Olorinab (APD371), a peripherally acting, highly selective, full agonist of the cannabinoid receptor 2, reduces colitis-induced acute and chronic visceral hypersensitivity in rodents

Joel Castro, Sonia Garcia-Caraballo, Jessica Maddern, Gudrun Schober, Amanda Lumsden, Andrea Harrington, Shirdi Schmiel, Beatriz Lindstrom, John Adams, Stuart M Brierley, Joel Castro, Sonia Garcia-Caraballo, Jessica Maddern, Gudrun Schober, Amanda Lumsden, Andrea Harrington, Shirdi Schmiel, Beatriz Lindstrom, John Adams, Stuart M Brierley

Abstract

Abdominal pain is a key symptom of inflammatory bowel disease and irritable bowel syndrome, for which there are inadequate therapeutic options. We tested whether olorinab-a highly selective, full agonist of the cannabinoid receptor 2 (CB2)-reduced visceral hypersensitivity in models of colitis and chronic visceral hypersensitivity (CVH). In rodents, colitis was induced by intrarectal administration of nitrobenzene sulfonic acid derivatives. Control or colitis animals were administered vehicle or olorinab (3 or 30 mg/kg) twice daily by oral gavage for 5 days, starting 1 day before colitis induction. Chronic visceral hypersensitivity mice were administered olorinab (1, 3, 10, or 30 mg/kg) twice daily by oral gavage for 5 days, starting 24 days after colitis induction. Visceral mechanosensitivity was assessed in vivo by quantifying visceromotor responses (VMRs) to colorectal distension. Ex vivo afferent recordings determined colonic nociceptor firing evoked by mechanical stimuli. Colitis and CVH animals displayed significantly elevated VMRs to colorectal distension and colonic nociceptor hypersensitivity. Olorinab treatment significantly reduced VMRs to control levels in colitis and CVH animals. In addition, olorinab reduced nociceptor hypersensitivity in colitis and CVH states in a concentration- and CB2-dependent manner. By contrast, olorinab did not alter VMRs nor nociceptor responsiveness in control animals. Cannabinoid receptor 2 mRNA was detected in colonic tissue, particularly within epithelial cells, and dorsal root ganglia, with no significant differences between healthy, colitis, and CVH states. These results demonstrate that olorinab reduces visceral hypersensitivity through CB2 agonism in animal models, suggesting that olorinab may provide a novel therapy for inflammatory bowel disease- and irritable bowel syndrome-associated abdominal pain.

Trial registration: ClinicalTrials.gov NCT03155945.

Conflict of interest statement

S. Schmiel, B. Lindstrom, and J. Adams are employees of Arena Pharmaceuticals, Inc. S.M. Brierley received research funding from Arena Pharmaceuticals to conduct the study. The remaining authors have no conflicts of interest to declare. Data from this study were previously presented in part at Digestive Disease Week (DDW) on May 2, 2020, as a virtual ePoster; DDW 2019 on May 18 to 21, 2019, in San Diego, CA; American Neurogastroenterology and Motility Society (ANMS) Annual Meeting on August 16 to 18, 2019, in Chicago, IL; United European Gastroenterology (UEG) Week on October 11, 2020, as a virtual presentation; and UEGW on October 19 to 23, 2019, in Barcelona, Spain.

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

Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the International Association for the Study of Pain.

Figures

Figure 1.
Figure 1.
Study design schematic: Overview of the timing of colitis induction with intracolonic TNBS or DNBS administration, oral administration of olorinab or vehicle, and VMR recordings are outlined for (A) IBD (colitis) and (B) IBS (CVH) rodent models. BID, twice daily; CVH, chronic visceral hypersensitivity; DNBS, 2,4-dinitrobenzene sulfonic acid; IBD, inflammatory bowel disease; IBS, irritable bowel syndrome; TNBS, trinitrobenzene sulfonic acid; VMR, visceromotor response.
Figure 2.
Figure 2.
