1
|
Chiew AL, Isbister GK. Advances in the understanding of acetaminophen toxicity mechanisms: a clinical toxicology perspective. Expert Opin Drug Metab Toxicol 2023; 19:601-616. [PMID: 37714812 DOI: 10.1080/17425255.2023.2259787] [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: 05/15/2023] [Revised: 08/02/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023]
Abstract
INTRODUCTION Acetaminophen (paracetamol) is a commonly used analgesic and antipyretic agent, which is safe in therapeutic doses. Acetaminophen poisoning due to self-harm or repeated supratherapeutic ingestion is a common cause of acute liver injury. Acetylcysteine has been a mainstay of treatment for acetaminophen poisoning for decades and is efficacious if administered early. However, treatment failures occur if administered late, in 'massive' overdoses or in high-risk patients. AREAS COVERED This review provides an overview of the mechanisms of toxicity of acetaminophen poisoning (metabolic and oxidative phase) and how this relates to the assessment and treatment of the acetaminophen poisoned patient. The review focuses on how these advances offer further insight into the utility of novel biomarkers and the role of proposed adjunct treatments. EXPERT OPINION Advances in our understanding of acetaminophen toxicity have allowed the development of novel biomarkers and a better understanding of how adjunct treatments may prevent acetaminophen toxicity. Newly proposed adjunct treatments like fomepizole are being increasingly used without robust clinical trials. Novel biomarkers (not yet clinically available) may provide better assessment of these newly proposed adjunct treatments, particularly in clinical trials. These advances in our understanding of acetaminophen toxicity and liver injury hold promise for improved diagnosis and treatment.
Collapse
Affiliation(s)
- Angela L Chiew
- Department of Clinical Toxicology, Prince of Wales Hospital, Randwick, NSW, Australia
- Faculty of Medicine, The University of New South Wales, Sydney, New South Wales, Australia
- New South Wales Poisons Information Centre, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | - Geoffrey K Isbister
- New South Wales Poisons Information Centre, Sydney Children's Hospital, Sydney, New South Wales, Australia
- Clinical Toxicology Research Group, University of Newcastle, Callaghan, NSW, Australia
- Department of Clinical Toxicology, Calvary Mater Newcastle, Waratah, NSW, Australia
| |
Collapse
|
2
|
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
|
3
|
Deng Y, Zhao Z, Sheldon M, Zhao Y, Teng H, Martinez C, Zhang J, Lin C, Sun Y, Yao F, Zhu H, Ma L. LIFR recruits HGF-producing neutrophils to promote liver injury repair and regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.18.533289. [PMID: 36993315 PMCID: PMC10055204 DOI: 10.1101/2023.03.18.533289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
The molecular links between tissue repair and tumorigenesis remain elusive. Here, we report that loss of the liver tumor suppressor Lifr in mouse hepatocytes impairs the recruitment and activity of reparative neutrophils, resulting in the inhibition of liver regeneration after partial hepatectomy or toxic injuries. On the other hand, overexpression of LIFR promotes liver repair and regeneration after injury. Interestingly, LIFR deficiency or overexpression does not affect hepatocyte proliferation ex vivo or in vitro . In response to physical or chemical damage to the liver, LIFR from hepatocytes promotes the secretion of the neutrophil chemoattractant CXCL1 (which binds CXCR2 to recruit neutrophils) and cholesterol in a STAT3-dependent manner. Cholesterol, in turn, acts on the recruited neutrophils to secrete hepatocyte growth factor (HGF) to accelerate hepatocyte proliferation and regeneration. Altogether, our findings reveal a LIFR-STAT3- CXCL1-CXCR2 axis and a LIFR-STAT3-cholesterol-HGF axis that mediate hepatic damage- induced crosstalk between hepatocytes and neutrophils to repair and regenerate the liver.
Collapse
|
4
|
Offor SJ, Amadi CN, Chijioke-Nwauche I, Manautou JE, Orisakwe OE. Potential deleterious effects of paracetamol dose regime used in Nigeria versus that of the United States of America. Toxicol Rep 2022; 9:1035-1044. [PMID: 36561959 PMCID: PMC9764198 DOI: 10.1016/j.toxrep.2022.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 12/25/2022] Open
Abstract
Paracetamol, also known as acetaminophen (N-acetyl-para-aminophenol, APAP) is the world's most used over-the-counter analgesic-antipyretic drug. Despite its good safety profile, acetaminophen can cause severe hepatotoxicity in overdose, and poisoning from paracetamol has become a major public health concern. Paracetamol is now the major cause of acute liver failure in the United States and Europe. This systematic review aims at examining the likelihood of paracetamol use in Nigeria causing more liver toxicity vis-à-vis the reduced maximum recommended daily adult dose of 3 g for the 500 mg tablet. Online searches were conducted in the databases of PubMed, Google Scholar and MEDLINE for publications using terms like "paracetamol toxicity," "acetaminophen and liver toxicity," "paracetamol and liver diseases in Nigeria," and other variants. Further search of related references in PubMed was carried out, and synthesis of all studies included in this review finalized. There were 94 studies that met the inclusion criteria. Evaluation of hepatic disorder was predicated mostly on a constellation of clinical features and limited clinical laboratory investigations. Determination of blood paracetamol concentration was rarely reported, thus excluding paracetamol poisoning as one of the likely causes of liver disorders in Nigeria. In Nigeria and elsewhere, several factors are known to increase paracetamol's predisposition to liver injury. They include: the over-the-counter status of paracetamol, use of fixed-dose combinations of paracetamol with other drugs, malnutrition, dose miscalculations, and chronic alcohol consumption. The tendency to exceed the new paracetamol maximum daily dose of 3 g in Nigeria may increase its risk for hepatotoxicity than observed in the United States of America known for emphasizing lower dose of the drug. In addition to recommending the new maximal daily paracetamol dose allowance, the historical maximum daily adult dose of 4 g should be de-emphasized in Nigeria.
