Paradoxical Patterns of Sinusoidal Obstruction Syndrome-Like Liver Injury in Aged Female CD-1 Mice Triggered by Cannabidiol-Rich Cannabis Extract and Acetaminophen Co-Administration

Laura E Ewing, Mitchell R McGill, Eric U Yee, Charles M Quick, Charles M Skinner, Stefanie Kennon-McGill, Melissa Clemens, Joel H Vazquez, Sandra S McCullough, D Keith Williams, Kristy R Kutanzi, Larry A Walker, Mahmoud A ElSohly, Laura P James, Bill J Gurley, Igor Koturbash, Laura E Ewing, Mitchell R McGill, Eric U Yee, Charles M Quick, Charles M Skinner, Stefanie Kennon-McGill, Melissa Clemens, Joel H Vazquez, Sandra S McCullough, D Keith Williams, Kristy R Kutanzi, Larry A Walker, Mahmoud A ElSohly, Laura P James, Bill J Gurley, Igor Koturbash

Abstract

The goal of this study was to investigate the potential for a cannabidiol-rich cannabis extract (CRCE) to interact with the most common over-the-counter drug and the major known cause of drug-induced liver injury-acetaminophen (APAP)-in aged female CD-1 mice. Gavaging mice with 116 mg/kg of cannabidiol (CBD) [mouse equivalent dose (MED) of 10 mg/kg of CBD] in CRCE delivered with sesame oil for three consecutive days followed by intraperitoneally (i.p.) acetaminophen (APAP) administration (400 mg/kg) on day 4 resulted in overt toxicity with 37.5% mortality. No mortality was observed in mice treated with 290 mg/kg of CBD+APAP (MED of 25 mg/kg of CBD) or APAP alone. Following CRCE/APAP co-administration, microscopic examination revealed a sinusoidal obstruction syndrome-like liver injury-the severity of which correlated with the degree of alterations in physiological and clinical biochemistry end points. Mechanistically, glutathione depletion and oxidative stress were observed between the APAP-only and co-administration groups, but co-administration resulted in much greater activation of c-Jun N-terminal kinase (JNK). Strikingly, these effects were not observed in mice gavaged with 290 mg/kg CBD in CRCE followed by APAP administration. These findings highlight the potential for CBD/drug interactions, and reveal an interesting paradoxical effect of CBD/APAP-induced hepatotoxicity.

Keywords: acetaminophen; cannabidiol; liver injury; natural products; phytochemical; sinusoidal obstruction syndrome.

Conflict of interest statement

Quick serves as a scientific consultant for Allergan. James is a co-owner of Acetaminophen Toxicity Diagnostics, LLC. No conflicts of interest are declared by the other authors.

Figures

Figure 1
Figure 1
Co-administration of cannabidiol-rich cannabis extract (CRCE) and APAP results in sinusoid obstructive syndrome-like histomorphological alterations in the livers of aged female CD-1 mice. H&E stained liver sections from (A) vehicle-gavaged mice (sesame oil), (B) 116 mg/kg CBD, (C) 290 mg/kg CBD, (D) APAP (400 mg/kg), (E) 116 mg/kg CBD+APAP, (F) 290 mg/kg CBD+APAP. Black brackets indicate areas of clear cell changes (D), yellow brackets (E) and arrows (F) indicate areas of sinusoidal dilation with vascular congestion, and blue arrows (E) indicate areas of hepatic plate atrophy. CRCE—cannabidiol-rich cannabis extract; APAP—acetaminophen. Magnification: X10.
Figure 2
Figure 2
Effects of CRCE/APAP co-administration on commonly assessed clinical chemistry parameters for liver injury. Blood was collected at the time of sacrifice, and the serum was subsequently separated and removed for analysis. Veh (n = 7); Veh + APAP (n = 7); 116 mg/kg CBD (n = 7); 116 mg/kg CBD+APAP (n = 5); 290 mg/kg CBD (n = 7); 290 mg/kg CBD + APAP (n = 6). Data are presented as mean ± SEM (* p < 0.05). ALT: alanine aminotransferase; AST: aspartate aminotransferase; ALP: alkaline phosphatase; GGT: gamma-glutamyl transferase.
Figure 3
Figure 3
Effects of CRCE/APAP co-administration on intrahepatic expression of cytochrome P450s and UDP-glucuronosyltransferases. Livers were collected 5 h after APAP administration and gene expression was measured using the quantitative real-time (qRT) PCR. Veh (n = 7); Veh + APAP (n = 7); 116 mg/kg CBD (n = 7); 116 mg/kg CBD + APAP (n = 7); 290 mg/kg CBD (n = 8); 290 mg/kg CBD + APAP (n = 8). Data presented as mean ± SEM fold changed from vehicle (* p < 0.05; ** p < 0.01; *** p < 0.001; and **** p < 0.0001). The following genes were changed significantly with either CBD or APAP, but the two-way ANOVA did not yield any significant comparisons: Cyp2d22 (APAP, p = 0.0016), Ugt1a1 (CBD, p = 0.0186; APAP, p = 0.0255), Ugt1a6 (CBD, p = 0.0222), and Ugt1a9 (APAP, p = 0.0026).
Figure 4
Figure 4
Mechanisms of CRCE/APAP-induced liver injury. (A) APAP-Cys protein adducts; levels of (B) GSH, and (C) GSSG/GSH; (D) mRNA levels of Gclm as measured by quantitative real-time (qRT) PCR; E) protein levels of Jnk and pJNK as measured by Western blot; and F) representative blot. Veh (n = 7); Veh + APAP (n = 7); 116 mg/kg CBD (n = 7); 116 mg/kg CBD + APAP (n = 7); 290 mg/kg CBD (n = 8); 290 mg/kg CBD + APAP (n = 8). Data presented as mean ± SEM fold changed from vehicle (* p < 0.05; ** p < 0.01; and **** p < 0.0001). GSSG levels were significantly affected by APAP (p = 0.0151), but there are no significant comparisons.

