Skip to main content
Log in

Pharmacokinetic and Pharmacodynamic Properties of Indole-3-carbinol in Experimental Focal Ischemic Injury

  • Original Research Article
  • Published:
European Journal of Drug Metabolism and Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objectives

Indole-3-carbinol (I3C) is reported to have neuroprotective properties in an animal model of ischemic stroke. However, the pharmacokinetics of I3C in stroke animals are unknown. Furthermore, the most effective method of I3C delivery for the treatment of stroke has yet to be determined. Therefore, the objective of this study was to evaluate pharmacokinetics and pharmacodynamics of I3C to discover the most effective delivery route for protecting the brain from ischemic injury.

Methods

With oral and intravenous administration, the pharmacokinetics and pharmacodynamics of I3C in sham and middle cerebral artery occluded (MCAO) rats were investigated.

Results

I3C administration in sham and MCAO rats did not alter the pharmacokinetic parameters such as maximum plasma concentration (Cmax), time to reach Cmax, half-life, area under the curve, mean residential time, volume of distribution, clearance, bioavailability, and tissue distribution. A higher amount of diindolylmethane (DIM) was observed with oral administration of I3C compared to intravenous administration in the plasma (5 fold), brain (4 fold), and cerebrospinal fluid (CSF) (2–3 fold). Orally delivered I3C significantly reduced neurological deficits, brain infarction (20%), blood-brain barrier leakage (15 μg/g), and brain water content (75%) in MCAO rats compared to intravenous administration of I3C.

Conclusions

I3C pharmacokinetic parameters were similar in sham and MCAO rats, but I3C and DIM penetration in the brain and CSF was significantly higher in MCAO rats than in sham animals, and I3C oral intake significantly reduced MCAO-induced neurological impairments. Consequently, compared to intravenous treatment, I3C oral delivery is more effective in treating ischemic stroke.

Graphical abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Aggarwal BB, Ichikawa H. Molecular targets and anticancer potential of indole-3-carbinol and its derivatives. Cell Cycle. 2005;4(9):1201–15.

    Article  CAS  Google Scholar 

  2. Exon J, et al. Effects of indole-3-carbinol on immune responses, aberrant crypt foci, and colonic crypt cell proliferation in rats. J Toxicol Environ Health Part A. 2001;62(7):561–73.

    Article  CAS  Google Scholar 

  3. El-Naga RN, Mahran YF. Indole-3-carbinol protects against cisplatin-induced acute nephrotoxicity: role of calcitonin gene-related peptide and insulin-like growth factor-1. Sci Rep. 2016;6:29857.

    Article  CAS  Google Scholar 

  4. Ramakrishna, K. and S. Krishnamurthy, Indole-3-carbinol ameliorated the isoproterenol-induced myocardial infarction via multimodal mechanisms in Wistar rats. Nat Prod Res. 2022:36: 1–6.

  5. Garikapaty V, et al. Anti-carcinogenic and anti-metastatic properties of indole-3-carbinol in prostate cancer. Oncol Rep. 2005;13(1):89–93.

    CAS  PubMed  Google Scholar 

  6. El-Naga RN, Ahmed HI, Al Haleem ENA. Effects of indole-3-carbinol on clonidine-induced neurotoxicity in rats: Impact on oxidative stress, inflammation, apoptosis and monoamine levels. Neurotoxicology. 2014;44:48–57.

    Article  CAS  Google Scholar 

  7. Ping J, et al. Therapeutic effect of indole-3-carbinol on pig serum-induced hepatic fibrosis in rats. Yao Xue Xue Bao Acta Pharm Sin. 2011;46(8):915–21.

    CAS  Google Scholar 

  8. Souli E, et al. Indole-3-carbinol (I3C) exhibits inhibitory and preventive effects on prostate tumors in mice. Food Chem Toxicol. 2008;46(3):863–70.

    Article  CAS  Google Scholar 

  9. Paliwal P, Chauhan G, Gautam D, Dash D, Patne SCU, Krishnamurthy S. Indole-3-carbinol improves neurobehavioral symptoms in a cerebral ischemic stroke model. Naunyn Schmiedebergs Arch Pharmacol. 2018;391(6):613–25.

