1
|
Shariati S, Mohtadi S, Khodayar MJ, Salehcheh M, Azadnasab R, Mansouri E, Moosavi M. Quinic acid alleviates liver toxicity induced by acetaminophen in mice via anti-oxidative and anti-inflammatory effects. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03869-7. [PMID: 39985580 DOI: 10.1007/s00210-025-03869-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/30/2025] [Indexed: 02/24/2025]
Abstract
Acetaminophen (N-acetyl-para-aminophenol: APAP)-induced hepatotoxicity is a common toxicity that is associated with oxidative stress and inflammation. Quinic acid (QA) is a naturally occurring metabolite that exhibits antioxidant and anti-inflammatory properties. In this research, the effect of QA on hepatotoxicity caused by APAP was investigated. The mice were divided into six groups: control, APAP (300 mg/kg, i.p.), QA (100 mg/kg, i.p.), N-acetylcysteine (NAC) (100 mg/kg, i.p.), and treatment groups, which pretreated with QA at two doses of 50 and 100 mg/kg. NAC and QA were injected for 7 days, and APAP was injected on the seventh day. On day 8, mice were euthanized, and serum factors, markers of oxidative stress, tumor necrosis factor-α (TNF-α), and expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and cytochrome P450 2E1 (CYP2E1) proteins were measured. The results showed that the APAP-treated group significantly increased the activity of serum enzymes (alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase). APAP decreased hepatic total thiol content, as well as catalase, superoxide dismutase, and glutathione peroxidase activities, and increased thiobarbituric acid reactive substances and TNF-α levels. In addition, Nrf2 and CYP2E1 protein expression was upregulated in APAP-induced injury. Moreover, histopathological findings confirmed APAP hepatotoxicity. However, QA protected mice against the detrimental effects resulting from an imbalance in the oxidant/antioxidant system. QA ameliorated APAP-induced inflammation and histopathological changes and was able to upregulate the protein expression of Nrf2, while also reversing the increase in protein expression of CYP2E1 in APAP-intoxicated mice. These findings demonstrate the potential of QA in preventing APAP-induced hepatotoxicity, which is comparable to the effects of NAC.
Collapse
Affiliation(s)
- Saeedeh Shariati
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shokooh Mohtadi
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Salehcheh
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Azadnasab
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehrnoosh Moosavi
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
2
|
N-Acetyl Cysteine Overdose Inducing Hepatic Steatosis and Systemic Inflammation in Both Propacetamol-Induced Hepatotoxic and Normal Mice. Antioxidants (Basel) 2021; 10:antiox10030442. [PMID: 33809388 PMCID: PMC8000488 DOI: 10.3390/antiox10030442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Acetaminophen (APAP) overdose induces acute liver damage and even death. The standard therapeutic dose of N-acetyl cysteine (NAC) cannot be applied to every patient, especially those with high-dose APAP poisoning. There is insufficient evidence to prove that increasing NAC dose can treat patients who failed in standard treatment. This study explores the toxicity of NAC overdose in both APAP poisoning and normal mice. Two inbred mouse strains with different sensitivities to propacetamol-induced hepatotoxicity (PIH) were treated with different NAC doses. NAC therapy decreased PIH by reducing lipid oxidation, protein nitration and inflammation, and increasing glutathione (GSH) levels and antioxidative enzyme activities. However, the therapeutic effects of NAC on PIH were dose-dependent from 125 (N125) to 275 mg/kg (N275). Elevated doses of NAC (400 and 800 mg/kg, N400 and N800) caused additional deaths in both propacetamol-treated and normal mice. N800 treatments significantly decreased hepatic GSH levels and induced inflammatory cytokines and hepatic microvesicular steatosis in both propacetamol-treated and normal mice. Furthermore, both N275 and N400 treatments decreased serum triglyceride (TG) and induced hepatic TG, whereas N800 treatment significantly increased interleukin-6, hepatic TG, and total cholesterol levels. In conclusion, NAC overdose induces hepatic and systemic inflammations and interferes with fatty acid metabolism.