Olorinab reversed colitis-induced hypersensitivity to CRD in colitis animals but had no effect in healthy animals. (A) Representative examples of EMG signals in response to each distension pressure for all animal cohorts. (B) In healthy rats, compared with vehicle treatment, olorinab administration at the highest dose of 30 mg/kg failed to alter the VMR to CRD. Comparison was not significant based on the generalized estimating equation method followed by the LSD post hoc test (P > 0.05). (C) Total AUC (sum of the AUC obtained at all distension pressures) of the VMR to CRD showed no difference in response between healthy rats treated with vehicle or the highest dose of olorinab (30 mg/kg). Comparison was not significant using a 2-tailed unpaired t test (P > 0.05). (D) Vehicle-treated colitis rats exhibited significantly enhanced VMR to CRD compared with vehicle-treated healthy control rats. Significant increases in colitis rats were observed across all distension pressures from 40 mm Hg. Olorinab treatment of colitis rats with doses of either 3 mg/kg or 30 mg/kg significantly reduced VMR to CRD relative to vehicle-treated colitis rats. Comparisons were performed using the generalized estimating equation method followed by the LSD post hoc test. **P < 0.01; †P < 0.001; ‡P < 0.0001. (E) Total AUC (sum of the AUC obtained at all distension pressures) of the VMR to CRD shows significantly elevated responses in colitis rats compared with control rats. Olorinab 3 mg/kg and 30 mg/kg significantly reduced the total AUC of the VMR to CRD relative to vehicle-treated colitis rats. Comparisons were performed using a one-way ANOVA followed by a Tukey multiple comparisons test. *P < 0.05; **P < 0.01; †P < 0.001. (F) No significant changes in colonic compliance were observed between healthy rats treated with vehicle and colitis rats treated with vehicle or olorinab at either dose. (G) No significant changes in colonic compliance were observed between healthy rats treated with vehicle or olorinab 30 mg/kg. All compliance comparisons were performed using the generalized estimating equation method followed by the LSD post hoc test, and results were not significant (P > 0.05). Data are presented as mean ± SEM. AUC was calculated as the difference of area values obtained predistension (20 seconds) minus those obtained during distension (20 seconds). aSum of the AUC obtained at all distension pressures. ANOVA, analysis of variance; AUC, area under the curve; CRD, colorectal distension; EMG, electromyography; LSD, least squares difference; VMR, visceromotor response.
Figure 3.
Figure 3.
Olorinab reversed hypersensitivity to CRD in CVH mice but had no effect in healthy mice. (A) Representative examples of EMG signals in response to each distension pressure for all mouse cohorts. (B) Vehicle-treated CVH mice exhibited significantly enhanced VMR to CRD compared with vehicle-treated healthy control mice. Significant increases in CVH mice were observed for distension pressures higher than 40 mm Hg. Olorinab treatment of CVH mice with doses of 3 mg/kg, 10 mg/kg, or 30 mg/kg significantly reduced VMR to CRD relative to vehicle-treated colitis mice. Olorinab 1 mg/kg was not effective in reducing VMR to CRD in CVH mice. Comparisons were performed with generalized estimating equation using an LSD post hoc test. **P < 0.01; ‡P < 0.0001. (C) Total AUC of the VMR to CRD showed significantly elevated responses in CVH mice compared with control mice. Olorinab 3 mg/kg, 10 mg/kg, and 30 mg/kg significantly reduced the total AUC of the VMR to CRD relative to vehicle-treated CVH mice. Comparisons were performed using a one-way ANOVA followed by Tukey post hoc tests. *P < 0.05; †P < 0.001; ‡P < 0.0001. (D) No significant changes in colonic compliance were observed between healthy mice treated with vehicle and CVH mice treated with vehicle or olorinab at all doses. Data are presented as mean ± SEM. AUC was calculated as the difference of area values obtained predistension (20 seconds) minus those obtained during distension (20 seconds). aSum of all distension pressures. ANOVA, analysis of variance; AUC, area under the curve; CRD, colorectal distension; CVH, chronic visceral hypersensitivity; EMG, electromyography; LSD, least squares difference; VMR, visceromotor response.
Figure 4.
Figure 4.
Olorinab had no effect on colonic nociceptors from healthy mice. (A) The mechanosensitive response of colonic nociceptors from healthy mice was unaffected by increasing concentrations of olorinab. Comparisons were performed using a one-way ANOVA followed by the Bonferroni post hoc test (P > 0.05). (B) The change in baseline response of healthy nociceptors in the presence of increasing concentrations of olorinab indicated no effect of olorinab on nociceptor firing even at high concentrations. (C) Single-unit colonic nociceptor recordings from healthy mice showed mechanical responsiveness was unchanged with olorinab application. Data are presented as mean ± SEM. ANOVA, analysis of variance; vfh, von Frey hair.
Figure 5.
Figure 5.