Collapse
Affiliation(s)
- Samuel James Offor
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Uyo, Uyo, Akwa Ibom State, Nigeria
| | - Cecilia Nwadiuto Amadi
- Department of Experimental Pharmacology & Toxicology, Faculty of Pharmacy, University of Port-Harcourt, Port-Harcourt, Rivers State, Nigeria
| | - Ifeyinwa Chijioke-Nwauche
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Port Harcourt, Port Harcourt 5323, Rivers State, Nigeria
| | - Jose E. Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Orish E. Orisakwe
- Department of Experimental Pharmacology & Toxicology, Faculty of Pharmacy, University of Port-Harcourt, Port-Harcourt, Rivers State, Nigeria
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
| |
Collapse
|
5
|
The Timing and Effects of Low-Dose Ethanol Treatment on Acetaminophen-Induced Liver Injury. Life (Basel) 2021; 11:life11101094. [PMID: 34685467 PMCID: PMC8539755 DOI: 10.3390/life11101094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/27/2022] Open
Abstract
Acetaminophen (APAP) overdose is the major cause of drug-induced liver injury and acute liver failure. Approximately 10% of APAP is metabolized by cytochrome P450 (CYP2E1) into toxic N-acetyl-p-benzoquinone imine (NAPQI). CYP2E1 also contributes to ethanol metabolism, especially during conditions of high blood ethanol concentration. Acute and chronic ethanol consumption appears to have opposite effects on APAP-induced liver injury. We determined the effects of different doses, pre- and post-treatment, and various schedules of ethanol exposure in APAP-induced liver injury. Treatment with ethanol (0.5 g/kg) after 1 h of APAP (300 mg/kg) administration decreased serum ALT levels, histopathological features, and inflammatory cell infiltration. Moreover, ethanol treatment 1 h after APAP treatment reduced APAP-induced liver injury compared with later administration. Interestingly, ethanol pretreatment did not provide any protective effect. Furthermore, ethanol treatment was associated with a significant decrease in ERK and AKT phosphorylation during the acute injury phase. Ethanol exposure also increased CYP2E1 expression and decreased PCNA expression during the liver regeneration phase.
Collapse
|
6
|
Ahmed EA, Abd-Eldayem AM, Ahmed E. Can granulocyte colony stimulating factor (G-CSF) ameliorate acetaminophen-induced hepatotoxicity? Hum Exp Toxicol 2021; 40:1755-1766. [PMID: 33882750 DOI: 10.1177/09603271211008522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acetaminophen (APAP) is often used as an antipyretic and analgesic agent. Overdose hepatotoxicity, which often results in liver cell failure and liver transplantation, is a severe complication of APAP usage. To save the liver and save lives from acute liver damage caused by APAP, the search for new strategies for liver defense is important. Wistar rats have been used for the induction of APAP hepatotoxicity. Elevated levels of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) and lactate dehydrogenase (LDH) were evaluated for liver toxicity. In addition, the levels of hepatic tissue oxidative markers such as malondialdehyde (MDA), nitric oxide (NO) increased while glutathione (GSH) was depleted and catalase (CAT) activity was curtailed. The biochemical findings were consistent with the changes in histology that suggested liver damage and inflammation. Treated rats with N-acetylcysteine (N-AC) and granulocyte colony stimulating factor (G-CSF) showed a decrease in serum levels of ALT, AST and LDH, while the level of ALP in the G-CSF group was still high. After administration of APAP, treatment with N-AC or G-CSF substantially reduced the level of MDA and NO while maintaining the GSH content and CAT activity. Treatment with N-AC and G-CSF after administration of APAP has also attenuated inflammation and hepatocytes necrosis. The results of this study showed that G-CSF could be viewed as an alternative hepatoprotective agent against APAP-induced acute liver injury compared to N-AC.
Collapse
Affiliation(s)
- E A Ahmed
- Department of Pharmacology, Faculty of Medicine, 68796Assiut University, Assiut, Egypt.,Centre of Excellence in Environmental Studies (CEES), King Abdulaziz University, Jeddah, Saudi Arabia
| | - A M Abd-Eldayem
- Department of Pharmacology, Faculty of Medicine, 68796Assiut University, Assiut, Egypt.,Al-Ghad International Colleges of Applied Medical Sciences, ABHA / Male, Saudi Arabia
| | - E Ahmed
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, 68796Assiut University, Assiut, Egypt
| |
Collapse
|
7
|
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
|
8
|
Ahmed LA, Abd El-Rhman RH, Gad AM, Hassaneen SK, El-Yamany MF. Dibenzazepine combats acute liver injury in rats via amendments of Notch signaling and activation of autophagy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:337-348. [PMID: 32984915 DOI: 10.1007/s00210-020-01977-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/13/2020] [Indexed: 02/08/2023]
Abstract
Paracetamol is a commonly used over-the-counter analgesic and antipyretic drug. Nevertheless, an overdose of paracetamol leads to hepatic necrosis that can be lethal. This study aimed to assess the potential hepatoprotective effects of dibenzazepine, a Notch inhibitor, against acute liver injury in rats via interfering with oxidative stress, inflammation, apoptosis, autophagy, and Notch signaling. Silymarin (200 mg/kg, p.o.) or dibenzazepine (2 mg/kg, i.p.) were administered to rats for 5 days before a single hepatotoxic dose of paracetamol (800 mg/kg, i.p.). Pretreatment with silymarin and dibenzazepine significantly mitigated oxidative stress, inflammatory and apoptotic markers induced by paracetamol hepatotoxicity where dibenzazepine showed greater repression of inflammation. Furthermore, dibenzazepine was found to be significantly more efficacious than silymarin in inhibiting Notch signaling as represented by expression of Notch-1 and Hes-1. A significantly greater response was also demonstrated with dibenzazepine pretreatment with regard to the expression of autophagic proteins, Beclin-1 and LC-3. The aforementioned biochemical results were confirmed by histopathological examination. Autophagy and Notch signaling seem to play a significant role in protection provided by dibenzazepine for paracetamol-induced hepatotoxicity in rats, which could explain its superior results relative to silymarin. Graphical abstract.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Rana H Abd El-Rhman
- Department of Pharmacology, Egyptian Drug Authority formerly National Organization for Drug Control and Research, Giza, Egypt