Collapse
|
3
|
Pingili R, Pawar AK, Challa SR. Quercetin reduced the formation of
N
‐acetyl‐
p
‐benzoquinoneimine, a toxic metabolite of paracetamol in rats and isolated rat hepatocytes. Phytother Res 2019; 33:1770-1783. [DOI: 10.1002/ptr.6365] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 02/24/2019] [Accepted: 03/16/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Ravindrababu Pingili
- Research and Development, Department of PharmacyJawaharlal Nehru Technological University Kakinada India
- Department of PharmacologyKVSR Siddhartha College of Pharmaceutical Sciences Vijayawada India
| | - A. Krishnamanjari Pawar
- Department of Pharmaceutical Analysis, University College of Pharmaceutical SciencesAndhra University Visakhapatnam India
| | - Siva Reddy Challa
- Department of PharmacologyKVSR Siddhartha College of Pharmaceutical Sciences Vijayawada India
| |
Collapse
|
4
|
Hepatoprotective Effect of Polysaccharides Isolated from Dendrobium officinale against Acetaminophen-Induced Liver Injury in Mice via Regulation of the Nrf2-Keap1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6962439. [PMID: 30116489 PMCID: PMC6079321 DOI: 10.1155/2018/6962439] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/20/2018] [Accepted: 05/20/2018] [Indexed: 02/07/2023]
Abstract
The effect of polysaccharides isolated from Dendrobium officinale (DOP) on acetaminophen- (APAP-) induced hepatotoxicity and the underlying mechanisms involved are investigated. Male Institute of Cancer Research (ICR) mice were randomly assigned to six groups: (1) control, (2) vehicle (APAP, 230 mg/kg), (3) N-acetylcysteine (100 mg/kg), (4) 50 mg/kg DOP, (5) 100 mg/kg DOP, and (6) 200 mg/kg DOP. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels in the serum and glutathione (GSH), malondialdehyde (MDA), catalase (CAT), total antioxidant capacity (T-AOC), myeloperoxidase (MPO), and reactive oxygen species (ROS) levels in the liver were determined after the death of the mice. The histological examination of the liver was also performed. The effect of DOP on the Kelch-like ECH-associated protein 1- (Keap1-) nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway was evaluated using Western blot analysis and real-time polymerase chain reaction (PCR). The results showed that DOP treatment significantly alleviated the hepatic injury. The decrease in ALT and AST levels in the serum and ROS, MDA, and MPO contents in the liver, as well as the increases in GSH, CAT, and T-AOC in the liver, were observed after DOP treatment. DOP treatment significantly induced the dissociation of Nrf2 from the Nrf2-Keap1 complex and promoted the Nrf2 nuclear translocation. Subsequently, DOP-mediated Nrf2 activation triggered the transcription and expressions of the glutamate-cysteine ligase catalytic (GCLC) subunit, glutamate-cysteine ligase regulatory subunit (GCLM), heme oxygenase-1 (HO-1), and NAD(P)H dehydrogenase quinone 1 (NQO1) in APAP-treated mice. The present study revealed that DOP treatment exerted potentially hepatoprotective effects against APAP-induced liver injury. Further investigation about mechanisms indicated that DOP exerted the hepatoprotective effect by suppressing the oxidative stress and activating the Nrf2-Keap1 signaling pathway.
Collapse
|
5
|
Wang G, Xiao K, Gao J, Jiang S, Wang S, Weng S, Xu C, Wang T, Qiao HL. Inhibitory effect of chlormethiazole on the toxicokinetics of diethylnitrosamine in normal and hepatofibrotic rats. Drug Chem Toxicol 2018; 42:600-607. [DOI: 10.1080/01480545.2018.1455204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Gaoju Wang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kang Xiao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| | - Jie Gao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| | - Shan Jiang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| | - Shang Wang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| | - Shijia Weng
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| | - Chen Xu
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| | - Tong Wang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| | - Hai-Ling Qiao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
3-Amino-1,2,4-triazole Limits the Oxidative Damage in UVA-Irradiated Dysplastic Keratinocytes. BIOMED RESEARCH INTERNATIONAL 2018; 2017:4872164. [PMID: 29387721 PMCID: PMC5745652 DOI: 10.1155/2017/4872164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/28/2017] [Accepted: 11/23/2017] [Indexed: 11/24/2022]
Abstract
Reactive oxygen species (ROS) generated by UVA irradiation affect the keratinocyte cell membrane, DNA, and proteins and may cause serious injury to the skin. Treating human dysplastic keratinocytes (DOK) with 3-amino-1,2,4-triazole (AMT), a common catalase inhibitor, induced a compensatory mechanism for the hydrogen peroxide detoxification, which included a rise in glutathione peroxidase and glutathione reductase activities. Here, we examined a possible role of AMT in protecting a human DOK cell line against UVA-induced damage. In DOK cells exposed to UVA irradiation, we observed a substantial decrease in antioxidant enzymatic activities, such as catalase, glutathione peroxidase, glutathione reductase, and glutathione-S-transferase and an increase in lipid peroxidation and protein oxidation levels. Treating DOK cells with AMT prior to UVA exposure enhanced the activities of glutathione peroxidase, glutathione reductase, and glutathione-S-transferase, relative to nontreated cells. The enhanced antioxidant activities were correlated with decreased protein oxidation levels. Based on these results, we suggest that AMT may protect dysplastic keratinocytes against the harmful effects of UVA radiation.
