1
|
Ahmad I, Alam W, Pirzada AS, Darwish HW, Zafar R, Daglia M, Khan H. Oxindole derivatives alleviate paracetamol-induced nephrotoxicity and hepatotoxicity: biochemical, histological, and computational expressions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03568-9. [PMID: 39545986 DOI: 10.1007/s00210-024-03568-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
Oxindole is a nature-derived heteroaromatic nucleus with a history of preclinical uses in various conditions. In this study, oxindole derivatives, 6-chloro-3-(3-hydroxybenzylidene) indolin-2-one (3OH) and 6-chloro-3-(4-hydroxybenzylidene) indolin-2-one (4OH) were evaluated for nephroprotective and hepatoprotective effects. Paracetamol-induced nephrotoxicity and hepatotoxicity model was used in mice. Tissue histology and serum biochemistry were carried out to further support in vivo activity. Compound 3OH reduced serum urea and creatinine levels by 51.8% and 64.6%, respectively (p < 0.0001). Excretion of creatinine by 3OH 10 mg was 52.8% as compared to silymarin. In case of urinary excretion of urea, the significant rise in excretion was observed in 4OH 15 mg (30.4%; p < 0.05) and 3OH 10 mg group (29.24%; p < 0.05). The compound 3OH exhibited restorative pattern of the renal tissues with slight inflammatory infiltrations. In case of hepatoprotective activity, 3OH reduced (59.9%; p < 0.0001) serum ALT at 5 mg even more than silymarin and all other doses of oxindole derivatives. In case of serum AST, all treatment groups produced significant (p < 0.0001) reduction except 3OH 15 mg. Computational studies supported the results as both derivatives were found to have promising interactions with enzymes at lower binding energies. Compound 3OH which possesses a hydroxyl group based on aromatic ring at meta position was the most successful drug candidate throughout this study. In a nutshell, the selected compounds elicited significant nephroprotective and hepatoprotective-like effects in mice.
Collapse
Affiliation(s)
- Imad Ahmad
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Abdul Saboor Pirzada
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Hany W Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Rehman Zafar
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Riphah International Unversity, Islamabad, 44000, Pakistan
- School of Pharmacy, Iqra Institute of Health Sciences, Islamabad, 44000, Pakistan
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, Naples, Campania, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
- Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| |
Collapse
|
2
|
Yu Y, Sun B, Ye X, Wang Y, Zhao M, Song J, Geng X, Marx U, Li B, Zhou X. Hepatotoxic assessment in a microphysiological system: Simulation of the drug absorption and toxic process after an overdosed acetaminophen on intestinal-liver-on-chip. Food Chem Toxicol 2024; 193:115016. [PMID: 39304085 DOI: 10.1016/j.fct.2024.115016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
To compensate the limitation of animal models, new models were proposed for drug safety evaluation to refine and reduce existing models. To mimic drug absorption and metabolism and predict toxicokinetic and toxic effects in an in vitro intestinal-liver microphysiological system (MPS), we constructed an intestinal-liver-on-chip and detected the acute liver injury process after an overdose of acetaminophen (APAP). Caco-2 and HT29-MTX-E12 cell lines were utilized to establish intestinal equivalents, along with HepG2, HUVEC-T1, and THP-1 induced by PMA and human hepatic stellate cell to establish liver equivalents. The APAP concentration was determined using high-performance liquid chromatography, and the toxicokinetic parameters were fitted using the non-compartmental analysis method by Phoenix. Changes in liver injury biomarkers aspartate aminotransferase and alanine aminotransferase, and liver function marker albumin indicated that the short-term culture of the two organs-on-chip model was stable for 4 days. Reactive oxygen species signaling was enhanced after APAP administration, along with decreased mitochondrial membrane potential, activated caspase-3, and enhanced p53 signaling, indicating a toxic response induced by APAP overdose. In the gut-liver MPS model, we fitted the toxicokinetic parameters and simulated the hepatotoxicity procedure following an APAP overdose, which will facilitate the organ-on-chips application in drug toxicity assays.
Collapse
Affiliation(s)
- Yue Yu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China
| | - Baiyang Sun
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China
| | - Xiao Ye
- Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China
| | - Yupeng Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China
| | - Manman Zhao
- Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China
| | - Jie Song
- Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China
| | - Xingchao Geng
- Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China
| | - Uwe Marx
- TissUse GmbH, Oudenarder Str. 16, D-13347, Berlin, Germany.