Olorinab dose dependently inhibited colonic nociceptors from colitis mice through a CB2-dependent mechanism. (A) Application of increasing concentrations of olorinab to ex vivo colonic nociceptor endings isolated from colitis mice caused a dose-dependent decrease in action potential firing in response to mechanical stimulation (2 g vfh). Comparisons were performed using a one-way ANOVA followed by the Bonferroni post hoc test (*P < 0.05; **P < 0.01; †P < 0.001). (B) The change in colitis colonic nociceptor mechanosensitivity induced by olorinab compared with baseline responses indicated a dose-dependent decrease in nociceptor firing with increasing concentrations of olorinab. (C) Single-unit colonic nociceptor recordings from colitis mice showed mechanical responsiveness at baseline and dose-dependent inhibition with increasing concentrations of olorinab. (D) Application of the CB2 antagonist SR144528 had no effect on the baseline mechanosensitivity of colitis colonic nociceptors and prevented olorinab-induced inhibition of nociceptor hypersensitivity. Comparisons were performed using a one-way ANOVA followed by the Bonferroni post hoc test (P > 0.05). (E) The change in colitis colonic nociceptor mechanosensitivity compared with baseline demonstrated no inhibitory action of olorinab in the presence of the CB2 antagonist. (F) Single-unit colonic nociceptor recordings from colitis mice showed mechanical responsiveness was unchanged with olorinab application in the presence of the CB2 antagonist. Data are presented as mean ± SEM. ANOVA, analysis of variance; CB2, cannabinoid receptor 2; IBD, inflammatory bowel disease; vfh, von Frey hair.
Figure 6.
Figure 6.
Olorinab dose dependently inhibited colonic nociceptors from CVH mice through a CB2-dependent mechanism. (A) Ex vivo application of increasing concentrations of olorinab to colonic nociceptor endings isolated from CVH mice caused a decrease in action potential firing in response to mechanical stimulation (2 g vfh; *P < 0.05, **P < 0.01, †P < 0.001). (B) The change in CVH colonic nociceptor mechanosensitivity induced by olorinab compared with baseline responses indicated a dose-dependent decrease in nociceptor response with increasing concentrations of olorinab. (C) Single-unit colonic nociceptor recordings from CVH mice showed mechanical responsiveness at baseline and dose-dependent inhibition with increasing concentrations of olorinab. (D) Application of the CB2 antagonist SR144528 had no effect on the baseline mechanosensitivity of CVH colonic nociceptors and prevented olorinab-induced inhibition of nociceptor action potential firing in response to mechanical stimulation (2 g vfh; P > 0.05). (E) The change in CVH colonic nociceptor mechanosensitivity induced by olorinab compared with baseline demonstrated no inhibitory action of olorinab in the presence of the CB2 antagonist. (F) Single-unit colonic nociceptor recordings from CVH mice showed the mechanical responsiveness was unchanged with olorinab application in the presence of the CB2 antagonist. Data are presented as mean ± SEM. Statistical analysis was performed using a one-way ANOVA with Bonferroni post hoc tests. ANOVA, analysis of variance; CB2, cannabinoid receptor 2; CVH, colonic visceral hypersensitivity; IBS, irritable bowel syndrome; vfh, von Frey hair.
Figure 7.
Figure 7.
CB1 and CB2 mRNA expression in tissue from healthy, colitis, and CVH mice. (A) CB2 (CB2A+B isoforms) was predominantly expressed over CB1 in the colonic mucosa from healthy, colitis, and CVH mice, with CB2A as the most prevalent CB2 isoform. (B) In the colonic longitudinal and circular muscle also containing the myenteric plexus (ENS) from healthy, colitis, and CVH mice, CB1 had the highest relative abundance of all CB receptor transcripts compared with CB2A+B, CB2A, and CB2B. (C) In TL and LS DRG from healthy, colitis, and CVH mice, CB1 had the highest relative abundance of all CB receptor transcripts. CB2 mRNA was also detected in the TL and LS DRG, with the CB2A isoform as the predominantly expressed CB2 isoform. Expression profiles did not significantly differ between healthy, colitis, and CVH states in the (A) colonic mucosa, (B) colonic muscle + ENS, or (C) TL or LS DRG (P > 0.05). All comparisons shown were performed using a one-way ANOVA followed by the Tukey multiple comparison post hoc test (*P < 0.05; **P < 0.01; †P < 0.001; ‡P < 0.0001). CB1 and CB2 mRNA expression was measured relative to reference gene mRNA expression (Ppia, Gapdh, and β-actin) quantified by geometric mean. Data are presented as mean ± SEM. ANOVA, analysis of variance; CB1, cannabinoid receptor 1; CB2, cannabinoid receptor 2; DRG, dorsal root ganglia; ENS, enteric nervous system; Gapdh, glyceraldehyde-3-phosphate dehydrogenase; IBD, inflammatory bowel disease; IBS, irritable bowel syndrome; LS, lumbosacral; mRNA, messenger ribonucleic acid; Ppia, peptidylprolyl isomerase A; TL, thoracolumbar.
Figure 8.
Figure 8.
