) (n = 5) and SNI rats treated with veh (n = 4) and CBD (5 mg/kg/day, s.c., for 7 days) (n = 7) were tested. Two-way ANOVA followed by Bonferroni post hoc comparisons. In A and C, each point represents a single neuron recorded in each group. In F, each point represents data from an individual rat. **P < 0.01, ***P < 0.001 vs sham/veh., #P < 0.05, and ###P < 0.001 vs SNI/veh. ANOVA, analysis of variance; CBD, cannabidiol; COV, coefficient of variation; DRN, dorsal raphe nucleus; SNI, spared nerve injury.

References

    1. Allers K, Sharp T. Neurochemical and anatomical identification of fast- and slow-firing neurones in the rat dorsal raphe nucleus using juxtacellular labelling methods in vivo. Neuroscience 2003;122:193–204.
    1. Bambico FR, Cassano T, Dominguez-Lopez S, Katz N, Walker CD, Piomelli D, Gobbi G. Genetic deletion of fatty acid amide hydrolase alters emotional behavior and serotonergic transmission in the dorsal raphe, prefrontal cortex, and hippocampus. Neuropsychopharmacol 2010;35:2083–100.
    1. Bambico FR, Katz N, Debonnel G, Gobbi G. Cannabinoids elicit antidepressant-like behavior and activate serotonergic neurons through the medial prefrontal cortex. J Neurosci 2007;27:11700–11.
    1. Bambico FR, Nguyen NT, Gobbi G. Decline in serotonergic firing activity and desensitization of 5-HT1A autoreceptors after chronic unpredictable stress. Eur Neuropsychopharmacol 2009;19:215–28.
    1. Bambico FR, Nguyen NT, Katz N, Gobbi G. Chronic exposure to cannabinoids during adolescence but not during adulthood impairs emotional behaviour and monoaminergic neurotransmission. Neurobiol Dis 2010;37:641–55.
    1. Baraban J, Aghajanian G. Noradrenergic innervation of serotonergic neurons in the dorsal raphe: demonstration by electron microscopic autoradiography. Brain Res 1981;204:1–11.
    1. Bardin L, Lavarenne J, Eschalier A. Serotonin receptor subtypes involved in the spinal antinociceptive effect of 5-HT in rats. PAIN 2000;86:11–18.
    1. Bergamaschi MM, Queiroz RHC, Chagas MHN, De Oliveira DCG, De Martinis BS, Kapczinski F, Quevedo J, Roesler R, Schröder N, Nardi AE. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology 2011;36:1219–26.
    1. Berlin NI, Huff RL, Van Dyke DC, Hennessy TG. The blood volume of the adult rat, as determined by Fe59 and P32 labelled red cells. Proc Soc Exp Biol Med 1949;71:176–8.
    1. Bisogno T, Hanuš L, De Petrocellis L, Tchilibon S, Ponde DE, Brandi I, Moriello AS, Davis JB, Mechoulam R, Di Marzo V. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol 2001;134:845–52.
    1. Blessing EM, Steenkamp MM, Manzanares J, Marmar CR. Cannabidiol as a potential treatment for anxiety disorders. Neurotherapeutics 2015;12:825–36.
    1. Blier P, De Montigny C. Electrophysiological investigations on the effect of repeated zimelidine administration on serotonergic neurotransmission in the rat. J Neurosci 1983;3:1270–8.
    1. Blier P, De Montigny C. Modification of 5-HT neuron properties by sustained administration of the 5-HT1A agonist gepirone: electrophysiological studies in the rat brain. Synapse 1987;1:470–80.
    1. Campos AC, Ferreira FR, Guimarães FS. Cannabidiol blocks long-lasting behavioral consequences of predator threat stress: possible involvement of 5HT1A receptors. J Psychiatr Res 2012;46:1501–10.
    1. Campos AC, Fogaça MV, Sonego AB, Guimarães FS. Cannabidiol, neuroprotection and neuropsychiatric disorders. Pharmacol Res 2016;112:119–27.
    1. Campos AC, Guimarães FS. Involvement of 5HT1A receptors in the anxiolytic-like effects of cannabidiol injected into the dorsolateral periaqueductal gray of rats. Psychopharmacology 2008;199:223.
    1. Campos AC, Moreira FA, Gomes FV, Del Bel EA, Guimaraes FS. Multiple mechanisms involved in the large-spectrum therapeutic potential of cannabidiol in psychiatric disorders. Philos Trans R Soc Lond B Biol Sci 2012;367:3364–78.