References

    1. Devinsky O., Cross J.H., Wright S. Trial of Cannabidiol for Drug-Resistant Seizures in the Dravet Syndrome. N. Engl. J. Med. 2017;377:699–700. doi: 10.1056/NEJMoa1611618.
    1. Thiele E.A., Marsh E.D., French J.A., Mazurkiewicz-Beldzinska M., Benbadis S.R., Joshi C., Lyons P.D., Taylor A., Roberts C., Sommerville K., et al. Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2018;391:1085–1096. doi: 10.1016/S0140-6736(18)30136-3.
    1. Bonini S.A., Premoli M., Tambaro S., Kumar A., Maccarinelli G., Memo M., Mastinu A. Cannabis sativa: A comprehensive ethnopharmacological review of a medicinal plant with a long history. J. Ethnopharmacol. 2018;227:300–315. doi: 10.1016/j.jep.2018.09.004.
    1. Premoli M., Aria F., Bonini S.A., Maccarinelli G., Gianoncelli A., Pina S.D., Tambaro S., Memo M., Mastinu A. Cannabidiol: Recent advances and new insights for neuropsychiatric disorders treatment. Life Sci. 2019;224:120–127. doi: 10.1016/j.lfs.2019.03.053.
    1. Rosenkrantz H., Fleischman R.W., Grant R.J. Toxicity of short-term administration of cannabinoids to rhesus monkeys. Toxicol. Appl. Pharmacol. 1981;58:118–131. doi: 10.1016/0041-008X(81)90122-8.
    1. Gamble L.J., Boesch J.M., Frye C.W., Schwark W.S., Mann S., Wolfe L., Brown H., Berthelsen E.S., Wakshlag J.J. Pharmacokinetics, Safety, and Clinical Efficacy of Cannabidiol Treatment in Osteoarthritic Dogs. Front. Vet. Sci. 2018;5:165. doi: 10.3389/fvets.2018.00165.
    1. Ewing L.E., Skinner C.M., Quick C.M., Kennon-McGill S., McGill M.R., Walker L.A., ElSohly M.A., Gurley B.J., Koturbash I. Hepatotoxicity of a Cannabidiol-Rich Cannabis Extract in the Mouse Model. Molecules. 2019;24:1694. doi: 10.3390/molecules24091694.
    1. Devinsky O., Patel A.D., Cross J.H., Villanueva V., Wirrell E.C., Privitera M., Greenwood S.M., Roberts C., Checketts D., VanLandingham K.E., et al. Effect of Cannabidiol on Drop Seizures in the Lennox-Gastaut Syndrome. N. Engl. J. Med. 2018;378:1888–1897. doi: 10.1056/NEJMoa1714631.
    1. Mazur A., Lichti C.F., Prather P.L., Zielinska A.K., Bratton S.M., Gallus-Zawada A., Finel M., Miller G.P., Radominska-Pandya A., Moran J.H. Characterization of human hepatic and extrahepatic UDP-glucuronosyltransferase enzymes involved in the metabolism of classic cannabinoids. Drug Metab. Dispos. 2009;37:1496–1504. doi: 10.1124/dmd.109.026898.
    1. Stout S.M., Cimino N.M. Exogenous cannabinoids as substrates, inhibitors, and inducers of human drug metabolizing enzymes: A systematic review. Drug Metab. Rev. 2014;46:86–95. doi: 10.3109/03602532.2013.849268.
    1. Zendulka O., Dovrtelova G., Noskova K., Turjap M., Sulcova A., Hanus L., Jurica J. Cannabinoids and Cytochrome P450 Interactions. Curr. Drug Metab. 2016;17:206–226. doi: 10.2174/1389200217666151210142051.
    1. Suh J.H., Shenvi S.V., Dixon B.M., Liu H., Jaiswal A.K., Liu R.M., Hagen T.M. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proc. Natl. Acad. Sci. USA. 2004;101:3381–3386. doi: 10.1073/pnas.0400282101.
    1. Klotz U. Pharmacokinetics and drug metabolism in the elderly. Drug Metab. Rev. 2009;41:67–76. doi: 10.1080/03602530902722679.
    1. Kaufman D.W., Kelly J.P., Rosenberg L., Anderson T.E., Mitchell A.A. Recent patterns of medication use in the ambulatory adult population of the United States: The Slone survey. JAMA. 2002;287:337–344. doi: 10.1001/jama.287.3.337.