ISH analysis of CB2 mRNA expression in the mouse colon: (A) Positive control tissue for CB2 shows abundant expression of CB2 mRNA using ISH in cross-sections of the spleen (left panel), whereas the negative control probe (dapB) shows a lack of punctate staining (right panel). (B) CB2 labeling in the colon from a healthy mouse was prominent within epithelial cells lining the lumen edge and crypts (see arrows and inset) with sparser labeling observed in the lamina propria, muscularis mucosae, and myenteric plexus. Representative examples of in situ hybridizations for CB2 on cross-sections of the colon from (C) healthy, (D) colitis, and (E) CVH mice. (F) Quantitative analysis revealed that there was no significant difference in CB2 expression when expressed as CB2-positive area per section (left panel), average CB2-positive area per section (middle panel), or CB2-positive area per mouse (right panel). All comparisons shown were performed using a one-way ANOVA followed by the Tukey multiple comparison post hoc test (P > 0.05). Data are presented as mean ± SEM. CB2, cannabinoid receptor 2; cm, circular muscle; CVH, chronic visceral hypersensitivity; dapB, dihydrodipicolinate reductase; ISH, in situ hybridization; lm, longitudinal muscle; lp, lamina propria; LS, lumbosacral; mm, muscularis mucosae; mp, myenteric plexus.
Figure 9.
Figure 9.
ISH of CB2 mRNA expression in DRG and proposed mechanism of action of olorinab: (A–C) Representative images of sections of thoracolumbar DRG from healthy mice that underwent hematoxylin staining (blue) and ISH labeling for (A, B) CB2 (brown dots) or (C) the negative probe dapB. Representative images of CB2 labeling in sections of (D) thoracolumbar and (E) lumbosacral DRG from healthy, acute colitis, and CVH mice. Scale bars = 20 μm. (F) Hypothesized mechanism of action of olorinab, which is a highly selective full agonist for CB2 that exhibits low brain penetration. To modulate abdominal pain, olorinab may activate CB2 located on one or multiple cell types including epithelial cells, immune cells, and afferent nerves within the gastrointestinal wall. Downstream or direct effects of CB2 activation may then reduce action potential firing of colonic nociceptors. This would reduce the nociceptive signal being sent from the gastrointestinal tract to the spinal cord, where this nociceptive information is processed and then sent to the brain where pain is perceived. CB2, cannabinoid receptor 2; CVH, chronic visceral hypersensitivity; DRG, dorsal root ganglia; ISH, in situ hybridization; LS, lumbosacral; TL, thoracolumbar.

References

    1. Adam B, Liebregts T, Gschossmann JM, Krippner C, Scholl F, Ruwe M, Holtmann G. Severity of mucosal inflammation as a predictor for alterations of visceral sensory function in a rat model. PAIN 2006;123:179–86.
    1. Adams JW, Unett D, Anthony T, Gaitlin J, Gaidarov I. APD371: a Potent, highly selective, full agonist of the human CB2 receptor with sustained analgesic effects in rodents. J Pain 2018;19(S2).
    1. American Gastroenterological Association. IBS in America. Survey summary findings. Vol. 2020. Bethesda, MD: American Gastroenterological Association, 2015.
    1. Bellono NW, Bayrer JR, Leitch DB, Castro J, Zhang C, O'Donnell TA, Brierley SM, Ingraham HA, Julius D. Enterochromaffin cells are gut chemosensors that couple to sensory neural pathways. Cell 2017;170:185–98.e116.
    1. Borrelli F, Romano B, Petrosino S, Pagano E, Capasso R, Coppola D, Battista G, Orlando P, Di Marzo V, Izzo AA. Palmitoylethanolamide, a naturally occurring lipid, is an orally effective intestinal anti-inflammatory agent. Br J Pharmacol 2015;172:142–58.
    1. Brierley SM, Jones RC, III, Gebhart GF, Blackshaw LA. Splanchnic and pelvic mechanosensory afferents signal different qualities of colonic stimuli in mice. Gastroenterology 2004;127:166–78.
    1. Brierley SM, Linden DR. Neuroplasticity and dysfunction after gastrointestinal inflammation. Nat Rev Gastroenterol Hepatol 2014;11:611–27.
    1. Brust A, Croker DE, Colless B, Ragnarsson L, Andersson A, Jain K, Garcia-Caraballo S, Castro J, Brierley SM, Alewood PF, Lewis RJ. Conopeptide-derived kappa-opioid agonists (conorphins): potent, selective, and metabolic stable dynorphin a mimetics with antinociceptive properties. J Med Chem 2016;59:2381–95.
    1. Cain KC, Headstrom P, Jarrett ME, Motzer SA, Park H, Burr RL, Surawicz CM, Heitkemper MM. Abdominal pain impacts quality of life in women with irritable bowel syndrome. Am J Gastroenterol 2006;101:124–32.