Collapse
|
7
|
Sun JB, Li Y, Cai YF, Huang Y, Liu S, Yeung PK, Deng MZ, Sun GS, Zilundu PL, Hu QS, An RX, Zhou LH, Wang LX, Cheng X. Scutellarin protects oxygen/glucose-deprived astrocytes and reduces focal cerebral ischemic injury. Neural Regen Res 2018; 13:1396-1407. [PMID: 30106052 PMCID: PMC6108207 DOI: 10.4103/1673-5374.235293] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Scutellarin, a bioactive flavone isolated from Scutellaria baicalensis, has anti-inflammatory, anti-neurotoxic, anti-apoptotic and anti-oxidative effects and has been used to treat cardiovascular and cerebrovascular diseases in China. However, the mechanisms by which scutellarin mediates neuroprotection in cerebral ischemia remain unclear. The interaction between scutellarin and nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) was assessed by molecular docking study, which showed that scutellarin selectively binds to NOX2 with high affinity. Cultures of primary astrocytes isolated from the cerebral cortex of neonatal Sprague-Dawley rats were pretreated with 2, 10 or 50 μM scutellarin for 30 minutes. The astrocytes were then subjected to oxygen/glucose deprivation by incubation for 2 hours in glucose-free Dulbecco's modified Eagle's medium in a 95% N2/5% CO2 incubator, followed by simulated reperfusion for 22 hours. Cell viability was assessed by cell counting kit-8 assay. Expression levels of NOX2, connexin 43 and caspase-3 were assessed by western blot assay. Reactive oxygen species were measured spectrophotometrically. Pretreatment with 10 or 50 μM scutellarin substantially increased viability, reduced the expression of NOX2 and caspase-3, increased the expression of connexin 43, and diminished the levels of reactive oxygen species in astrocytes subjected to ischemia-reperfusion. We also assessed the effects of scutellarin in vivo in the rat transient middle cerebral artery occlusion model of cerebral ischemia-reperfusion injury. Rats were given intraperitoneal injection of 100 mg/kg scutellarin 2 hours before surgery. The Bederson scale was used to assess neurological deficit, and 2,3,5-triphenyltetrazolium chloride staining was used to measure infarct size. Western blot assay was used to assess expression of NOX2 and connexin 43 in brain tissue. Enzyme-linked immunosorbent assay was used to detect 8-hydroxydeoxyguanosine (8-OHdG), 4-hydroxy-2-nonenal (4-HNE) and 3-nitrotyrosin (3-NT) in brain tissue. Immunofluorescence double staining was used to determine the co-expression of caspase-3 and NeuN. Pretreatment with scutellarin improved the neurological function of rats with focal cerebral ischemia, reduced infarct size, diminished the expression of NOX2, reduced levels of 8-OHdG, 4-HNE and 3-NT, and reduced the number of cells co-expressing caspase-3 and NeuN in the injured brain tissue. Furthermore, we examined the effect of the NOX2 inhibitor apocynin. Apocynin substantially increased connexin 43 expression in vivo and in vitro. Collectively, our findings suggest that scutellarin protects against ischemic injury in vitro and in vivo by downregulating NOX2, upregulating connexin 43, decreasing oxidative damage, and reducing apoptotic cell death.