| | - Amany M Gad
- Department of Pharmacology, Egyptian Drug Authority formerly National Organization for Drug Control and Research, Giza, Egypt
| | - Sherifa K Hassaneen
- Department of Pharmacology, Egyptian Drug Authority formerly National Organization for Drug Control and Research, Giza, Egypt
| | - Mohamad F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Jaeschke H, Murray FJ, Monnot AD, Jacobson-Kram D, Cohen SM, Hardisty JF, Atillasoy E, Hermanowski-Vosatka A, Kuffner E, Wikoff D, Chappell GA, Bandara SB, Deore M, Pitchaiyan SK, Eichenbaum G. Assessment of the biochemical pathways for acetaminophen toxicity: Implications for its carcinogenic hazard potential. Regul Toxicol Pharmacol 2021; 120:104859. [PMID: 33388367 DOI: 10.1016/j.yrtph.2020.104859] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
In 2019 California's Office of Environmental Health Hazard Assessment (OEHHA) initiated a review of the carcinogenic hazard potential of acetaminophen. In parallel with this review, herein we evaluated the mechanistic data related to the steps and timing of cellular events following therapeutic recommended (≤4 g/day) and higher doses of acetaminophen that may cause hepatotoxicity to evaluate whether these changes indicate that acetaminophen is a carcinogenic hazard. At therapeutic recommended doses, acetaminophen forms limited amounts of N-acetyl-p-benzoquinone-imine (NAPQI) without adverse cellular effects. Following overdoses of acetaminophen, there is potential for more extensive formation of NAPQI and depletion of glutathione, which may result in mitochondrial dysfunction and DNA damage, but only at doses that result in cell death - thus making it implausible for acetaminophen to induce the kind of stable, genetic damage in the nucleus indicative of a genotoxic or carcinogenic hazard in humans. The collective data demonstrate a lack of a plausible mechanism related to carcinogenicity and are consistent with rodent cancer bioassays, epidemiological results reviewed in companion manuscripts in this issue, as well as conclusions of multiple international health authorities.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- University of Kansas Medical Center, Department of Pharmacology, Toxicology & Therapeutics, Kansas City, KS, USA
| | | | | | | | - Samuel M Cohen
- University of Nebraska Medical Center, Havlik-Wall Professor of Oncology, Department of Pathology and Microbiology, Omaha, NE, USA
| | - Jerry F Hardisty
- Experimental Pathology Laboratories, Inc., Research Triangle Park, NC, USA
| | | | | | - Edwin Kuffner
- Johnson & Johnson Consumer Health, Fort Washington, PA, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
McGill MR, Hinson JA. The development and hepatotoxicity of acetaminophen: reviewing over a century of progress. Drug Metab Rev 2020; 52:472-500. [PMID: 33103516 DOI: 10.1080/03602532.2020.1832112] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acetaminophen (APAP) was first synthesized in the 1800s, and came on the market approximately 65 years ago. Since then, it has become one of the most used drugs in the world. However, it is also a major cause of acute liver failure. Early investigations of the mechanisms of toxicity revealed that cytochrome P450 enzymes catalyze formation of a reactive metabolite in the liver that depletes glutathione and covalently binds to proteins. That work led to the introduction of N-acetylcysteine (NAC) as an antidote for APAP overdose. Subsequent studies identified the reactive metabolite N-acetyl-p-benzoquinone imine, specific P450 enzymes involved, the mechanism of P450-mediated oxidation, and major adducted proteins. Significant gaps remain in our understanding of the mechanisms downstream of metabolism, but several events appear critical. These events include development of an initial oxidative stress, reactive nitrogen formation, altered calcium flux, JNK activation and mitochondrial translocation, inhibition of mitochondrial respiration, the mitochondrial permeability transition, and nuclear DNA fragmentation. Additional research is necessary to complete our knowledge of the toxicity, such as the source of the initial oxidative stress, and to greatly improve our understanding of liver regeneration after APAP overdose. A better understanding of these mechanisms may lead to additional treatment options. Even though NAC is an excellent antidote, its effectiveness is limited to the first 16 hours following overdose.
Collapse
Affiliation(s)
- Mitchell R McGill
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, Little Rock, AR, USA.,Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jack A Hinson
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
11
|
Murray FJ, Monnot AD, Jacobson-Kram D, Cohen SM, Hardisty JF, Bandara SB, Kovochich M, Deore M, Pitchaiyan SK, Gelotte CK, Lai JCK, Atillasoy E, Hermanowski-Vosatka A, Kuffner E, Unice KM, Yang K, Gebremichael Y, Howell BA, Eichenbaum G. A critical review of the acetaminophen preclinical carcinogenicity and tumor promotion data and their implications for its carcinogenic hazard potential. Regul Toxicol Pharmacol 2020; 118:104801. [PMID: 33039518 DOI: 10.1016/j.yrtph.2020.104801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 12/22/2022]
Abstract
In 2019 the California Office of Environmental Health Hazard Assessment (OEHHA) initiated a review of the carcinogenic hazard potential of acetaminophen, including an assessment of the long-term rodent carcinogenicity and tumor initiation/promotion studies. The objective of the analysis herein was to inform this review process with a weight-of-evidence assessment of these studies and an assessment of the relevance of these models to humans. In most of the 14 studies, there were no increases in the incidences of tumors in any organ system. In the few studies in which an increase in tumor incidence was observed, there were factors such as absence of a dose response and a rodent-specific tumor supporting that these findings are not relevant to human hazard identification. In addition, we performed qualitative analysis and quantitative simulations of the exposures to acetaminophen and its metabolites and its toxicity profile; the data support that the rodent models are toxicologically relevant to humans. The preclinical carcinogenicity results are consistent with the broader weight of evidence assessment and evaluations of multiple international health authorities supporting that acetaminophen is not a carcinogenic hazard.