    Article  CAS  Google Scholar 

  10. Bansal S, Sangha KS, Khatri P. Drug treatment of acute ischemic stroke. Am J Cardiovasc Drugs. 2013;13(1):57–69.

    Article  CAS  Google Scholar 

  11. Fisher M, et al. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke. 2009;40(6):2244–50.

    Article  Google Scholar 

  12. Park MK, Rhee Y-H, Lee H-J, Lee E-O, Kim K-H, Park M-J, Jeon B-H, Shim B-S, Jung C-H, Choi S-H. Antiplatelet and antithrombotic activity of indole-3-carbinol in vitro and in vivo. Phytother Res Int J Devoted Pharmacol Toxicol Eval Nat Prod Deriv. 2008;22(1):58–64.

    CAS  Google Scholar 

  13. De Kruif C, et al. Structure elucidation of acid reaction products of indole-3-carbinol: detection in vivo and enzyme induction in vitro. Chem Biol Interact. 1991;80(3):303–15.

    Article  Google Scholar 

  14. Shertzer HG, Senft AP. The micronutrient indole-3-carbinol: implications for disease and chemoprevention. Drug Metab Drug Interact. 2000;17(1–4):159–88.

    CAS  Google Scholar 

  15. Moussata J, Wang Z, Wang J. Development and validation of an HPLC method for the simultaneous quantification of indole-3-carbinol acetate, indole-3-carbinol, and 3, 3′-diindolylmethane in mouse plasma, liver, and kidney tissues. J Chromatogr B. 2014;958:1–9.

    Article  CAS  Google Scholar 

  16. Anderton MJ, et al. Pharmacokinetics and tissue disposition of indole-3-carbinol and its acid condensation products after oral administration to mice. Clin Cancer Res. 2004;10(15):5233–41.

    Article  CAS  Google Scholar 

  17. Anderton MJ, et al. Liquid chromatographic assay for the simultaneous determination of indole-3-carbinol and its acid condensation products in plasma. J Chromatogr B. 2003;787(2):281–91.

    Article  CAS  Google Scholar 

  18. Staub RE, et al. Fate of indole-3-carbinol in cultured human breast tumor cells. Chem Res Toxicol. 2002;15(2):101–9.

    Article  CAS  Google Scholar 

  19. Li Y, et al. Pharmacokinetic comparison of scutellarin and paeoniflorin in sham-operated and middle cerebral artery occlusion ischemia and reperfusion injury rats after intravenous administration of xin-shao formula. Molecules. 2016;21(9):1191.

    Article  Google Scholar 

  20. Ramakrishna K, Srinivasan K, Sharma SS. Chronic treatment of 4-phenylbutyric acid ameliorates cognitive impairment after focal cerebral ischemia/reperfusion injury in rats. Indian J Physiol Pharmacol. 2021;64(3):188–94.

    Article  Google Scholar 

  21. Brand M, et al. The role of mitochondrial function and cellular bioenergetics in ageing and disease. Br J Dermatol. 2013;169:1–8.

    Article  CAS  Google Scholar 

  22. Longa EZ, et al. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 1989;20(1):84–91.

    Article  CAS  Google Scholar 

  23. Belayev L, et al. Middle cerebral artery occlusion in the rat by intraluminal suture. Neurological and pathological evaluation of an improved model. Stroke. 1996;27(9):1616–22 (discussion 1623).

    Article  CAS  Google Scholar 

  24. Shah FA, et al. Melatonin protects MCAO-induced neuronal loss via NR2A mediated prosurvival pathways. Front Pharmacol. 2019;10:297.

    Article  CAS  Google Scholar 

  25. Zhang W, et al. Omega-3 polyunsaturated fatty acids mitigate blood–brain barrier disruption after hypoxic–ischemic brain injury. Neurobiol Dis. 2016;91:37–46.

