Journal Home > Volume 1 , Issue 1
Background

Phyto-constituents are widely recognized for hepatoprotective effects. Ascorbic acid is extensively studied for the radical scavenging effect. The objective of the present study was to evaluate the hepatoprotective effect of ascorbic acid against isoniazid and rifampicin-induced liver damage in rats.

Methods

Wistar rats were used in the present study. Hepato-cellular damage was produced by the administration of isoniazid (100 mg/kg) and rifampicin (100 mg/kg) for 21 days. Ascorbic acid was administered in the dose of 50, 100, and 200 mg/kg body weight. At the end of the study, blood was collected and biochemical studies were performed to determine antioxidant status.

Results

Ascorbic acid administration (50, 100, and 200 mg/kg body weight) caused restoration of serum glutamic oxaloacetic transaminase, serum glutamic-pyruvic transaminase, and serum alkaline phosphatase. The level of superoxide dismutase and catalase were also restored due to the administration of ascorbic acid. There was a decrement in the expression of tumor necrosis factor-α, interleukin-1β, interleukin-6, malondialdehyde and nitric oxide.

Conclusion

The outcome of the study established ascorbic acid as a notable hepatoprotective effect. The radical scavenging and cytokine suppression effect is the possible mechanism associated with the hepatoprotective effect of ascorbic acid.


menu
Abstract
Full text
Outline
About this article

Ascorbic acid ameliorates isoniazid-rifampicin-induced hepatocellular damage in rats

Show Author's information Shubham PatelAman ChaturvediNazneen DubeyAbhishek ShrivastavaAditya Ganeshpurkar( )
Shri Ram Institute of Technology-Pharmacy, Jabalpur, India

Abstract

Background

Phyto-constituents are widely recognized for hepatoprotective effects. Ascorbic acid is extensively studied for the radical scavenging effect. The objective of the present study was to evaluate the hepatoprotective effect of ascorbic acid against isoniazid and rifampicin-induced liver damage in rats.

Methods

Wistar rats were used in the present study. Hepato-cellular damage was produced by the administration of isoniazid (100 mg/kg) and rifampicin (100 mg/kg) for 21 days. Ascorbic acid was administered in the dose of 50, 100, and 200 mg/kg body weight. At the end of the study, blood was collected and biochemical studies were performed to determine antioxidant status.

Results

Ascorbic acid administration (50, 100, and 200 mg/kg body weight) caused restoration of serum glutamic oxaloacetic transaminase, serum glutamic-pyruvic transaminase, and serum alkaline phosphatase. The level of superoxide dismutase and catalase were also restored due to the administration of ascorbic acid. There was a decrement in the expression of tumor necrosis factor-α, interleukin-1β, interleukin-6, malondialdehyde and nitric oxide.

Conclusion

The outcome of the study established ascorbic acid as a notable hepatoprotective effect. The radical scavenging and cytokine suppression effect is the possible mechanism associated with the hepatoprotective effect of ascorbic acid.

Keywords: Cytokines, Toxicity, Liver, Antitubercular, Peroxidation, Ascorbic acid

References(35)

[1]
Younossi ZM, Koenig AB, Abdelatif D, et al. Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016.https://doi.org/10.1002/hep.28431
DOI
[2]

Li S, Tan HY, Wang N, et al. The role of oxidative stress and antioxidants in liver diseases. Int J Mol Sci 2015.

[3]

Bessone F, Dirchwolf M, Rodil MA, et al. Review article: drug-induced liver injury in the context of nonalcoholic fatty liver disease – a physiopathological and clinical integrated view. Aliment Pharmacol Therapeut 2018.

[4]

Zumla A, Chakaya J, Centis R, et al. Tuberculosis treatment and management-an update on treatment regimens, trials, new drugs, and adjunct therapies. Lancet Respir Med 2015.

[5]

Biswas SSA, Bishu D, Dhali GK, et al. Anti-TB drugs, isoniazid and rifampicin produce hepatic fibrosis through a oxidative stress-dependent mechanism. J Clin Exp Hepatol 2015;5: S80.

[6]

Karunaweera N, Raju R, Gyengesi E, et al. Plant polyphenols as inhibitors of NF-κB induced cytokine production—a potential anti-inflammatory treatment for Alzheimer's disease? Front Mol Neurosci 2015;8.

[7]

Rafique S, Hassan SM, Mughal SS, et al. Biological attributes of lemon: a review. J Addict Med Ther Sci 2020;6(1): 30–4.

[8]

Al-Niaimi F, Chiang NYZ. Topical vitamin C and the skin: mechanisms of action and clinical applications. J Clin Aesthet Dermatol 2017;10(7): 14.

[9]

Naidu KA. Vitamin C in human health and disease is still a mystery? An overview. Nutr J 2003;2(1): 7.

[10]

Su M, Liang X, Xu X, et al. Hepatoprotective benefits of vitamin C against perfluorooctane sulfonate-induced liver damage in mice through suppressing inflammatory reaction and ER stress. Environ Toxicol Pharmacol 2019;65: 60–5.

[11]

Abdulrazzaq AM, Badr M, Gammoh O, et al. Hepatoprotective actions of ascorbic acid, alpha lipoic acid and silymarin or their combination against acetaminopheninduced hepatotoxicity in rats. Medicina 2019;55(5): 181.