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China.
| | - Xiaobing Zhou
- Institute for Safety Evaluation, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing, 100176, China.
| |
Collapse
|
3
|
Yu C, Guo X, Cui X, Su G, Wang H. Functional Food Chemical Ingredient Strategies for Non-alcoholic Fatty Liver Disease (NAFLD) and Hepatic Fibrosis: Chemical Properties, Health Benefits, Action, and Application. Curr Nutr Rep 2024; 13:1-14. [PMID: 38172459 DOI: 10.1007/s13668-023-00514-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE OF REVIEW The liver is an important digestive gland in the body. Lifestyle and dietary habits are increasingly damaging our liver, leading to various diseases and health problems. Non-alcoholic fatty liver disease (NAFLD) has become one of the most serious liver disease problems in the world. Diet is one of the important factors in maintaining liver health. Functional foods and their components have been identified as novel sources of potential preventive agents in the prevention and treatment of liver disease in daily life. However, the effects of functional components derived from small molecules in food on different types of liver diseases have not been systematically summarized. RECENT FINDINGS The components and related mechanisms in functional foods play a significant role in the development and progression of NAFLD and liver fibrosis. A variety of structural components are found to treat and prevent NAFLD and liver fibrosis through different mechanisms, including flavonoids, alkaloids, polyphenols, polysaccharides, unsaturated fatty acids, and peptides. On the other hand, the relevant mechanisms include oxidative stress, inflammation, and immune regulation, and a large number of literature studies have confirmed a close relationship between the mechanisms. The purpose of this article is to examine the current literature related to functional foods and functional components used for the treatment and protection against NAFLD and hepatic fibrosis, focusing on chemical properties, health benefits, mechanisms of action, and application in vitro and in vivo. The roles of different components in the biological processes of NAFLD and liver fibrosis were also discussed.
Collapse
Affiliation(s)
- Chong Yu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Xiaohe Guo
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Xiaohang Cui
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Guangyue Su
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Haifeng Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
| |
Collapse
|
4
|
Du H, Tong S, Kuang G, Gong X, Jiang N, Yang X, Liu H, Li N, Xie Y, Xiang Y, Guo J, Li Z, Yuan Y, Wu S, Wan J. Sesamin Protects against APAP-Induced Acute Liver Injury by Inhibiting Oxidative Stress and Inflammatory Response via Deactivation of HMGB1/TLR4/NF κB Signal in Mice. J Immunol Res 2023; 2023:1116841. [PMID: 37663051 PMCID: PMC10471453 DOI: 10.1155/2023/1116841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/16/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Acetaminophen (APAP) overdose would lead to liver toxicity and even acute liver failure in severe cases by triggering an inflammatory response and oxidative stress. Sesamin has been reported to possess anti-inflammatory and antioxidant actions in several animal disease models. In the present study, the effects and mechanisms of sesamin on APAP-induced acute liver injury (ALI) were explored. The results showed that pretreatment with sesamin significantly alleviated APAP-induced ALI, as indicated by decreased serum aminotransferase activities, hepatic pathological damages, and hepatic cellular apoptosis. But sesamin has no significant effects on the expression of cytochrome P450 2E1 (CYP2E1), APAP-cysteine adducts (APAP-CYS) production, and glutathione content in the liver of APAP-administered mice. Moreover, APAP-induced liver oxidative stress and inflammatory response also were remarkedly attenuated by sesamin, including reducing hepatic reactive oxygen species levels, promoting antioxidant generation, and inhibiting the expression of TNF-α and IL-1β, as well as decreasing inflammatory cell recruitment. Notably, sesamin inhibited serum high-mobility group box 1 (HMGB1) releases and blocked hepatic activation of Toll-like receptor 4 (TLR4)-interleukin 1 receptor-associated kinase 3-nuclear factor kappa B (NF-κB) signaling pathway in APAP-administered mice. These findings indicated that sesamin could mitigate APAP-induced ALI through suppression of oxidative stress and inflammatory response, which might be mediated by the deactivation of HMGB1/TLR4/NF-κB signaling in mice.