    1. Camilleri M. Physiological underpinnings of irritable bowel syndrome: neurohormonal mechanisms. J Physiol 2014;592:2967–80.
    1. Carstens BB, Berecki G, Daniel JT, Lee HS, Jackson KA, Tae HS, Sadeghi M, Castro J, O'Donnell T, Deiteren A, Brierley SM, Craik DJ, Adams DJ, Clark RJ. Structure-activity studies of cysteine-rich alpha-conotoxins that inhibit high-voltage-activated calcium channels via GABA(B) receptor activation reveal a minimal functional motif. Angew Chem Int Ed Engl 2016;55:4692–6.
    1. Castro J, Grundy L, Deiteren A, Harrington AM, O'Donnell T, Maddern J, Moore J, Garcia-Caraballo S, Rychkov GY, Yu R, Kaas Q, Craik DJ, Adams DJ, Brierley SM. Cyclic analogues of alpha-conotoxin Vc1.1 inhibit colonic nociceptors and provide analgesia in a mouse model of chronic abdominal pain. Br J Pharmacol 2018;175:2384–98.
    1. Castro J, Harrington AM, Garcia-Caraballo S, Maddern J, Grundy L, Zhang J, Page G, Miller PE, Craik DJ, Adams DJ, Brierley SM. alpha-Conotoxin Vc1.1 inhibits human dorsal root ganglion neuroexcitability and mouse colonic nociception via GABAB receptors. Gut 2017;66:1083–94.
    1. Castro J, Harrington AM, Hughes PA, Martin CM, Ge P, Shea CM, Jin H, Jacobson S, Hannig G, Mann E, Cohen MB, MacDougall JE, Lavins BJ, Kurtz CB, Silos-Santiago I, Johnston JM, Currie MG, Blackshaw LA, Brierley SM. Linaclotide inhibits colonic nociceptors and relieves abdominal pain via guanylate cyclase-C and extracellular cyclic guanosine 3',5'-monophosphate. Gastroenterology 2013;145:1334–6.e1331-1311.
    1. Castro J, Harrington AM, Lieu T, Garcia-Caraballo S, Maddern J, Schober G, O'Donnell T, Grundy L, Lumsden AL, Miller P, Ghetti A, Steinhoff MS, Poole DP, Dong X, Chang L, Bunnett NW, Brierley SM. Activation of pruritogenic TGR5, MrgprA3, and MrgprC11 on colon-innervating afferents induces visceral hypersensitivity. JCI Insight 2019;4:e131712.
    1. Cremon C, Stanghellini V, Barbaro MR, Cogliandro RF, Bellacosa L, Santos J, Vicario M, Pigrau M, Alonso Cotoner C, Lobo B, Azpiroz F, Bruley des Varannes S, Neunlist M, DeFilippis D, Iuvone T, Petrosino S, Di Marzo V, Barbara G. Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Aliment Pharmacol Ther 2017;45:909–22.
    1. de Araujo AD, Mobli M, Castro J, Harrington AM, Vetter I, Dekan Z, Muttenthaler M, Wan J, Lewis RJ, King GF, Brierley SM, Alewood PF. Selenoether oxytocin analogues have analgesic properties in a mouse model of chronic abdominal pain. Nat Commun 2014;5:3165.
    1. Desormeaux C, Bautzova T, Garcia-Caraballo S, Rolland C, Barbaro MR, Brierley SM, Barbara G, Vergnolle N, Cenac N. Protease-activated receptor 1 is implicated in irritable bowel syndrome mediators-induced signaling to thoracic human sensory neurons. PAIN 2018;159:1257–67.
    1. Dothel G, Chang L, Shih W, Barbaro MR, Cremon C, Stanghellini V, De Ponti F, Mayer EA, Barbara G, Sternini C. micro-opioid receptor, beta-endorphin, and cannabinoid receptor-2 are increased in the colonic mucosa of irritable bowel syndrome patients. Neurogastroenterol Motil 2019;31:e13688.
    1. Duncan M, Mouihate A, Mackie K, Keenan CM, Buckley NE, Davison JS, Patel KD, Pittman QJ, Sharkey KA. Cannabinoid CB2 receptors in the enteric nervous system modulate gastrointestinal contractility in lipopolysaccharide-treated rats. Am J Physiol Gastrointest Liver Physiol 2008;295:G78–87.
    1. Enck P, Aziz Q, Barbara G, Farmer AD, Fukudo S, Mayer EA, Niesler B, Quigley EM, Rajilic-Stojanovic M, Schemann M, Schwille-Kiuntke J, Simren M, Zipfel S, Spiller RC. Irritable bowel syndrome. Nat Rev Dis Primers 2016;2:16014.