Collapse
Affiliation(s)
| | | | | | - Samuel M Cohen
- Havlik-Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Kyunghee Yang
- DILIsym Services Inc., Research Triangle Park, NC, USA
| | | | | | | |
Collapse
|
12
|
Stefanello ST, de Carvalho NR, Reis SB, Soares FAA, Barcelos RP. Acetaminophen Oxidation and Inflammatory Markers - A Review of Hepatic Molecular Mechanisms and Preclinical Studies. Curr Drug Targets 2020; 21:1225-1236. [PMID: 32386489 DOI: 10.2174/1389450121666200510014418] [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: 11/01/2019] [Revised: 01/06/2020] [Accepted: 02/21/2020] [Indexed: 11/22/2022]
Abstract
Acetaminophen is a widely used analgesic for pain management, especially useful in chronic diseases, such as rheumatoid arthritis. However, easy access to this medicine has increased the occurrence of episodes of poisoning. Patients often develop severe liver damage, which may quickly lead to death. Consequently, numerous studies have been conducted to identify new biomarkers that allow the prediction of the degree of acetaminophen intoxication and thus intervene in a timely manner to save patients' lives. This review highlights the main mechanisms of the induction and progression of liver damage arising from acetaminophen poisoning. In addition, we have discussed the possibility of using new clinical biomarkers for detecting acetaminophen poisoning.
Collapse
Affiliation(s)
- Silvio Terra Stefanello
- Programa de Pos-Graduacao em Bioquimica Toxicologica, Centro de Ciencias Naturais e Exatas (CCNE), Universidade Federal de Santa Maria, Brazil
| | | | - Simone Beder Reis
- Institudo de Ciencias Biologicas (ICB), Programa de Posgraduacao em Bioexperimentacao, Universidade de Passo Fundo, Passo Fundo, Brazil
| | - Felix Alexandre Antunes Soares
- Programa de Pos-Graduacao em Bioquimica Toxicologica, Centro de Ciencias Naturais e Exatas (CCNE), Universidade Federal de Santa Maria, Brazil
| | - Rômulo Pillon Barcelos
- Programa de Pos-Graduacao em Bioquimica Toxicologica, Centro de Ciencias Naturais e Exatas (CCNE), Universidade Federal de Santa Maria, Brazil
| |
Collapse
|
13
|
Abstract
Acetaminophen (APAP) is one of the most popular and safe pain medications worldwide. However, due to its wide availability, it is frequently implicated in intentional or unintentional overdoses where it can cause severe liver injury and even acute liver failure (ALF). In fact, APAP toxicity is responsible for 46% of all ALF cases in the United States. Early mechanistic studies in mice demonstrated the formation of a reactive metabolite, which is responsible for hepatic glutathione depletion and initiation of the toxicity. This insight led to the rapid introduction of N-acetylcysteine as a clinical antidote. However, more recently, substantial progress was made in further elucidating the detailed mechanisms of APAP-induced cell death. Mitochondrial protein adducts trigger a mitochondrial oxidant stress, which requires amplification through a MAPK cascade that ultimately results in activation of c-jun N-terminal kinase (JNK) in the cytosol and translocation of phospho-JNK to the mitochondria. The enhanced oxidant stress is responsible for the membrane permeability transition pore opening and the membrane potential breakdown. The ensuing matrix swelling causes the release of intermembrane proteins such as endonuclease G, which translocate to the nucleus and induce DNA fragmentation. These pathophysiological signaling mechanisms can be additionally modulated by removing damaged mitochondria by autophagy and replacing them by mitochondrial biogenesis. Importantly, most of the mechanisms have been confirmed in human hepatocytes and indirectly through biomarkers in plasma of APAP overdose patients. The extensive necrosis caused by APAP overdose leads to a sterile inflammatory response. Although recruitment of inflammatory cells is necessary for removal of cell debris in preparation for regeneration, these cells have the potential to aggravate the injury. This review touches on the newest insight into the intracellular mechanisms of APAP-induced cells death and the resulting inflammatory response. Furthermore, it discusses the translation of these findings to humans and the emergence of new therapeutic interventions.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
Cha H, Lee S, Lee JH, Park JW. Protective effects of p-coumaric acid against acetaminophen-induced hepatotoxicity in mice. Food Chem Toxicol 2018; 121:131-139. [DOI: 10.1016/j.fct.2018.08.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 08/03/2018] [Accepted: 08/23/2018] [Indexed: 12/16/2022]
|
15
|
Ndetan H, Evans MW, Singal AK, Brunner LJ, Calhoun K, Singh KP. Light to moderate drinking and therapeutic doses of acetaminophen: An assessment of risks for renal dysfunction. Prev Med Rep 2018; 12:253-258. [PMID: 30377576 PMCID: PMC6205325 DOI: 10.1016/j.pmedr.2018.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/15/2018] [Accepted: 10/21/2018] [Indexed: 11/17/2022] Open
Abstract
This study investigated the potential effect of therapeutic doses of acetaminophen (APAP) in combination with light-moderate amounts of alcohol on kidney functions controlling for factors such as hypertension, diabetes and obesity that may predispose the kidney to APAP and/or alcohol toxicity. Secondary analysis of the 2003–2004 National Health and Nutrition Examination Survey data was performed using SAS 9.4. Odds ratios (OR) and 95% confidence intervals (CI) comparing the likelihood that individuals who ingested therapeutic doses of APAP and light-moderate amount of alcohol, compared to those who did not, would have kidney dysfunction were generated from multiple logistics regression models by further controlling for potential predisposing factors namely hypertension, diabetes and obesity. Kidney dysfunction was defined based on self-reports and laboratory examination of serum creatinine (SCr), blood urea nitrogen (BUN), glomerular filtration rate (GFR) and albumin creatinine ratio (ABCR). Statistically significant increased odds of renal dysfunction were noted among respondents who reported use of therapeutic doses of APAP and light-moderate amount of alcohol [OR(95% CI) = 1.64(1.28–2.10) self-report, 2.18(1.81–2.63) SCr, 4.60(3.03–7.00) BUN, 3.14(2.42–4.07) GFR, and 1.71(1.36–2.14) ALBCR)] even after adjusting for hypertension, diabetes and obesity [Adjusted OR (95% CI) = 1.78 (1.22–2.58) self-report, 2.05 (1.07–3.92) GFR]. The toxic effects of APAP and alcohol on the kidney were hypothesized. The threshold doses at which these effects begin to occur are unknown. The findings of this study suggest that even therapeutic doses of APAP and light-moderate amount of alcohol could be health problematic if consumed concomitantly. Even therapeutic doses of acetaminophen may be associated with renal dysfunction. Light-moderate drinking alone may be protective of renal dysfunction. Light-moderate alcohol may enhance effect of therapeutic doses of APAP on kidney. Hypertension, obesity & diabetes may strengthen effect of APAP & alcohol on kidney.