    Article  Google Scholar 

  26. Maleki SN, Aboutaleb N, Souri F. Berberine confers neuroprotection in coping with focal cerebral ischemia by targeting inflammatory cytokines. J Chem Neuroanat. 2018;87:54–9.

    Article  CAS  Google Scholar 

  27. FDA, US. Guidance for industry: bioanalytical method validation (2018). Center for Drug Evaluation and Research (CDER), Silver Spring, MD and/or Center for Veterinary Medicine (CVM): Rockville, MD. 2001:1-27.

  28. Martinez MN, Papich MG, Drusano GL. Dosing regimen matters: the importance of early intervention and rapid attainment of the pharmacokinetic/pharmacodynamic target. Antimicrob Agents Chemother. 2012;56(6):2795–805.

    Article  CAS  Google Scholar 

  29. Grose KR, Bjeldanes LF. Oligomerization of indole-3-carbinol in aqueous acid. Chem Res Toxicol. 1992;5(2):188–93.

    Article  CAS  Google Scholar 

  30. Deng Y, et al. Icariside II protects against cerebral ischemia–reperfusion injury in rats via nuclear factor-κB inhibition and peroxisome proliferator-activated receptor up-regulation. Neurochem Int. 2016;96:56–61.

    Article  CAS  Google Scholar 

  31. Mukherjee S, Kumar G, Patnaik R. Withanolide a penetrates brain via intra-nasal administration and exerts neuroprotection in cerebral ischemia reperfusion injury in mice. Xenobiotica. 2020;50(8):957–66.

    Article  CAS  Google Scholar 

  32. Jin G, et al. Protecting against cerebrovascular injury: contributions of 12/15-lipoxygenase to edema formation after transient focal ischemia. Stroke. 2008;39(9):2538–43.

    Article  CAS  Google Scholar 

  33. Singh AA, et al. Biomedical application of Indole-3-carbinol: a mini-review. Phytochem Lett. 2021;41:49–54.

    Article  CAS  Google Scholar 

  34. Stresser DM, et al. Substrate-dependent modulation of CYP3A4 catalytic activity: analysis of 27 test compounds with four fluorometric substrates. Drug Metab Dispos. 2000;28(12):1440–8.

    CAS  PubMed  Google Scholar 

  35. Ahmad A, Sakr WA, Rahman K. Mechanisms and therapeutic implications of cell death induction by indole compounds. Cancers. 2011;3(3):2955–74.

    Article  CAS  Google Scholar 

  36. Ramakrishna K, Singh N, Krishnamurthy S. Diindolylmethane ameliorates platelet aggregation and thrombosis: in silico, in vitro, and in vivo studies. Eur J Pharmacol. 2022;919:174812.

    Article  CAS  Google Scholar 

Download references

Acknowledgments

Kakarla Ramakrishna thanks IIT (BHU), Varanasi, and MHRD, India, for providing the teaching assistantship for carrying out the experimental work. We also thank Pankaj Paliwal and Akanksha Mishra, research scholars of Pharmaceutical Engineering and Technology, IIT (BHU), Varanasi, India, for their help in the experiments.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sairam Krishnamurthy.

Ethics declarations

Funding

No source of funding.

Conflict of interest

The authors report no conflict of interest.

Ethics approval

The experimental protocol was approved by IMS, BHU, Varanasi (protocol no. Dean/2016/CAEC/31). The approved protocol strictly adhered to the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA) and follows the ARRIVE guidelines.

Availability of data and material

All data generated or analyzed during this study are included in this manuscript and also available in a supplementary file.

Code availability

Not applicable.

Author contributions

KR designed and performed the experiment, analyzed the data, and wrote the manuscript. SKJ helped perform experiments and edited the manuscript. SK designed and analyzed the data and edited the manuscript.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 133 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ramakrishna, K., Jain, S.K. & Krishnamurthy, S. Pharmacokinetic and Pharmacodynamic Properties of Indole-3-carbinol in Experimental Focal Ischemic Injury. Eur J Drug Metab Pharmacokinet 47, 593–605 (2022). https://doi.org/10.1007/s13318-022-00771-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13318-022-00771-y

Navigation