    1. Engel MA, Kellermann CA, Burnat G, Hahn EG, Rau T, Konturek PC. Mice lacking cannabinoid CB1-, CB2-receptors or both receptors show increased susceptibility to trinitrobenzene sulfonic acid (TNBS)-induced colitis. J Physiol Pharmacol 2010;61:89–97.
    1. Grundy L, Erickson A, Brierley SM. Visceral pain. Annu Rev Physiol 2019;81:261–84.
    1. Grundy L, Harrington AM, Castro J, Garcia-Caraballo S, Deiteren A, Maddern J, Rychkov GY, Ge P, Peters S, Feil R, Miller P, Ghetti A, Hannig G, Kurtz CB, Silos-Santiago I, Brierley SM. Chronic linaclotide treatment reduces colitis-induced neuroplasticity and reverses persistent bladder dysfunction. JCI Insight 2018;3:e121841.
    1. Han S, Thatte J, Buzard DJ, Jones RM. Therapeutic utility of cannabinoid receptor type 2 (CB(2)) selective agonists. J Med Chem 2013;56:8224–56.
    1. Han S, Thoresen L, Jung JK, Zhu X, Thatte J, Solomon M, Gaidarov I, Unett DJ, Yoon WH, Barden J, Sadeque A, Usmani A, Chen C, Semple G, Grottick AJ, Al-Shamma H, Christopher R, Jones RM. Discovery of APD371: identification of a highly potent and selective CB2 agonist for the treatment of chronic pain. ACS Med Chem Lett 2017;8:1309–13.
    1. Harvey BS, Nicotra LL, Vu M, Smid SD. Cannabinoid CB2 receptor activation attenuates cytokine-evoked mucosal damage in a human colonic explant model without changing epithelial permeability. Cytokine 2013;63:209–17.
    1. Hillsley K, McCaul C, Aerssens J, Peeters PJ, Gijsen H, Moechars D, Coulie B, Grundy D, Stead RH. Activation of the cannabinoid 2 (CB2) receptor inhibits murine mesenteric afferent nerve activity. Neurogastroenterol Motil 2007;19:769–77.
    1. Hockley JRF, Taylor TS, Callejo G, Wilbrey AL, Gutteridge A, Bach K, Winchester WJ, Bulmer DC, McMurray G, Smith ESJ. Single-cell RNAseq reveals seven classes of colonic sensory neuron. Gut 2019;68:633–44.
    1. Hua T, Li X, Wu L, Iliopoulos-Tsoutsouvas C, Wang Y, Wu M, Shen L, Johnston CA, Nikas SP, Song F, Song X, Yuan S, Sun Q, Wu Y, Jiang S, Grim TW, Benchama O, Stahl EL, Zvonok N, Zhao S, Bohn LM, Makriyannis A, Liu ZJ. Activation and signaling mechanism revealed by cannabinoid receptor-gi complex structures. Cell 2020;180:655–65 e618.
    1. Hughes PA, Brierley SM, Martin CM, Brookes SJ, Linden DR, Blackshaw LA. Post-inflammatory colonic afferent sensitisation: different subtypes, different pathways and different time courses. Gut 2009;58:1333–41.
    1. Hughes PA, Castro J, Harrington AM, Isaacs N, Moretta M, Hicks GA, Urso DM, Brierley SM. Increased kappa-opioid receptor expression and function during chronic visceral hypersensitivity. Gut 2014;63:1199–200.
    1. Inserra MC, Israel MR, Caldwell A, Castro J, Deuis JR, Harrington AM, Keramidas A, Garcia-Caraballo S, Maddern J, Erickson A, Grundy L, Rychkov GY, Zimmermann K, Lewis RJ, Brierley SM, Vetter I. Multiple sodium channel isoforms mediate the pathological effects of Pacific ciguatoxin-1. Sci Rep 2017;7:42810.
    1. Iwata Y, Ando K, Taniguchi K, Koba N, Sugiura A, Sudo M. Identification of a highly potent and selective CB2 agonist, RQ-00202730, for the treatment of irritable bowel syndrome. Bioorg Med Chem Lett 2015;25:236–40.
    1. Jimenez-Vargas NN, Pattison LA, Zhao P, Lieu T, Latorre R, Jensen DD, Castro J, Aurelio L, Le GT, Flynn B, Herenbrink CK, Yeatman HR, Edgington-Mitchell L, Porter CJH, Halls ML, Canals M, Veldhuis NA, Poole DP, McLean P, Hicks GA, Scheff N, Chen E, Bhattacharya A, Schmidt BL, Brierley SM, Vanner SJ, Bunnett NW. Protease-activated receptor-2 in endosomes signals persistent pain of irritable bowel syndrome. Proc Natl Acad Sci U S A 2018;115:E7438–47.