Collapse
Affiliation(s)
- Harrison Ndetan
- Department of Epidemiology and Biostatistics, School of Community and Rural Health, University of Texas Health Science Center, 11937 US Hwy 271, Tyler, TX 75708, United States of America
| | - Marion W Evans
- Department of Food Science, Nutrition, and Health Promotion, 105 Herzer Building, Box 9805, Mississippi State, MS 39762, United States of America
| | - Ashwani K Singal
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Alabama Birmingham, 1720 2 Ave, South, BDB 380, Birmingham, AL 35294-0012, United States of America
| | - Lane J Brunner
- Ben and Maytee Fisch College of Pharmacy, University of Texas, Tyler, 3900 University Blvd, Tyler, TX 75799, United States of America
| | - Kirk Calhoun
- University of Texas Health Science Center, 11937 US Hwy 271, Tyler, TX 75708, United States of America
| | - Karan P Singh
- Department of Epidemiology and Biostatistics, School of Community and Rural Health, University of Texas Health Science Center, 11937 US Hwy 271, Tyler, TX 75708, United States of America
| |
Collapse
|
16
|
Tsuchiya Y, Sakai H, Hirata A, Yanai T. Brazilian green propolis suppresses acetaminophen-induced hepatocellular necrosis by modulating inflammation-related factors in rats. J Toxicol Pathol 2018; 31:275-282. [PMID: 30393431 PMCID: PMC6206282 DOI: 10.1293/tox.2018-0027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/05/2018] [Indexed: 12/11/2022] Open
Abstract
Propolis is a resin-like material produced by honey bees from bud exudates and sap of plants and their own secretions. An ethanol extract of Brazilian green propolis (EEBGP) contains prenylated phenylpropanoids and flavonoids and has antioxidative and anti-inflammatory effects. Acetaminophen (N-acetyl-p-aminophenol; APAP) is a typical hepatotoxic drug, and APAP-treated rats are widely used as a model of drug-induced liver injury. Oxidative stress and inflammatory reactions cause APAP-induced hepatocellular necrosis and are also related to expansion of the lesion. In the present study, we investigated the preventive effects of EEBGP on APAP-induced hepatocellular necrosis in rats and the protective mechanism including the expression of antioxidative enzyme genes and inflammation-related genes. A histological analysis revealed that administration 0.3% EEBGP in the diet for seven days reduced centrilobular hepatocellular necrosis with inflammatory cell infiltration induced by oral administration of APAP (800 mg/kg) and significantly reduced the area of necrosis. EEBGP administration did not significantly change the mRNA expression levels of antioxidant enzyme genes in the liver of APAP-treated rats but decreased the mRNA expression of cytokines including Il10 and Il1b, with a significant difference in Il10 expression. In addition, the decrease in the mRNA levels of the Il1b and Il10 genes significantly correlated with the decrease in the percentage of hepatocellular necrosis. These findings suggest that EEBGP could suppress APAP-induced hepatocellular necrosis by modulating cytokine expression.
Collapse
Affiliation(s)
- Yuya Tsuchiya
- Nagaragawa Research Center, API Co., Ltd., 692-3 Nagara, Gifu-shi, Gifu 502-0071, Japan.,Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Hiroki Sakai
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Akihiro Hirata
- Division of Animal Experiment, Life Science Research Center, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1194, Japan
| | - Tokuma Yanai
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| |
Collapse
|
17
|
Nikravesh H, Khodayar MJ, Mahdavinia M, Mansouri E, Zeidooni L, Dehbashi F. Protective Effect of Gemfibrozil on Hepatotoxicity Induced by Acetaminophen in Mice: the Importance of Oxidative Stress Suppression. Adv Pharm Bull 2018; 8:331-339. [PMID: 30023335 PMCID: PMC6046434 DOI: 10.15171/apb.2018.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 03/10/2018] [Accepted: 04/08/2018] [Indexed: 12/24/2022] Open
Abstract
Purpose: Gemfibrozil (GEM) apart from agonist activity at peroxisome proliferator-activated receptor-alpha (PPAR-α) has antioxidant and anti-inflammatory properties. Accordingly, the present study was designed to investigate the protective effect of GEM on acute liver toxicity induced by acetaminophen (APAP) in mice. Methods: In this study, mice divided in seven groups include, control group, APAP group, GEM group, three APAP groups pretreated with GEM at the doses of 25, 50 and 100 mg/kg respectively and APAP group pretreated with N-Acetyl cysteine. GEM, NAC or vehicle were administered for 10 days. In last day, GEM and NAC were gavaged 1 h before and 1 h after APAP injection. Twenty four hours after APAP, mice were sacrificed. Serum parameters include alanine aminotransferase (ALT), aspartate aminotransferase (AST) and liver tissue markers including catalase enzyme activity, reactive oxygen species (ROS), malondialdehyde and reduced glutathione (GSH) levels determined and histopathological parameters measured. Results: GEM led to significant decrease in serum ALT and AST activities and increase in catalase activity and hepatic GSH level and reduces malondialdehyde and ROS levels in the liver tissue. In confirmation, histopathological findings revealed that GEM decrease degeneration, vacuolation and necrosis of hepatocytes and infiltration of inflammatory cells. Conclusion: Present data demonstrated that GEM has antioxidant properties and can protect the liver from APAP toxicity, just in the same pathway that toxicity occurs by toxic ROS and that GEM may be an alternative therapeutic agent to NAC in APAP toxicity.