Collapse
Affiliation(s)
- Hui Du
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Shiwen Tong
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ge Kuang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Ningman Jiang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xian Yang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Hao Liu
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Nana Li
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yao Xie
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yang Xiang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jiashi Guo
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Zhenhan Li
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Yinglin Yuan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengwang Wu
- Department of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Hammad AM, Shawaqfeh B, Hikmat S, Al-Qirim T, Hamadneh L, Al-Kouz S, Awad MM, Hall FS. The Role of Vitamin E in Protecting against Oxidative Stress, Inflammation, and the Neurotoxic Effects of Acute Paracetamol in Pregnant Female Rats. TOXICS 2023; 11:368. [PMID: 37112594 PMCID: PMC10141164 DOI: 10.3390/toxics11040368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/29/2023] [Accepted: 04/06/2023] [Indexed: 06/19/2023]
Abstract
Paracetamol (acetaminophen, APAP) is the most common non-prescription analgesic drug used during pregnancy. The aim of this study was to investigate the effect of vitamin E on acute APAP toxicity in pregnant rats. Toxicity in the liver, kidney, and brain (hippocampus, cerebellum, and olfactory bulb) was examined. Twenty pregnant female Wistar rats at gestational day 18 were used. Pregnant rats were divided into four groups: Control, APAP, E + APAP, and APAP + E. The Control group was treated with 0.5 mL p.o. corn oil. The APAP group received 3000 mg/kg p.o. APAP. The E + APAP group received 300 mg/kg p.o. vitamin E one hour before 3000 mg/kg APAP. The APAP + E group received 3000 mg/kg paracetamol one hour before 300 mg/kg p.o. vitamin E. Twenty-four hours after the last treatment administration, rats were euthanized and blood, brain, liver, and kidney samples were collected. Alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), creatinine levels, uric acid (UA), and superoxide dismutase (SOD) levels, as well as the relative mRNA expression of Cyp1a4, Cyp2d6, and Nat2, were determined. Acute APAP treatment upregulated ALT, AST, BUN, and creatinine levels. APAP treatment downregulated UA and SOD levels. APAP treatment upregulated the relative mRNA expression of Cyp1a4 and Cyp2d6, but downregulated Nat2 expression. Vitamin E treatment, either before or after APAP administration, attenuated the toxic effects of APAP. In conclusion, the results showed that an acute toxic APAP dose in late pregnancy can cause oxidative stress and dysregulation in Cyp isoform expression, and that vitamin E treatment attenuates these effects.
Collapse
Affiliation(s)
- Alaa M. Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Baraa Shawaqfeh
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Suhair Hikmat
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Tariq Al-Qirim
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Lama Hamadneh
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt 19117, Jordan
| | - Sameer Al-Kouz
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Mariam M. Awad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Frank S. Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43606, USA;
| |
Collapse
|
6
|
Cytoprotective remedies for ameliorating nephrotoxicity induced by renal oxidative stress. Life Sci 2023; 318:121466. [PMID: 36773693 DOI: 10.1016/j.lfs.2023.121466] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 02/11/2023]
Abstract
AIMS Nephrotoxicity is the hallmark of anti-neoplastic drug metabolism that causes oxidative stress. External chemical agents and prescription drugs release copious amounts of free radicals originating from molecular oxidation and unless sustainably scavenged, they stimulate membrane lipid peroxidation and disruption of the host antioxidant mechanisms. This review aims to provide a comprehensive collection of potential cytoprotective remedies in surmounting the most difficult aspect of cancer therapy as well as preventing renal oxidative stress by other means. MATERIALS AND METHODS Over 400 published research and review articles spanning several decades were scrutinised to obtain the relevant data which is presented in 3 categories; sources, mechanisms, and mitigation of renal oxidative stress. KEY-FINDINGS Drug and chemical-induced nephrotoxicity commonly manifests as chronic or acute kidney disease, nephritis, nephrotic syndrome, and nephrosis. Renal replacement therapy requirements and mortalities from end-stage renal disease are set to rapidly increase in the next decade for which 43 different cytoprotective compounds which have the capability to suppress experimental nephrotoxicity are described. SIGNIFICANCE The renal system performs essential homeostatic functions that play a significant role in eliminating toxicants, and its accumulation and recurrence in nephric tissues results in tubular degeneration and subsequent renal impairment. Global statistics of the latest chronic kidney disease prevalence is 13.4 % while the end-stage kidney disease requiring renal replacement therapy is 4-7 million per annum. The remedial compounds discussed herein had proven efficacy against nephrotoxicity manifested consequent to impaired antioxidant mechanisms in preclinical models produced by renal oxidative stress activators.