    1. Campos AC, Ortega Z, Palazuelos J, Fogaca MV, Aguiar DC, Diaz-Alonso J, Ortega-Gutierrez S, Vazquez-Villa H, Moreira FA, Guzman M, Galve-Roperh I, Guimaraes FS. The anxiolytic effect of cannabidiol on chronically stressed mice depends on hippocampal neurogenesis: involvement of the endocannabinoid system. Int J Neuropsychopharmacol 2013;16:1407–19.
    1. Chen J, Song Y, Yang J, Zhang Y, Zhao P, Zhu XJ, Su HC. The contribution of TNF-α in the amygdala to anxiety in mice with persistent inflammatory pain. Neurosci Lett 2013;541:275–80.
    1. Comai S, Ochoa-Sanchez R, Dominguez-Lopez S, Bambico FR, Gobbi G. Melancholic-like behaviors and circadian neurobiological abnormalities in melatonin MT1 receptor knockout mice. Int J Neuropsychopharmacol 2015;18:pyu075. 10.1093/ijnp/pyu075.
    1. Costa B, Colleoni M, Conti S, Parolaro D, Franke C, Trovato AE, Giagnoni G. Oral anti-inflammatory activity of cannabidiol, a non-psychoactive constituent of cannabis, in acute carrageenan-induced inflammation in the rat paw. Naunyn Schmiedebergs Arch Pharmacol 2004;369:294–9.
    1. Costa B, Giagnoni G, Franke C, Trovato AE, Colleoni M. Vanilloid TRPV1 receptor mediates the antihyperalgesic effect of the nonpsychoactive cannabinoid, cannabidiol, in a rat model of acute inflammation. Br J Pharmacol 2004;143:247–50.
    1. Costa B, Trovato AE, Comelli F, Giagnoni G, Colleoni M. The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. Eur J Pharmacol 2007;556:75–83.
    1. Crippa JAS, Derenusson GN, Ferrari TB, Wichert-Ana L, Duran FL, Martin-Santos R, Simões MV, Bhattacharyya S, Fusar-Poli P, Atakan Z. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J Psychopharmacol 2011;25:121–30.
    1. Dawson L, Nguyen H, Smith D, Schechter L. Effect of chronic fluoxetine and WAY-100635 treatment on serotonergic neurotransmission in the frontal cortex. J Psychopharmacol 2002;16:145–52.
    1. Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. PAIN 2000;87:149–58.
    1. Deiana S, Watanabe A, Yamasaki Y, Amada N, Arthur M, Fleming S, Woodcock H, Dorward P, Pigliacampo B, Close S. Plasma and brain pharmacokinetic profile of cannabidiol (CBD), cannabidivarine (CBDV), Δ 9-tetrahydrocannabivarin (THCV) and cannabigerol (CBG) in rats and mice following oral and intraperitoneal administration and CBD action on obsessive–compulsive behaviour. Psychopharmacology 2012;219:859–73.
    1. Devinsky O, Marsh E, Friedman D, Thiele E, Laux L, Sullivan J, Miller I, Flamini R, Wilfong A, Filloux F. Cannabidiol in patients with treatment-resistant epilepsy: an open-label interventional trial. Lancet Neurol 2016;15:270–8.
    1. Dixon WJ. Efficient analysis of experimental observations. Annu Rev Pharmacol Toxicol 1980;20:441–62.
    1. Domínguez-López S, Mahar I, Bambico FR, Labonté B, Ochoa-Sánchez R, Leyton M, Gobbi G. Short-term effects of melatonin and pinealectomy on serotonergic neuronal activity across the light–dark cycle. J Psychopharmacol 2012;26:830–44.
    1. Dong J, De Montigny C, Blier P. Assessment of the serotonin reuptake blocking property of YM992: electrophysiological studies in the rat hippocampus and dorsal raphe. Synapse 1999;34:277–89.
    1. El Mansari M, Sánchez C, Chouvet G, Renaud B, Haddjeri N. Effects of acute and long-term administration of escitalopram and citalopram on serotonin neurotransmission: an in vivo electrophysiological study in rat brain. Neuropsychopharmacology 2005;30:1269–77.
    1. Fogaça MV, Campos AC, Coelho LD, Duman RS, Guimarães FS. The anxiolytic effects of cannabidiol in chronically stressed mice are mediated by the endocannabinoid system: role of neurogenesis and dendritic remodeling. Neuropharmacology 2018;135:22–33.
    1. Ford A, Castonguay A, Cottet M, Little JW, Chen Z, Symons-Liguori AM, Doyle T, Egan TM, Vanderah TW, De Konnick Y. Engagement of the GABA to KCC2 signaling pathway contributes to the analgesic effects of A3AR agonists in neuropathic pain. J Neurosci 2015;35:6057–67.