Collapse
Affiliation(s)
- Hojatolla Nikravesh
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Zeidooni
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fereshteh Dehbashi
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
18
|
Tsuchiya Y, Sakai H, Hirata A, Yanai T. Effects of food restriction on the expression of genes related to acetaminophen-induced liver toxicity in rats. J Toxicol Pathol 2018; 31:267-274. [PMID: 30393430 PMCID: PMC6206280 DOI: 10.1293/tox.2018-0009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/17/2018] [Indexed: 01/26/2023] Open
Abstract
It is well known that fasting substantially affects the metabolism of drugs and chemicals. Food restriction also affects drug kinetics, such as absorption, metabolism, and excretion, and therefore, it can potentially modulate the onset of chemical toxicity or drug-induced adverse reactions. In the present study, the expression of drug-metabolizing enzyme genes and total glutathione content in the liver, which are related to toxicity induced by overdose of the hepatotoxic drug acetaminophen (N-acetyl-p-aminophenol; APAP), were examined in rats reared under different feeding conditions: ad libitum feeding, 16-h fasting, and food restriction (fed 70% of the average intake of ad libitum feeding for 10 days) conditions. The rats under food restriction conditions as well as fasted rats showed significantly higher expression of Cyp2e1, the gene encoding the enzyme that metabolizes APAP to its toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). They also had lower levels of liver total glutathione, which detoxifies NAPQI. In contrast, the gene expression of UDP-glucuronosyltransferase 1A6 (Ugt1a6), sulfotransferase 1A1 (Sult1a1), and glutathione S-transferase M1 (Gstm1) was not affected by food restriction or fasting. When APAP was administered (800 mg/kg), histopathological changes were not observed in rats fed ad libitum, while hepatocellular necrosis was observed in most of the rats treated with APAP after fasting or food restriction. Taken together, these results suggest that not only fasting but also food restriction exacerbate APAP-induced acute liver injury, probably by the induction of CYP2E1 and the reduction of liver glutathione contents, in rodents.
Collapse
Affiliation(s)
- Yuya Tsuchiya
- Nagaragawa Research Center, API Co., Ltd., 692-3 Nagara, Gifu-shi, Gifu 502-0071, Japan.,Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Hiroki Sakai
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Akihiro Hirata
- Division of Animal Experiment, Life Science Research Center, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Tokuma Yanai
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| |
Collapse
|
19
|
Cao P, Sun J, Sullivan MA, Huang X, Wang H, Zhang Y, Wang N, Wang K. Angelica sinensis polysaccharide protects against acetaminophen-induced acute liver injury and cell death by suppressing oxidative stress and hepatic apoptosis in vivo and in vitro. Int J Biol Macromol 2018; 111:1133-1139. [DOI: 10.1016/j.ijbiomac.2018.01.139] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/16/2018] [Accepted: 01/19/2018] [Indexed: 02/07/2023]
|
20
|
Abstract
Acetaminophen (APAP) is the leading cause of acute liver failure (ALF), although the worldwide frequency is variable. APAP hepatotoxicity develops either following intentional overdose or unintentional ingestion (therapeutic misadventure) in the background of several factors, such as concomitant use of alcohol and certain medications that facilitate the formation of reactive and toxic metabolites. Spontaneous survival is more common in APAP-induced ALF compared with non-APAP etiologies. N-acetylcysteine is recommended for all patients with APAP-induced ALF and it reduces mortality. Liver transplantation should be offered early to those who are unlikely to survive based on described prognostic criteria.
Collapse
|
21
|
Le paracétamol à dose thérapeutique : quelles populations à risque d’hépatotoxicité ? TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2018. [DOI: 10.1016/j.toxac.2017.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Papackova Z, Heczkova M, Dankova H, Sticova E, Lodererova A, Bartonova L, Poruba M, Cahova M. Silymarin prevents acetaminophen-induced hepatotoxicity in mice. PLoS One 2018; 13:e0191353. [PMID: 29342206 PMCID: PMC5771617 DOI: 10.1371/journal.pone.0191353] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
Acetaminophen or paracetamol (APAP) overdose is a common cause of liver injury. Silymarin (SLM) is a hepatoprotective agent widely used for treating liver injury of different origin. In order to evaluate the possible beneficial effects of SLM, Balb/c mice were pretreated with SLM (100 mg/kg b.wt. per os) once daily for three days. Two hours after the last SLM dose, the mice were administered APAP (300 mg/kg b.wt. i.p.) and killed 6 (T6), 12 (T12) and 24 (T24) hours later. SLM-treated mice exhibited a significant reduction in APAP-induced liver injury, assessed according to AST and ALT release and histological examination. SLM treatment significantly reduced superoxide production, as indicated by lower GSSG content, lower HO-1 induction, alleviated nitrosative stress, decreased p-JNK activation and direct measurement of mitochondrial superoxide production in vitro. SLM did not affect the APAP-induced decrease in CYP2E1 activity and expression during the first 12 hrs. Neutrophil infiltration and enhanced expression of inflammatory markers were first detected at T12 in both groups. Inflammation progressed in the APAP group at T24 but became attenuated in SLM-treated animals. Histological examination suggests that necrosis the dominant cell death pathway in APAP intoxication, which is partially preventable by SLM pretreatment. We demonstrate that SLM significantly protects against APAP-induced liver damage through the scavenger activity of SLM and the reduction of superoxide and peroxynitrite content. Neutrophil-induced damage is probably secondary to necrosis development.