Collapse
|
7
|
Curative Effects of Dianthus orientalis against Paracetamol Triggered Oxidative Stress, Hepatic and Renal Injuries in Rabbit as an Experimental Model. SEPARATIONS 2023. [DOI: 10.3390/separations10030182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
The aim of the present study investigates the hepatoprotective, nephroprotective and hematopoietic and antioxidant effects of Dianthus orientalis leaves aqueous extract (DO.AQ) in rabbits intoxicated with paracetamol. Different experimental groups were formed, i.e., group N, group T, group ELD, group EMD, group EHD and group SM. The groups with leaves aqueous extract of Dianthus orientalis of 200 and 400 mg/kg body weight, i.e., group EMD and group EHD, showed remedial effects; however, a high dose extract significantly (p < 0.05) reduced the elevated serum levels of alanine transaminase ALT, aspartate transaminase AST and alkaline phosphatase ALP and renal related indices such as serum creatinine, urea and uric acid, and serum electrolytes such as Ca, Mg, P, Na and K, as well as the total count of RBC, WBC, platelets and hemoglobin Hb concentration, mean corpuscular hemoglobin MCH concentration and hematocrit HCT values. Additionally, the extract showed positive effects on the lipid profile, i.e., decreasing levels of cholesterol, triglycerides and LDL and increasing levels of HDL. The levels of thiobarbituric acid reactive substances TBARS, glutathione GSH and radical scavenging activity were also evaluated in liver and kidney homogenates. Paracetamol fed animals had high levels of thiobarbituric acid reactive substances and low levels of glutathione GSH and radical scavenging activity (RSA). Extract ingestion caused a significant increase in glutathione and radical scavenging activity RSA levels, while reducing the (TBARS) levels, showing that the extracts have antioxidant potentials. The antioxidant capacity of the Dianthus orientalis leaves aqueous extract at various dosages demonstrated an increased inhibition of DPPH, i.e., 2, 2-diphenyl-1-picrylehydrazyle free radical. The histological study of the liver and kidney supports the protective activity of Dianthus orientalis leaves aqueous extract against paracetamol intoxication with optimistic effects regarding oxidative stress condition and serum electrolytes balance.
Collapse
|
8
|
Dua TK, Palai S, Roy A, Paul P. Protective effect of probiotics against acetaminophen induced nephrotoxicity. Mol Biol Rep 2022; 49:8139-8143. [PMID: 35661049 DOI: 10.1007/s11033-022-07534-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/24/2022]
Abstract
Acetaminophen (APAP) is commonly prescribed as an antipyretic and analgesic agent in the practical field. Like every other drug(s), APAP also undergo metabolism by oxidation or conjugation by glucuronate and sulphate to form the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). Moreover, the NAPQI is detoxified by conjugation with reduced glutathione (GSH). Interestingly, APAP is also metabolized in the kidney by deacetylation reaction in the presence of N-deacetylase enzyme into another severely toxic but minor metabolite, p-aminophenol. Both NAPQI and p-aminophenol shows nephrotoxicity as well as hepatotoxicity. Hence, the long-term therapeutic dose use and unnecessary overdose of APAP are of great concern as prolonged negligence may cost the nephrotoxicity that may lead to uremia and finally to kidney failure. It has recently been investigated that probiotic supplementation inhibits the sequential events associated with APAP-induced nephrotoxicity. This review emphasizes the role of different probiotics that have already been investigated in nephrotoxicity or uremia caused by APAP overdose.