    1. Garcia-Garcia AL, Newman-Tancredi A, Leonardo ED. P5-HT1A receptors in mood and anxiety: recent insights into autoreceptor versus heteroreceptor function. Psychopharmacology 2014;231:623–36.
    1. Gauldie S, McQueen D, Pertwee R, Chessell I. Anandamide activates peripheral nociceptors in normal and arthritic rat knee joints. Br J Pharmacol 2001;132:617–21.
    1. Gobbi G, Murphy DL, Lesch KP, Blier P. Modifications of the serotonergic system in mice lacking serotonin transporters: an in vivo electrophysiological study. J Pharmacol Exp Ther 2001;296:987–95.
    1. Gomes FV, Resstel LB, Guimarães FS. The anxiolytic-like effects of cannabidiol injected into the bed nucleus of the stria terminalis are mediated by 5-HT1A receptors. Psychopharmacology 2011;213:465–73.
    1. Graeff FG, Guimarães FS, De Andrade TG, Deakin JF. Role of 5-HT in stress, anxiety, and depression. Pharmacol Biochem Behav 1996;54:129–41.
    1. Guida F, Luongo L, Marmo F, Romano R, Iannotta M, Napolitano F, Belardo C, Marabese I, D'Aniello A, De Gregorio D. Palmitoylethanolamide reduces pain-related behaviors and restores glutamatergic synapses homeostasis in the medial prefrontal cortex of neuropathic mice. Mol Brain 2015;8:47.
    1. Guo B, Wang J, Yao H, Ren K, Chen J, Yang J, Cai G, Liu H, Fan Y, Wang W. Chronic inflammatory pain impairs mGluR5-mediated depolarization-induced suppression of excitation in the anterior cingulate cortex. Cereb Cortex 2017;28:2118–30.
    1. Haddjeri N, Blier P. Effect of neurokinin-1 receptor antagonists on the function of 5-HT and noradrenaline neurons. Neuroreport 2000;11:1323–7.
    1. Haddjeri N, de Montigny C, Blier P. Modulation of the firing activity of rat serotonin and noradrenaline neurons by (±) pindolol. Biol Psychiatry 1999;45:1163–9.
    1. Haddjeri N, De Montigny C, Curet O, Blier P. Effect of the reversible monoamine oxidase-A inhibitor befloxatone on the rat 5-hydroxytryptamine neurotransmission. Eur J Pharmacol 1998;343:179–92.
    1. Hassan S, Eldeeb K, Millns PJ, Bennett AJ, Alexander SP, Kendall DA. Cannabidiol enhances microglial phagocytosis via transient receptor potential (TRP) channel activation. Br J Pharmacol 2014;171:2426–39.
    1. Holmes A, Murphy DL, Crawley JN. Abnormal behavioral phenotypes of serotonin transporter knockout mice: parallels with human anxiety and depression. Biol Psychiatry 2003;54:953–9.
    1. Huestis M. Pharmacokinetics and metabolism of the plant cannabinoids, Δ 9-tetrahydrocannabinol, cannabidiol and cannabinol. Cannabinoids. New York, NY: Springer, 2005. p. 657–90.
    1. Iannotti FA, Hill CL, Leo A, Alhusaini A, Soubrane C, Mazzarella E, Russo E, Whalley BJ, Di Marzo V, Stephens GJ. Nonpsychotropic plant cannabinoids, cannabidivarin (CBDV) and cannabidiol (CBD), activate and desensitize transient receptor potential vanilloid 1 (TRPV1) channels in vitro: potential for the treatment of neuronal hyperexcitability. ACS Chem Neurosci 2014;5:1131–41.
    1. Ji G, Fu Y, Ruppert KA, Neugebauer V. Pain-related anxiety-like behavior requires CRF1 receptors in the amygdala. Mol Pain 2007;3:13.
    1. Kaplan JS, Stella N, Catterall WA, Westenbroek RE. Cannabidiol attenuates seizures and social deficits in a mouse model of Dravet syndrome. Proc Natl Acad Sci U S A 2017;114:11229–34.
    1. Komaki A, Hashemi-Firouzi N, Shojaei S, Souri Z, Heidari S, Shahidi S. Study the effect of endocannabinoid system on rat behavior in elevated plus-maze. Basic Clin Neurosci 2015;6:147.
    1. Kwiatkowska M, Parker LA, Burton P, Mechoulam R. A comparative analysis of the potential of cannabinoids and ondansetron to suppress cisplatin-induced emesis in the Suncus murinus (house musk shrew). Psychopharmacology 2004;174:254–9.