Collapse
Affiliation(s)
- Zuzana Papackova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Veterinary Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czech Republic
- * E-mail:
| | - Marie Heczkova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Helena Dankova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Sticova
- Clinical and Transplant Pathology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Alena Lodererova
- Clinical and Transplant Pathology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lenka Bartonova
- Clinical and Transplant Pathology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Poruba
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Monika Cahova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
23
|
Kong SZ, Lin GS, Liu JJ, Su LY, Zeng L, Luo DD, Su ZR, Wang HF. Hepatoprotective Effect of Ultrafine Powder of Dendrobium officinale against Acetaminophen-Induced Liver Injury in Mice. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2018. [DOI: 10.3136/fstr.24.339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Song-Zhi Kong
- Guangdong Ocean University, Faculty of Chemistry and Environmental Science
| | - Guo-Sheng Lin
- Guangzhou University of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine
- Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine
| | - Jing-Jing Liu
- Guangzhou University of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine
- Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine
| | - Ling-Ye Su
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization
- Guangdong Academy of Forestry
| | - Lei Zeng
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization
- Guangdong Academy of Forestry
| | - Dan-Dan Luo
- Guangzhou University of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine
- Guangdong Academy of Forestry
| | - Zi-Ren Su
- Guangzhou University of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine
- Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine
| | - Hong-Feng Wang
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization
- Guangdong Academy of Forestry
| |
Collapse
|
24
|
Andonakakis A, Kwiatt K. Anesthesia for Weight Reduction Surgery. Anesthesiology 2018. [DOI: 10.1007/978-3-319-74766-8_81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
25
|
Bos JC, Mistício MC, Nunguiane G, Mathôt RAA, van Hest RM, Prins JM. Paracetamol clinical dosing routine leads to paracetamol underexposure in an adult severely ill sub-Saharan African hospital population: a drug concentration measurement study. BMC Res Notes 2017; 10:671. [PMID: 29202789 PMCID: PMC5715499 DOI: 10.1186/s13104-017-3016-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022] Open
Abstract
Background Hospitals in sub-Saharan Africa (SSA) continue to receive high numbers of severely ill (HIV-infected) patients with physical pain that may suffer from hepatic and renal dysfunction. Paracetamol is widely used for pain relief in this setting but it is unknown whether therapeutic drug concentrations are attained. The aim of this study was to assess the occurrence of therapeutic, sub-therapeutic and toxic paracetamol concentrations in SSA adult hospital population. Methods In a cross-sectional study, plasma paracetamol concentrations were measured in patients with an oral prescription in a referral hospital in Mozambique. From August to November 2015, a maximum of four blood samples were drawn on different time points for paracetamol concentration measurement and biochemical analysis. Study endpoints were the percentage of participants with therapeutic (≥ 10 and ≤ 20 mg/L), sub-therapeutic (< 10 mg/L) and toxic (> 75 mg/L) concentrations. Results Seventy-six patients with a median age of 37 years, a body mass index of 18.2, a haemoglobin concentration of 10.3 g/dL and an albumin of 29 g/L yielded 225 samples. 13.4% of participants had one or more therapeutic paracetamol concentrations. 86.6% had a sub-therapeutic concentration at all time points and 70.2% had two or more concentrations below the lower limit of quantification. No potentially toxic concentrations were found. Conclusions Routine oral dosing practices in a SSA hospital resulted in substantial underexposure to paracetamol. Palliation is likely to be sub-standard and oral palliative drug pharmacokinetics and dispensing procedures in this setting need further investigation.
Collapse
Affiliation(s)
- Jeannet C Bos
- Division of Infectious Diseases, Department of Internal Medicine, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Mabor C Mistício
- Faculty of Health Sciences, Research Centre for Infectious Diseases (CIDI), Catholic University of Mozambique, Rua Marquês do Soveral 960, C.P. 821, Beira, Mozambique
| | - Ginto Nunguiane
- Faculty of Health Sciences, Research Centre for Infectious Diseases (CIDI), Catholic University of Mozambique, Rua Marquês do Soveral 960, C.P. 821, Beira, Mozambique
| | - Ron A A Mathôt
- Division of Clinical Pharmacology, Department of Hospital Pharmacy, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Reinier M van Hest
- Division of Clinical Pharmacology, Department of Hospital Pharmacy, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jan M Prins
- Division of Infectious Diseases, Department of Internal Medicine, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Caparrotta TM, Antoine DJ, Dear JW. Are some people at increased risk of paracetamol-induced liver injury? A critical review of the literature. Eur J Clin Pharmacol 2017; 74:147-160. [PMID: 29067481 PMCID: PMC5765191 DOI: 10.1007/s00228-017-2356-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/15/2017] [Indexed: 12/18/2022]
Abstract