Collapse
Affiliation(s)
- Jing-Bo Sun
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine; Guangdong Provincial Academy of Chinese Medical Sciences; Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, Guangdong Province, China
| | - Yan Li
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine; Guangdong Provincial Academy of Chinese Medical Sciences; Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, Guangdong Province, China
| | - Ye-Feng Cai
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine; Guangdong Provincial Academy of Chinese Medical Sciences; Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, Guangdong Province, China
| | - Yan Huang
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine; Guangdong Provincial Academy of Chinese Medical Sciences; Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, Guangdong Province, China
| | - Shu Liu
- Department of Anatomy, An Hui Medical University, Hefei, Anhui Province, China
| | - Patrick Kk Yeung
- Department of Biomedical Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Min-Zhen Deng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine; Guangdong Provincial Academy of Chinese Medical Sciences; Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, Guangdong Province, China
| | - Guang-Shun Sun
- Department of Preventive Medicine, School of Public Health, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Prince Lm Zilundu
- Guangzhou Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Qian-Sheng Hu
- Department of Preventive Medicine, School of Public Health, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Rui-Xin An
- Guangzhou Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Li-Hua Zhou
- Guangzhou Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Li-Xin Wang
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine; Guangdong Provincial Academy of Chinese Medical Sciences; Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, Guangdong Province, China
| | - Xiao Cheng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine; Guangdong Provincial Academy of Chinese Medical Sciences; Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, Guangdong Province, China
| |
Collapse
|
8
|
Choi N, Kim JW, Jeong H, Shin DG, Seo JH, Kim JH, Lim CW, Han KM, Kim B. Fermented ginseng, GBCK25, ameliorates steatosis and inflammation in nonalcoholic steatohepatitis model. J Ginseng Res 2017; 43:196-208. [PMID: 30962734 PMCID: PMC6437395 DOI: 10.1016/j.jgr.2017.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/27/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022] Open
Abstract
Background Nonalcoholic steatohepatitis (NASH) is one of the chronic inflammatory liver diseases and a leading cause of advanced liver fibrosis, cirrhosis, and hepatocellular carcinoma. The main purpose of this study was to clarify the effects of GBCK25 fermented by Saccharomyces servazzii GB-07 and pectinase, on NASH severity in mice. Methods Six-wk-old male mice were fed either a normal diet (ND) or a Western diet (WD) for 12 wks to induce NASH. Each group was orally administered with vehicle or GBCK25 once daily at a dose of 10 mg/kg, 20 mg/kg, 100 mg/kg, 200 mg/kg, or 400 mg/kg during that time. The effects of GBCK25 on cellular damage and inflammation were determined by in vitro experiments. Results Histopathologic analysis and hepatic/serum biochemical levels revealed that WD-fed mice showed severe steatosis and liver injury compared to ND-fed mice. Such lesions were significantly decreased in the livers of WD-fed mice with GBCK25 administration. Consistently, mRNA expression levels of NASH-related inflammatory-, fibrogenic-, and lipid metabolism-related genes were decreased in the livers of WD-fed mice administered with GBCK25 compared to WD-fed mice. Western blot analysis revealed decreased protein levels of cytochrome P450 2E1 (CYP2E1) with concomitantly reduced activation of c-Jun N-terminal kinase (JNK) in the livers of WD-fed mice administered with GBCK25. Also, decreased cellular damage and inflammation were observed in alpha mouse liver 12 (AML12) cells and RAW264.7 cells, respectively. Conclusion Administration of GBCK25 ameliorates NASH severity through the modulation of CYP2E1 and its associated JNK-mediated cellular damage. GBCK25 could be a potentially effective prophylactic strategy to prevent metabolic diseases including NASH.
Collapse
Affiliation(s)
- Naeun Choi
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program), College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Jong Won Kim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program), College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Hyeneui Jeong
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program), College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Dong Gue Shin
- Research & Development Center of GENERAL BIO Co., Ltd, Namwon City, Jeollabuk-Do, Republic of Korea
| | - Jeong Hun Seo
- Research & Development Center of GENERAL BIO Co., Ltd, Namwon City, Jeollabuk-Do, Republic of Korea
| | - Jong Hoon Kim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program), College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Chae Woong Lim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program), College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Kang Min Han
- Department of Pathology, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Bumseok Kim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program), College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| |
Collapse
|
9
|
Zheng W, Zhang Z, Liu S, Bi J, Zhang J, Du L, Ding X, Liu C. Remote ischemic conditioning protects against acetaminophen-induced acute liver injury in mice. Hepatol Res 2017; 47:234-245. [PMID: 26990366 DOI: 10.1111/hepr.12702] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/22/2016] [Accepted: 03/14/2016] [Indexed: 12/20/2022]
Abstract