Collapse
Affiliation(s)
- Tarun Kumar Dua
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, 734013, Darjeeling, West Bengal, India.
| | - Sangita Palai
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, 734013, Darjeeling, West Bengal, India
| | - Abani Roy
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, 734013, Darjeeling, West Bengal, India
| | - Paramita Paul
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, 734013, Darjeeling, West Bengal, India
| |
Collapse
|
9
|
Wang YQ, Geng XP, Wang MW, Wang HQ, Zhang C, He X, Liang SM, Xu DX, Chen X. Vitamin D deficiency exacerbates hepatic oxidative stress and inflammation during acetaminophen-induced acute liver injury in mice. Int Immunopharmacol 2021; 97:107716. [PMID: 33951559 DOI: 10.1016/j.intimp.2021.107716] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022]
Abstract
Several experiments confirmed that vitamin D3 protected against acetaminophen (APAP)-induced acute liver injury (ALI). This research aimed to evaluate the influence of vitamin D deficiency (VDD) on APAP-induced ALI. In VDD and VDD + APAP groups, mice were fed with VDD diet. In APAP and VDD + APAP groups, mice were intraperitoneally injected with a sublethal dose of APAP (150 mg/kg). A sublethal dose of APAP caused a slight elevation of ALT and AST. Interestingly, APAP-induced elevation of ALT and AST was aggravated in VDD-fed mice. APAP-induced hepatic necrosis was exacerbated in VDD-fed mice. In addition, APAP-induced hepatocyte death, measured using TUNEL assay, was exacerbated in VDD-fed mice. Additional experiment showed that APAP-induced hepatic GSH depletion and lipid peroxidation were exacerbated in VDD-fed mice. Moreover, APAP-induced upregulation of antioxidant genes, such as hepatic heme oxygenase-1 (Ho-1), glutathione peroxidase (Gshpx), superoxide dismutase 1 (Sod1) and catalase enzymes (Cat), was aggravated in VDD-fed mice. Although a sublethal dose of APAP did not cause hepatic inflammation, hepatic proinflammatory cytokines and chemokines, such as Tnf-α, Kc, Mcp-1 and Mip2, were upregulated in VDD-fed mice treated with APAP. These results provide experimental data that VDD exacerbates hepatic oxidative stress and inflammation during APAP-induced ALI.
Collapse
Affiliation(s)
- Ya-Qi Wang
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Xiao-Pan Geng
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Ming-Wei Wang
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Hong-Qian Wang
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Xue He
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Shi-Min Liang
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.
| | - Xi Chen
- First Affiliated Hospital, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
10
|
Bhatt S, Kumar V, Dogra A, Ojha PK, Wazir P, Sangwan PL, Singh G, Nandi U. Amalgamation of in-silico, in-vitro and in-vivo approach to establish glabridin as a potential CYP2E1 inhibitor. Xenobiotica 2021; 51:625-635. [PMID: 33539218 DOI: 10.1080/00498254.2021.1883769] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CYP2E1 is directly or indirectly involved in the metabolism of ethanol and endogenous fatty acids but it plays a major role in the bio-activation of toxic substances that produce reactive metabolites leading to hepatotoxicity. Therefore, identification of CYP2E1 inhibitor from bioflavonoids class having useful pharmacological properties has dual benefit regarding avoidance of severe food-drug/nutraceutical-drug interaction and scope to develop a phytotherapeutics through an intended pharmacokinetic interaction.In the present study, we aimed to identify CYP2E1 inhibitor from experimental bioflavonoids which are unexplored for CYP2E1 inhibition till date using in-silico, in-vitro and in-vivo approaches.Results of in-vitro CYP2E1 inhibitory studies using CYP2E1-mediated chlorzoxazone 6-hydroxylation in human liver microsomes showed that glabridin have the highest potential than fisetin, epicatechin, nobiletin, and chrysin to inhibit CYP2E1 enzyme. Mechanistic investigations indicate that glabridin is a competitive CYP2E1 inhibitor. Molecular docking study results demonstrate that glabridin strongly interacted with the active site of human CYP2E1 enzyme. Pharmacokinetics of a CYP2E1 substrate in mice model indicates a significant alteration of chlorzoxazone and 6-hydroxychlorzoxazone plasma levels in the presence of glabridin. Further studies are needed to confirm the results at clinical level.Overall, glabridin is found to be a potential CYP2E1 inhibitor.