    1. Lesch KP, Bengel D, Heils A, Sabol SZ, Greenberg BD, Petri S, Benjamin J, Müller CR, Hamer DH, Murphy DL. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science 1996;274:1527–31.
    1. Lopez-Alvarez VM, Puigdomenech M, Navarro X, Cobianchi S. Monoaminergic descending pathways contribute to modulation of neuropathic pain by increasing-intensity treadmill exercise after peripheral nerve injury. Exp Neurol 2018;299:42–55.
    1. Mason P. Contributions of the medullary raphe and ventromedial reticular region to pain modulation and other homeostatic functions. Annu Rev Neurosci 2001;24:737–77.
    1. McPartland J, Glass M, Pertwee R. Meta-analysis of cannabinoid ligand binding affinity and receptor distribution: interspecies differences. Br J Pharmacol 2007;152:583–93.
    1. McWilliams LA, Cox BJ, Enns MW. Mood and anxiety disorders associated with chronic pain: an examination in a nationally representative sample. PAIN 2003;106:127–33.
    1. McWilliams LA, Goodwin RD, Cox BJ. Depression and anxiety associated with three pain conditions: results from a nationally representative sample. PAIN 2004;111:77–83.
    1. Mechoulam R, Parker LA, Gallily R. Cannabidiol: an overview of some pharmacological aspects. J Clin Pharmacol 2002;42:11S–9S.
    1. Narita M, Kaneko C, Miyoshi K, Nagumo Y, Kuzumaki N, Nakajima M, Nanjo K, Matsuzawa K, Yamazaki M, Suzuki T. Chronic pain induces anxiety with concomitant changes in opioidergic function in the amygdala. Neuropsychopharmacology 2006;31:739–50.
    1. Nurmikko TJ, Serpell MG, Hoggart B, Toomey PJ, Morlion BJ, Haines D. Sativex successfully treats neuropathic pain characterised by allodynia: a randomised, double-blind, placebo-controlled clinical trial. PAIN 2007;133:210–20.
    1. Palazzo E, Luongo L, Guida F, Marabese I, Romano R, Iannotta M, Rossi F, D'Aniello A, Stella L, Marmo F. D-Aspartate drinking solution alleviates pain and cognitive impairment in neuropathic mice. Amino Acids 2016;48:1553–67.
    1. Parker LA, Burton P, Sorge RE, Yakiwchuk C, Mechoulam R. Effect of low doses of Δ 9-tetrahydrocannabinol and cannabidiol on the extinction of cocaine-induced and amphetamine-induced conditioned place preference learning in rats. Psychopharmacology 2004;175:360–6.
    1. Parker LA, Kwiatkowska M, Mechoulam R. Delta-9-tetrahydrocannabinol and cannabidiol, but not ondansetron, interfere with conditioned retching reactions elicited by a lithium-paired context in Suncus murinus: an animal model of anticipatory nausea and vomiting. Physiol Behav 2006;87:66–71.
    1. Parker LA, Mechoulam R, Schlievert C. Cannabidiol, a non-psychoactive component of cannabis and its synthetic dimethylheptyl homolog suppress nausea in an experimental model with rats. Neuroreport 2002;13:567–70.
    1. Paxinos GAWC, Watson C. The rat brain atlas in stereotaxic coordinates. San Diego: Academic, 1998.
    1. Philpott HT, O'brien M, McDougall JJ. Attenuation of early phase inflammation by cannabidiol prevents pain and nerve damage in rat osteoarthritis. PAIN 2017;158:2442.
    1. Porsolt RD, Brossard G, Hautbois C, Roux S. Rodent models of depression: forced swimming and tail suspension behavioral despair tests in rats and mice. Curr Protoc Neurosci 2001;8:10A.
    1. Qin N, Neeper MP, Liu Y, Hutchinson TL, Lubin ML, Flores CM. TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root ganglion neurons. J Neurosci 2008;28:6231–8.
    1. Qing-Ping W, Nakai Y. The dorsal raphe: an important nucleus in pain modulation. Brain Res Bull 1994;34:575–85.
    1. Ressler KJ, Nemeroff CB. Role of serotonergic and noradrenergic systems in the pathophysiology of depression and anxiety disorders. Depress Anxiety 2000;12:2–19.