[12]

Antunes LMG, Darin JDC, Bianchi MDELP. Protective effects of vitamin C against cisplatin-induced nephrotoxicity and lipid peroxidation in adult rats: a dosedependent study. Pharmacol Res 2000;41(4): 405–11.

[13]

Masayasu M, Hiroshi Y. A simplified assay method of superoxide dismutase activity for clinical use. Clin Chim Acta 1979;92(3): 337–42. https://doi.org/10.1016/0009-8981(79)90211-0.

[14]

Sinha AK. Colorimetric assay of catalase. Anal Biochem 1972;47(2): 389–94. https://doi.org/10.1016/0003-2697(72)90132-7.

[15]

Ohkawa H, Ohishi N, Yagi K. Reaction of linoleic acid hydroperoxide with thiobarbituric acid. JLR (J Lipid Res) 1978;19(8): 1053–7.

[16]

Moshage H, Kok B, Huizenga JR, et al. Nitrite and nitrate determinations in plasma: a critical evaluation. Clin Chem 1995;41(6): 892–6.

[17]

Meng X, Maggs JL, Usui T, et al. Auto-oxidation of isoniazid leads to isonicotinic-lysine adducts on human serum albumin. Chem Res Toxicol 2015; 28(1): 51–8.

[18]

Metushi IG, Sanders C, Group ALS, et al. Detection of anti-isoniazid and anti–cytochrome P450 antibodies in patients with isoniazid-induced liver failure. Hepatology 2014;59(3): 1084–93.

[19]

Baskaran UL, Sabina EP. Clinical and experimental research in antituberculosis drug-induced hepatotoxicity: a review. J Integrat Med 2017;15(1): 27–36.

[20]

Zhang G, Zhu J, Zhou Y, et al. Hesperidin alleviates oxidative stress and upregulates the multidrug resistance protein 2 in isoniazid and rifampicin-induced liver injury in rats. J Biochem Mol Toxicol 2016;30(7): 342–9.

[21]

Abdel-Ghaffar O, Mahmoud ST, Said AA, et al. Hepatoprotective effect of rutin against oxidative stress of isoniazid in albino rats. Int J Pharmacol 2017;13: 516–28.

[22]

Wang C, Fan R-Q, Zhang Y-X, et al. Naringenin protects against isoniazid-and rifampicin-induced apoptosis in hepatic injury. World J Gastroenterol 2016;22(44): 9775.

[23]

Sabina EP, Peter S J, Geetha A. A comparison of hepatoprotective activity of Bacoside to Silymarin treatment against a combined Isoniazid and Rifampininduced hepatotoxicity in female Wistar rats. J Histotechnol 2019;42(3): 128–36.

[24]

Kumar R, Bhatia V, Khanal S, et al. Antituberculosis therapy–induced acute liver failure: magnitude, profile, prognosis, and predictors of outcome. Hepatology 2010; 51(5): 1665–74.

[25]

Ighodaro OM, Akinloye OA. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): their fundamental role in the entire antioxidant defence grid. Alexandria J Med 2018;54(4): 287–93.

[26]

Ahmadinejad F, Geir Møller S, Hashemzadeh-Chaleshtori M, et al. Molecular mechanisms behind free radical scavengers function against oxidative stress. Antioxidants 2017;6(3): 51.

[27]

Lopez-Castejon G, Brough D. Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev 2011. https://doi.org/10.1016/j.cytogfr.2011.10.001.

[28]

Sadatomo A, Inoue Y, Ito H, et al. Interaction of neutrophils with macrophages promotes IL-1β maturation and contributes to hepatic ischemia–reperfusion injury. J Immunol 2017;199(9): 3306–15.

[29]

Schmidt-Arras D, Rose-John S. IL-6 pathway in the liver: from physiopathology to therapy. J Hepatol 2016;64(6): 1403–15.

[30]
Boakye AN. Serum cytokine profiles as biomarker in Multi-drug resistant tuberculosis (MDR-TB). University Of Ghana; 2017.
[31]

Ramachandran A, Jaeschke H. Oxidative stress and acute hepatic injury. Current Opin Toxicol 2018;7: 17–21.

[32]

Arrigoni O, De Tullio MC. Ascorbic acid: much more than just an antioxidant. Biochim Biophys Acta Gen Subj 2002;1569(1–3): 1–9.

[33]

Sorice A, Guerriero E, Capone F, et al. Ascorbic acid: its role in immune system and chronic inflammation diseases. Mini Rev Med Chem 2014;14(5): 444–52.

[34]

Somasundaram V, Basudhar D, Bharadwaj G, et al. Molecular mechanisms of nitric oxide in cancer progression, signal transduction, and metabolism. Antioxidants Redox Signal 2019;30(8): 1124–43.

[35]

Ruart M, Chavarria L, Camprecios G, et al. Impaired endothelial autophagy promotes liver fibrosis by aggravating the oxidative stress response during acute liver injury. J Hepatol 2019;70(3): 458–69.

Publication history
Copyright
Rights and permissions

Publication history

Received: 12 January 2022
Revised: 18 February 2022
Accepted: 02 March 2022
Published: 10 March 2022
Issue date: March 2022

Copyright

© 2022 The Author(s). Published by Elsevier Ltd on behalf of Tsinghua University Press.

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/)

Return