Collapse
Affiliation(s)
- Shipra Bhatt
- PK-PD, Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vinay Kumar
- Drug Theoretics and Chemoinformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Ashish Dogra
- PK-PD, Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Probir Kumar Ojha
- Drug Theoretics and Chemoinformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Priya Wazir
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Payare Lal Sangwan
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,Bio-Organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Gurdarshan Singh
- PK-PD, Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Utpal Nandi
- PK-PD, Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
11
|
New insight and potential therapy for NAFLD: CYP2E1 and flavonoids. Biomed Pharmacother 2021; 137:111326. [PMID: 33556870 DOI: 10.1016/j.biopha.2021.111326] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Over the years, the prevalence of nonalcoholic fatty liver disease (NAFLD) has increased year by year; however, due to its complicated pathogenesis, there is no effective treatment so far. It is reported that Cytochrome P450 2E1 (CYP2E1) plays an indispensable role in the development of NAFLD, and numerous studies have shown that flavonoids have a hepatoprotective effect and can exert a beneficial effect on NAFLD by regulating the activity of CYP2E1. Therefore, flavonoids may become effective drugs for the treatment of NAFLD in the future. This prompted us to review the research progress of the pathological mechanism of NAFLD and the impact of CYP2E1 activity changes during the pathological process, and to summarize the protective effect of flavonoids against CYP2E1 activity.
Collapse
|
12
|
Alshahrani S, Ashafaq M, Hussain S, Mohammed M, Sultan M, Jali AM, Siddiqui R, Islam F. Renoprotective effects of cinnamon oil against APAP-Induced nephrotoxicity by ameliorating oxidative stress, apoptosis and inflammation in rats. Saudi Pharm J 2021; 29:194-200. [PMID: 33679180 PMCID: PMC7910143 DOI: 10.1016/j.jsps.2021.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/05/2021] [Indexed: 12/22/2022] Open
Abstract
Acetaminophen (APAP) is used as a primary medication in relieving moderate pain and fever. However, APAP is associated with toxic effects in renal tissue that appear because of its free radicals property. The principle goal of the present work is to assess the kidney damage by APAP and its restore antioxidative property of cinnamon oil (CO). Animals were distributed into six animals each in six groups. Rats were administered with three varying doses of CO from 50 to 200 mg/kg b.w. respectively and only a single dose of APAP. APAP induced an alteration in serum biochemical markers, imbalance in oxidative parameters, morphological changes in kidney tissue along with increased interleukins cytokines (IL-1β & 6) and caspase (3, 9) levels. CO administration significantly ameliorates all the parameters and histopathological changes were restored. Moreover, it also restored the activities of antioxidative enzymes. Our work proved that an variance of oxidative markers in the kidney by APAP is ameliorated by CO in rats. Thus, CO could be used in reducing APAP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Saudi Arabia
| | - Mohammad Ashafaq
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Saudi Arabia
| | - Sohail Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Saudi Arabia
| | - Manal Mohammed
- Substance Abuse Research Center (SARC), College of Pharmacy, Jazan University, Saudi Arabia
| | - Muhammad Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Saudi Arabia
| | - Abdulmajeed M. Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Saudi Arabia
| | - Rahimullah Siddiqui
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Saudi Arabia
| | - Fakhrul Islam
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Saudi Arabia
| |
Collapse
|
13
|
Ding CH, Zhu H. Isatidis Folium alleviates acetaminophen-induced liver injury in mice by enhancing the endogenous antioxidant system. ENVIRONMENTAL TOXICOLOGY 2020; 35:1251-1259. [PMID: 32677766 DOI: 10.1002/tox.22990] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/11/2020] [Accepted: 06/06/2020] [Indexed: 06/11/2023]
Abstract
Isatidis Folium (IF) has been clinically combined with acetaminophen (APAP), but the rationality of combinational therapy is still ambiguous. In the present study, the protective effect and related mechanism of IF on APAP-induced hepatotoxicity were evaluated. Hepatic histopathology and blood biochemistry investigations clearly demonstrated that IF could restore APAP-induced hepatotoxicity. Liver distribution study indicated that the hepatoprotective effect of IF on APAP is attributed to the reduction of N-acetyl-p-benzoquinone imine (NAPQI) in liver, which is a known hepatotoxic metabolite of APAP. Further study suggested the reduction is not via decreasing the generation of NAPQI through inhibiting the enzyme activities of CYP 1A2, 2E1, and 3A4 but via accelerating the transformation of NAPQI to NAPQI-GSH by promoting GSH and decreasing GSSG contents in liver. Furthermore, IF significantly enhanced the hepatic activities of GSH-associated enzymes in APAP-treated mice. In summary, IF could alleviate APAP-induced hepatotoxicity by reducing the content of NAPQI via enhancing the level of GSH and the followed generation of NAPQI-GSH which might be ascribed to the upregulation of GSH-associated enzymes.