    1. Rock E, Bolognini D, Limebeer C, Cascio M, Anavi-Goffer S, Fletcher P, Mechoulam R, Pertwee R, Parker L. Cannabidiol, a non-psychotropic component of cannabis, attenuates vomiting and nausea-like behaviour via indirect agonism of 5-HT1A somatodendritic autoreceptors in the dorsal raphe nucleus. Br J Pharmacol 2012;165:2620–34.
    1. Rock EM, Limebeer CL, Petrie GN, Williams LA, Mechoulam R, Parker LA. Effect of prior foot shock stress and Δ9-tetrahydrocannabinol, cannabidiolic acid, and cannabidiol on anxiety-like responding in the light-dark emergence test in rats. Psychopharmacology 2017;234:2207–17.
    1. Roeska K, Doods H, Arndt K, Treede RD, Ceci A. Anxiety-like behaviour in rats with mononeuropathy is reduced by the analgesic drugs morphine and gabapentin. PAIN 2008;139:349–57.
    1. Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res 2005;30:1037–43.
    1. Ryz NR, Remillard DJ, Russo EB. Cannabis roots: a traditional therapy with future potential for treating inflammation and pain. Cannabis Cannabinoid Res 2017;2:210–16.
    1. Sagheddu C, Aroni S, De Felice M, Lecca S, Luchicchi A, Melis M, Muntoni AL, Romano R, Palazzo E, Guida F. Enhanced serotonin and mesolimbic dopamine transmissions in a rat model of neuropathic pain. Neuropharmacology 2015;97:383–93.
    1. Sales AJ, Fogaça MV, Sartim AG, Pereira VS, Wegener G, Guimarães FS, Joca SR. Cannabidiol induces rapid and sustained antidepressant-like effects through increased BDNF signaling and synaptogenesis in the prefrontal cortex. Mol Neurobiol 2018. 10.1007/s12035-018-1143-4. [Epub ahead of print].
    1. Samara E, Bialer M, Mechoulam R. Pharmacokinetics of cannabidiol in dogs. Drug Metab disposition 1988;16:469–72.
    1. Schweimer J, Ungless M. Phasic responses in dorsal raphe serotonin neurons to noxious stimuli. Neuroscience 2010;171:1209–15.
    1. Scott KM, Bruffaerts R, Tsang A, Ormel J, Alonso J, Angermeyer M, Benjet C, Bromet E, De Girolamo G, De Graaf R. Depression–anxiety relationships with chronic physical conditions: results from the World Mental Health Surveys. J Affect Dis 2007;103:113–20.
    1. Scuderi C, Filippis DD, Iuvone T, Blasio A, Steardo A, Esposito G. Cannabidiol in medicine: a review of its therapeutic potential in CNS disorders. Phytother Res 2009;23:597–602.
    1. Tal M, Bennett GJ. Neuropathic pain sensations are differentially sensitive to dextrorphan. Neuroreport 1994;5:1438–40.
    1. Tsang A, Von Korff M, Lee S, Alonso J, Karam E, Angermeyer MC, Borges GLG, Bromet EJ, De Girolamo G, De Graaf R. Common chronic pain conditions in developed and developing countries: gender and age differences and comorbidity with depression-anxiety disorders. J Pain 2008;9:883–91.
    1. Wade DT, Robson P, House H, Makela P, Aram J. A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clin Rehabil 2003;17:21–9.
    1. Ward SJ, McAllister SD, Kawamura R, Murase R, Neelakantan H, Walker EA. Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT1A receptors without diminishing nervous system function or chemotherapy efficacy. Br J Pharmacol 2014;171:636–45.
    1. Ward SJ, Ramirez MD, Neelakantan H, Walker EA. Cannabidiol prevents the development of cold and mechanical allodynia in paclitaxel-treated female C57Bl6 mice. Anesth Analg 2011;113:947.
    1. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, Keurentjes JC, Lang S, Misso K, Ryder S. Cannabinoids for medical use: a systematic review and meta-analysis. JAMA 2015;313:2456–73.
    1. Wolfe F, Russell I, Vipraio G, Ross K, Anderson J. Serotonin levels, pain threshold, and fibromyalgia symptoms in the general population. J Rheumatol 1997;24:555–9.
    1. Wu Y, Yao X, Jiang Y, He X, Shao X, Du J, Shen Z, He Q, Fang J. Pain aversion and anxiety-like behavior occur at different times during the course of chronic inflammatory pain in rats. J Pain Res 2017;10:2585.
    1. Zanelati TV, Biojone C, Moreira FA, Guimaraes FS, Joca SR. Antidepressant-like effects of cannabidiol in mice: possible involvement of 5-HT1A receptors. Br J Pharmacol 2010;159:122–8.

Source: PubMed

3
Abonnere