    1. Kikuchi A, Ohashi K, Sugie Y, Sugimoto H, Omura H. Pharmacological evaluation of a novel cannabinoid 2 (CB2) ligand, PF-03550096, in vitro and in vivo by using a rat model of visceral hypersensitivity. J Pharmacol Sci 2008;106:219–24.
    1. Kimball ES, Schneider CR, Wallace NH, Hornby PJ. Agonists of cannabinoid receptor 1 and 2 inhibit experimental colitis induced by oil of mustard and by dextran sulfate sodium. Am J Physiol Gastrointest Liver Physiol 2006;291:G364–71.
    1. Krishna Kumar K, Shalev-Benami M, Robertson MJ, Hu H, Banister SD, Hollingsworth SA, Latorraca NR, Kato HE, Hilger D, Maeda S, Weis WI, Farrens DL, Dror RO, Malhotra SV, Kobilka BK, Skiniotis G. Structure of a signaling cannabinoid receptor 1-G protein complex. Cell 2019;176:448–58.e412.
    1. Leinwand KL, Jones AA, Huang RH, Jedlicka P, Kao DJ, de Zoeten EF, Ghosh S, Moaddel R, Wehkamp J, Ostaff MJ, Bader J, Aherne CM, Collins CB. Cannabinoid receptor-2 ameliorates inflammation in murine model of Crohn's Disease. J Crohns Colitis 2017;11:1369–80.
    1. Li X, Hua T, Vemuri K, Ho JH, Wu Y, Wu L, Popov P, Benchama O, Zvonok N, Locke K, Qu L, Han GW, Iyer MR, Cinar R, Coffey NJ, Wang J, Wu M, Katritch V, Zhao S, Kunos G, Bohn LM, Makriyannis A, Stevens RC, Liu ZJ. Crystal structure of the human cannabinoid receptor CB2. Cell 2019;176:459–67.e413.
    1. Liu QR, Pan CH, Hishimoto A, Li CY, Xi ZX, Llorente-Berzal A, Viveros MP, Ishiguro H, Arinami T, Onaivi ES, Uhl GR. Species differences in cannabinoid receptor 2 (CNR2 gene): identification of novel human and rodent CB2 isoforms, differential tissue expression and regulation by cannabinoid receptor ligands. Genes Brain Behav 2009;8:519–30.
    1. Mearin F, Lacy BE, Chang L, Chey WD, Lembo AJ, Simren M, Spiller R. Bowel disorders. Gastroenterology 2016;S0016-5085:00222–5.
    1. Merriam FV, Wang ZY, Guerios SD, Bjorling DE. Cannabinoid receptor 2 is increased in acutely and chronically inflamed bladder of rats. Neurosci Lett 2008;445:130–4.
    1. Moayyedi P, Mearin F, Azpiroz F, Andresen V, Barbara G, Corsetti M, Emmanuel A, Hungin APS, Layer P, Stanghellini V, Whorwell P, Zerbib F, Tack J. Irritable bowel syndrome diagnosis and management: a simplified algorithm for clinical practice. United Eur Gastroenterol J 2017;5:773–88.
    1. Moreira FA, Grieb M, Lutz B. Central side-effects of therapies based on CB1 cannabinoid receptor agonists and antagonists: focus on anxiety and depression. Best Pract Res Clin Endocrinol Metab 2009;23:133–44.
    1. Naguib M, Diaz P, Xu JJ, Astruc-Diaz F, Craig S, Vivas-Mejia P, Brown DL. MDA7: a novel selective agonist for CB2 receptors that prevents allodynia in rat neuropathic pain models. Br J Pharmacol 2008;155:1104–16.
    1. Ness TJ, Gebhart GF. Colorectal distension as a noxious visceral stimulus: physiologic and pharmacologic characterization of pseudaffective reflexes in the rat. Brain Res 1988;450:153–69.
    1. Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, Panaccione R, Ghosh S, Wu JCY, Chan FKL, Sung JJY, Kaplan GG. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 2018;390:2769–78.
    1. Osteen JD, Herzig V, Gilchrist J, Emrick JJ, Zhang C, Wang X, Castro J, Garcia-Caraballo S, Grundy L, Rychkov GY, Weyer AD, Dekan Z, Undheim EA, Alewood P, Stucky CL, Brierley SM, Basbaum AI, Bosmans F, King GF, Julius D. Selective spider toxins reveal a role for the Nav1.1 channel in mechanical pain. Nature 2016;534:494–9.