AIM Acetaminophen (APAP) overdose is a major cause of drug-induced acute liver failure. Studies have shown that remote ischemic pre- and post-conditioning (R-IPC and R-IPOST) can protect the liver against ischemia-reperfusion (I/R) and lipopolysaccharide-induced injuries. The aim of this study was to investigate the effect of R-IPC and R-IPOST on APAP-induced hepatotoxicity in mice. METHODS Mice were randomized (n = 6 per group) to seven major groups: (i) normal control; (ii) sham operated; (iii) APAP; (iv) R-IPC + APAP; (v) R-IPC + APAP + zinc protoporphyrin (ZnPP); (vi) R-IPOST + APAP; and (vii) R-IPOST + APAP + ZnPP. Sixteen hours after APAP treatment, mouse liver and serum were collected to determine the severity of liver injury. RESULTS The results showed that R-IPC and R-IPOST significantly decreased APAP-induced serum levels of alanine aminotransferase, aspartate aminotransferase, tumor necrosis factor-α, interleukin-6, and hepatic malondialdehyde, as well as nitrotyrosine formation. Both R-IPC and R-IPOST could improve the hepatic superoxide dismutase, glutathione, and glutathione peroxidase activities and depress the expressions of pro-inflammatory associated proteins, such as inducible nitric oxide synthetase and nuclear factor-κB. They could also increase heme oxygenase-1 expression; however, ZnPP could counteract this protective effect. CONCLUSION Remote ischemic conditioning has significant therapeutic potential in APAP-induced hepatotoxicity by inhibiting oxidative stress and inflammation and promoting heme oxygenase-1 expression.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University.,Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital
| | - Zhiyong Zhang
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital
| | - Sushun Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Jianbin Bi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Lixue Du
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| |
Collapse
|
10
|
Chen XH, Ma L, Hu YX, Wang DX, Fang L, Li XL, Zhao JC, Yu HR, Ying HZ, Yu CH. Transcriptome profiling and pathway analysis of hepatotoxicity induced by tris (2-ethylhexyl) trimellitate (TOTM) in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 41:62-71. [PMID: 26650799 DOI: 10.1016/j.etap.2015.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/02/2015] [Accepted: 11/08/2015] [Indexed: 06/05/2023]
Abstract
Tris (2-ethylhexyl) trimellitate (TOTM) is commonly used as an alternative plasticizer for medical devices. But very little information was available on its biological effects. In this study, we investigated toxicity effects of TOTM on hepatic differential gene expression analyzed by using high-throughput sequencing analysis for over-represented functions and phenotypically anchored to complementary histopathologic, and biochemical data in the liver of mice. Among 1668 candidate genes, 694 genes were up-regulated and 974 genes were down-regulated after TOTM exposure. Using Gene Ontology analysis, TOTM affected three processes: the cell cycle, metabolic process and oxidative activity. Furthermore, 11 key genes involved in the above processes were validated by real time PCR. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that these genes were involved in the cell cycle pathway, lipid metabolism and oxidative process. It revealed the transcriptome gene expression response to TOTM exposure in mouse, and these data could contribute to provide a clearer understanding of the molecular mechanisms of TOTM-induced hepatotoxicity in human.
Collapse
Affiliation(s)
- Xian-Hua Chen
- Key Laboratory for Medical Device Safety Evaluation and Research, Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou 310018, China
| | - Li Ma
- Key Laboratory for Medical Device Safety Evaluation and Research, Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou 310018, China
| | - Yi-Xiang Hu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Dan-Xian Wang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Li Fang
- Key Laboratory for Medical Device Safety Evaluation and Research, Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou 310018, China
| | - Xue-Lai Li
- Key Laboratory for Medical Device Safety Evaluation and Research, Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou 310018, China
| | - Jin-Chuan Zhao
- Key Laboratory for Medical Device Safety Evaluation and Research, Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou 310018, China
| | - Hai-Rong Yu
- Key Laboratory for Medical Device Safety Evaluation and Research, Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou 310018, China
| | - Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China.
| |
Collapse
|
11
|
Patel N, Bayliss GP. Developments in extracorporeal therapy for the poisoned patient. Adv Drug Deliv Rev 2015; 90:3-11. [PMID: 26050528 DOI: 10.1016/j.addr.2015.05.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/25/2015] [Accepted: 05/30/2015] [Indexed: 12/18/2022]
Abstract
The modern use of extracorporeal therapies to treat poisoning and drug overdoses dates back to the early 20th century and has evolved along with their use as treatment for acute kidney injury or as maintenance therapy in advanced kidney disease. As our understanding of drug pharmacokinetics and membrane materials has increased, the technologies of extracorporeal therapy and their applications have become more sophisticated. Despite that, there is little robust evidence to guide clinicians on the optimal use of extracorporeal therapy in treating poisoning beyond case reports and series. New efforts are underway to remedy that: the Extracorporeal Treatments in Poisoning Workgroup (EXTRIP) is an international effort on the part of nephrologists, pharmacists and toxicologists to review the available data and formulate evidence-based guidelines on how to use extracorporeal techniques to treat poisoning and improve patient outcomes. Meanwhile, new techniques and membranes are under development. This review will summarize those key scientific and technologic developments, the efforts to optimize their use and new directions in research.
Collapse
|