Collapse
Affiliation(s)
- Chuan-Hua Ding
- Department of Pharmacy, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - He Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pharmaceutical Analysis and Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
14
|
Mohos V, Fliszár-Nyúl E, Ungvári O, Bakos É, Kuffa K, Bencsik T, Zsidó BZ, Hetényi C, Telbisz Á, Özvegy-Laczka C, Poór M. Effects of Chrysin and Its Major Conjugated Metabolites Chrysin-7-Sulfate and Chrysin-7-Glucuronide on Cytochrome P450 Enzymes and on OATP, P-gp, BCRP, and MRP2 Transporters. Drug Metab Dispos 2020; 48:1064-1073. [PMID: 32661014 DOI: 10.1124/dmd.120.000085] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022] Open
Abstract
Chrysin is an abundant flavonoid in nature, and it is also contained by several dietary supplements. Chrysin is highly biotransformed in the body, during which conjugated metabolites chrysin-7-sulfate and chrysin-7-glucuronide are formed. These conjugates appear at considerably higher concentrations in the circulation than the parent compound. Based on previous studies, chrysin can interact with biotransformation enzymes and transporters; however, the interactions of its metabolites have been barely examined. In this in vitro study, the effects of chrysin, chrysin-7-sulfate, and chrysin-7-glucuronide on cytochrome P450 enzymes (2C9, 2C19, 3A4, and 2D6) as well as on organic anion-transporting polypeptides (OATPs; 1A2, 1B1, 1B3, and 2B1) and ATP binding cassette [P-glycoprotein, multidrug resistance-associated protein 2, and breast cancer resistance protein (BCRP)] transporters were investigated. Our observations revealed that chrysin conjugates are strong inhibitors of certain biotransformation enzymes (e.g., CYP2C9) and transporters (e.g., OATP1B1, OATP1B3, OATP2B1, and BCRP) examined. Therefore, the simultaneous administration of chrysin-containing dietary supplements with medications needs to be carefully considered due to the possible development of pharmacokinetic interactions. SIGNIFICANCE STATEMENT: Chrysin-7-sulfate and chrysin-7-glucuronide are the major metabolites of flavonoid chrysin. In this study, we examined the effects of chrysin and its conjugates on cytochrome P450 enzymes and on organic anion-transporting polypeptides and ATP binding cassette transporters (P-glycoprotein, breast cancer resistance protein, and multidrug resistance-associated protein 2). Our results demonstrate that chrysin and/or its conjugates can significantly inhibit some of these proteins. Since chrysin is also contained by dietary supplements, high intake of chrysin may interrupt the transport and/or the biotransformation of drugs.
Collapse
Affiliation(s)
- Violetta Mohos
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Orsolya Ungvári
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Éva Bakos
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Katalin Kuffa
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Tímea Bencsik
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Balázs Zoltán Zsidó
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Csaba Hetényi
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Ágnes Telbisz
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
15
|
Moghadamnia AA. Response to the Letter to the Editor by Hartmut Jaeschke and Anup Ramachandran Concerning Our Article "Chrysin Effect in Prevention of Acetaminophen-Induced Hepatotoxicity in Rat". Chem Res Toxicol 2020; 33:691-692. [PMID: 31986885 DOI: 10.1021/acs.chemrestox.0c00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ali Akbar Moghadamnia
- Cellular and Molecular Biology Research Center, Health Research Institute, Department of Pharmacology, Babol University of Medical Sciences, Babol 9811, Iran
| |
Collapse
|
16
|
Jaeschke H, Ramachandran A. Letter to the Editor Regarding the Article “Chrysin Effect in Prevention of Acetaminophen-Induced Hepatotoxicity in Rat” by Mohammadi and Co-Workers (2019). Chem Res Toxicol 2020; 33:689-690. [DOI: 10.1021/acs.chemrestox.9b00433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology, & Therapeutics University of Kansas Medical Center 3901 Rainbow Boulevard, MS 1018 Kansas City, Kansas 66160, United States
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology, & Therapeutics University of Kansas Medical Center 3901 Rainbow Boulevard, MS 1018 Kansas City, Kansas 66160, United States
| |
Collapse
|