    1. Sadeghi M, Erickson A, Castro J, Deiteren A, Harrington AM, Grundy L, Adams DJ, Brierley SM. Contribution of membrane receptor signalling to chronic visceral pain. Int J Biochem Cell Biol 2018;98:10–23.
    1. Salvatierra J, Castro J, Erickson A, Li Q, Braz J, Gilchrist J, Grundy L, Rychkov GY, Deiteren A, Rais R, King GF, Slusher BS, Basbaum A, Pasricha PJ, Brierley SM, Bosmans F. NaV1.1 inhibition can reduce visceral hypersensitivity. JCI Insight 2018;3:e121000.
    1. Sanson M, Bueno L, Fioramonti J. Involvement of cannabinoid receptors in inflammatory hypersensitivity to colonic distension in rats. Neurogastroenterol Motil 2006;18:949–56.
    1. Sharkey KA, Wiley JW. The role of the endocannabinoid system in the brain-gut axis. Gastroenterology 2016;151:252–66.
    1. Singh UP, Singh NP, Singh B, Price RL, Nagarkatti M, Nagarkatti PS. Cannabinoid receptor-2 (CB2) agonist ameliorates colitis in IL-10(-/-) mice by attenuating the activation of T cells and promoting their apoptosis. Toxicol Appl Pharmacol 2012;258:256–67.
    1. Soethoudt M, Grether U, Fingerle J, Grim TW, Fezza F, de Petrocellis L, Ullmer C, Rothenhausler B, Perret C, van Gils N, Finlay D, MacDonald C, Chicca A, Gens MD, Stuart J, de Vries H, Mastrangelo N, Xia L, Alachouzos G, Baggelaar MP, Martella A, Mock ED, Deng H, Heitman LH, Connor M, Di Marzo V, Gertsch J, Lichtman AH, Maccarrone M, Pacher P, Glass M, van der Stelt M. Cannabinoid CB2 receptor ligand profiling reveals biased signalling and off-target activity. Nat Commun 2017;8:13958.
    1. Spiegel BM, Bolus R, Harris LA, Lucak S, Chey WD, Sayuk G, Esrailian E, Lembo A, Karsan H, Tillisch K, Talley J, Chang L. Characterizing abdominal pain in IBS: guidance for study inclusion criteria, outcome measurement and clinical practice. Aliment Pharmacol Ther 2010;32:1192–202.
    1. Storr MA, Keenan CM, Zhang H, Patel KD, Makriyannis A, Sharkey KA. Activation of the cannabinoid 2 receptor (CB2) protects against experimental colitis. Inflamm Bowel Dis 2009;15:1678–85.
    1. Turcotte C, Blanchet MR, Laviolette M, Flamand N. The CB(2) receptor and its role as a regulator of inflammation. Cell Mol Life Sci 2016;73:4449–70.
    1. Weinland SR, Morris CB, Hu Y, Leserman J, Bangdiwala SI, Drossman DA. Characterization of episodes of irritable bowel syndrome using ecological momentary assessment. Am J Gastroenterol 2011;106:1813–20.
    1. Wright K, Rooney N, Feeney M, Tate J, Robertson D, Welham M, Ward S. Differential expression of cannabinoid receptors in the human colon: cannabinoids promote epithelial wound healing. Gastroenterology 2005;129:437–53.
    1. Yacyshyn BR, Hanauer S, Klassen P, English BA, Stauber K, Barish CF, Gilder K, Turner S, Higgins PDR. Safety, pharmacokinetics, and efficacy of olorinab, a peripherally acting, highly selective, full agonist of the cannabinoid receptor 2, in a phase 2a study of patients with chronic abdominal pain associated with Crohn's disease. Crohn's Colitis 2020;360:3.
    1. Yao BB, Mukherjee S, Fan Y, Garrison TR, Daza AV, Grayson GK, Hooker BA, Dart MJ, Sullivan JP, Meyer MD. In vitro pharmacological characterization of AM1241: a protean agonist at the cannabinoid CB2 receptor? Br J Pharmacol 2006;149:145–54.
    1. Zeitz J, Ak M, Muller-Mottet S, Scharl S, Biedermann L, Fournier N, Frei P, Pittet V, Scharl M, Fried M, Rogler G, Vavricka S. Pain in IBD patients: very frequent and frequently insufficiently taken into account. PLoS One 2016;11:e0156666.
    1. Zhang HY, Bi GH, Li X, Li J, Qu H, Zhang SJ, Li CY, Onaivi ES, Gardner EL, Xi ZX, Liu QR. Species differences in cannabinoid receptor 2 and receptor responses to cocaine self-administration in mice and rats. Neuropsychopharmacology 2015;40:1037–51.

Source: PubMed

3
Se inscrever