    1. Miousse I.R., Skinner C.M., Lin H., Ewing L.E., Kosanke S.D., Williams D.K., Avula B., Khan I.A., ElSohly M.A., Gurley B.J., et al. Safety assessment of the dietary supplement OxyELITE Pro (New Formula) in inbred and outbred mouse strains. Food Chem. Toxicol. 2017;109:194–209. doi: 10.1016/j.fct.2017.08.025.
    1. Skinner C.M., Miousse I.R., Ewing L.E., Sridharan V., Cao M., Lin H., Williams D.K., Avula B., Haider S., Chittiboyina A.G., et al. Impact of obesity on the toxicity of a multi-ingredient dietary supplement, OxyELITE Pro (New Formula), using the novel NZO/HILtJ obese mouse model: Physiological and mechanistic assessments. Food Chem. Toxicol. 2018;122:21–32. doi: 10.1016/j.fct.2018.09.067.
    1. Zhu J., Seo J.E., Wang S., Ashby K., Ballard R., Yu D., Ning B., Agarwal R., Borlak J., Tong W., et al. The Development of a Database for Herbal and Dietary Supplement Induced Liver Toxicity. Int. J. Mol. Sci. 2018;19:2955. doi: 10.3390/ijms19102955.
    1. Mitchell J.R., Jollow D.J., Potter W.Z., Davis D.C., Gillette J.R., Brodie B.B. Acetaminophen-induced hepatic necrosis. I. Role of drug metabolism. J. Pharmacol. Exp. Ther. 1973;187:185–194.
    1. James L.P., McCullough S.S., Lamps L.W., Hinson J.A. Effect of N-acetylcysteine on acetaminophen toxicity in mice: Relationship to reactive nitrogen and cytokine formation. Toxicol. Sci. 2003;75:458–467. doi: 10.1093/toxsci/kfg181.
    1. Fan C.Q., Crawford J.M. Sinusoidal obstruction syndrome (hepatic veno-occlusive disease) J. Clin. Exp. Hepatol. 2014;4:332–346. doi: 10.1016/j.jceh.2014.10.002.
    1. Devinsky O., Nabbout R., Miller I., Laux L., Zolnowska M., Wright S., Roberts C. Long-term cannabidiol treatment in patients with Dravet syndrome: An open-label extension trial. Epilepsia. 2019;60:294–302. doi: 10.1111/epi.14628.
    1. Wojcikowski K., Gobe G. Animal studies on medicinal herbs: Predictability, dose conversion and potential value. Phytother. Res. 2014;28:22–27. doi: 10.1002/ptr.4966.
    1. Muldrew K.L., James L.P., Coop L., McCullough S.S., Hendrickson H.P., Hinson J.A., Mayeux P.R. Determination of acetaminophen-protein adducts in mouse liver and serum and human serum after hepatotoxic doses of acetaminophen using high-performance liquid chromatography with electrochemical detection. Drug Metab. Dispos. 2002;30:446–451. doi: 10.1124/dmd.30.4.446.
    1. McGill M.R., Lebofsky M., Norris H.R., Slawson M.H., Bajt M.L., Xie Y., Williams C.D., Wilkins D.G., Rollins D.E., Jaeschke H. Plasma and liver acetaminophen-protein adduct levels in mice after acetaminophen treatment: Dose-response, mechanisms, and clinical implications. Toxicol. Appl. Pharmacol. 2013;269:240–249. doi: 10.1016/j.taap.2013.03.026.
    1. McGill M.R., Jaeschke H. A direct comparison of methods used to measure oxidized glutathione in biological samples: 2-vinylpyridine and N-ethylmaleimide. Toxicol. Mech. Methods. 2015;25:589–595. doi: 10.3109/15376516.2015.1094844.
    1. McGill M.R., Du K., Xie Y., Bajt M.L., Ding W.X., Jaeschke H. The role of the c-Jun N-terminal kinases 1/2 and receptor-interacting protein kinase 3 in furosemide-induced liver injury. Xenobiotica. 2015;45:442–449. doi: 10.3109/00498254.2014.986250.

Source: PubMed

3
Abonnere