Purpose Paracetamol is one of the world’s most commonly used drugs. In overdose, it is well established to be hepatotoxic. The aim of this review was to identify factors that have been, or actually are, associated with the development of liver injury after paracetamol exposure in humans. Method Google Scholar and PubMed were searched on various dates between December 2016 and March 2017. Papers identified had their references analysed for further studies that might be relevant. Results At the time of writing, there was little good quality clinical evidence—from studies of paracetamol overdose or therapeutic use—to suggest that any groups of people are relatively protected from, or are at greater risk of, liver injury. The factors that were historically used to indicate higher risk in the UK have no good quality clinical evidence to support their re-introduction into clinical practice. The safe (and still effective) oral dose of paracetamol in patients weighing less than 50 kg has not been established. Conclusion There is no patient group that is unequivocally at elevated risk of paracetamol-induced liver toxicity. We propose two clinical scenarios that warrant further research. Firstly, there is a need to establish whether the dose of paracetamol should be reduced in patients with low body weight. Secondly, if or when genomic information regarding individual patients becomes readily available to inform prescribing, we propose the contribution of the genome to paracetamol toxicity should be re-investigated with robustly designed studies. Such studies could enhance the safe use of one of the most frequently taken drugs. Electronic supplementary material The online version of this article (10.1007/s00228-017-2356-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Thomas M Caparrotta
- Speciality Registrar Clinical Pharmacology and Therapeutics, NHS Lothian, Edinburgh, UK
| | - Daniel J Antoine
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - James W Dear
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
27
|
Selenoprotein MsrB1 deficiency exacerbates acetaminophen-induced hepatotoxicity via increased oxidative damage. Arch Biochem Biophys 2017; 634:69-75. [PMID: 28986131 DOI: 10.1016/j.abb.2017.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/27/2017] [Accepted: 09/30/2017] [Indexed: 01/01/2023]
Abstract
Acetaminophen (APAP) overdose induces acute liver damage and failure via reactive oxygen species production and glutathione (GSH) depletion. Methionine sulfoxide reductase B1 (MsrB1) is an antioxidant selenoenzyme that specifically catalyzes the reduction of methionine R-sulfoxide residues. In this study, we used MsrB1 gene-knockout mice and primary hepatocytes to investigate the effect of MsrB1 on APAP-induced hepatotoxicity. Analyses of histological alterations and serum indicators of liver damage showed that MsrB1-/- mice were more susceptible to APAP-induced acute liver injury than wild-type (MsrB1+/+) mice. Consistent with the in vivo results, primary MsrB1-/- hepatocytes displayed higher susceptibility to APAP-induced cytotoxicity than MsrB1+/+ cells. MsrB1 deficiency increased hepatic oxidative stress after APAP challenge such as hydrogen peroxide production, lipid peroxidation, and protein oxidation levels. Additionally, basal and APAP-induced ratios of reduced-to-oxidized GSH (GSH/GSSG) were significantly lower in MsrB1-/- than in MsrB1+/+ livers. Nrf2 nuclear accumulation and heme oxygenase-1 expression levels after APAP challenge were lower in MsrB1-/- than in MsrB1+/+ livers, suggesting that MsrB1 deficiency attenuates the APAP-induced activation of Nrf2. Collectively, the results of this study suggest that selenoprotein MsrB1 plays a protective role against APAP-induced hepatotoxicity via its antioxidative function.
Collapse
|
28
|
Kress HG, Untersteiner G. Clinical update on benefit versus risks of oral paracetamol alone or with codeine: still a good option? Curr Med Res Opin 2017; 33:289-304. [PMID: 27842443 DOI: 10.1080/03007995.2016.1254606] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND After decades of worldwide use of paracetamol/acetaminophen as a popular and apparently safe prescription and over-the-counter medicine, the future role of this poorly understood analgesic has been seriously questioned by recent concerns about prenatal, cardiovascular (CV) and hepatic safety, and also about its analgesic efficacy. At the same time the usefulness of codeine in combination products has come under debate. METHODS Based on a PubMed database literature search on the terms efficacy, safety, paracetamol, acetaminophen, codeine and their combinations up to and including June 2016, this clinical update reviews the current evidence of the benefit and risks of oral paracetamol alone and with codeine for mild-to-moderate pain in adults, and compares the respective efficacy and safety profiles with those of nonsteroidal anti-inflammatory drugs (NSAIDs). RESULTS Whereas there is a clear strong association of NSAID use and gastrointestinal (GI) and CV morbidity and mortality, evidence for paracetamol with and without codeine supports the recommended use even in most vulnerable individuals, such as the elderly, pregnant women, alcoholics, and compromised GI and CV patients. The controversies and widespread misconceptions about the complex hepatic metabolism and potential hepatotoxicity have been corrected by recent reviews, and paracetamol remains the first-line nonopioid analgesic in patients with liver diseases if notes of caution are applied. CONCLUSION Due to its safety and tolerability profile paracetamol remained a first-line treatment in many international guidelines. Alone and with codeine it is a safe and effective option in adults, whilst NSAIDs are obviously less safe as alternatives, given the risk of potentially fatal GI and CV adverse effects.
Collapse
|
29
|
Singh MP, Kim KY, Kim HY. Methionine sulfoxide reductase A deficiency exacerbates acute liver injury induced by acetaminophen. Biochem Biophys Res Commun 2017; 484:189-194. [PMID: 28104395 DOI: 10.1016/j.bbrc.2017.01.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 01/06/2017] [Indexed: 01/12/2023]
Abstract
Acetaminophen (APAP) overdose induces acute liver injury via enhanced oxidative stress and glutathione (GSH) depletion. Methionine sulfoxide reductase A (MsrA) acts as a reactive oxygen species scavenger by catalyzing the cyclic reduction of methionine-S-sulfoxide. Herein, we investigated the protective role of MsrA against APAP-induced liver damage using MsrA gene-deleted mice (MsrA-/-). We found that MsrA-/- mice were more susceptible to APAP-induced acute liver injury than wild-type mice (MsrA+/+). The central lobule area of the MsrA-/- liver was more impaired with necrotic lesions. Serum alanine transaminase, aspartate transaminase, and lactate dehydrogenase levels were significantly higher in MsrA-/- than in MsrA+/+ mice after APAP challenge. Deletion of MsrA enhanced APAP-induced hepatic GSH depletion and oxidative stress, leading to increased susceptibility to APAP-induced liver injury in MsrA-deficient mice. APAP challenge increased Nrf2 activation more profoundly in MsrA-/- than in MsrA+/+ livers. Expression and nuclear accumulation of Nrf2 and its target gene expression were significantly elevated in MsrA-/- than in MsrA+/+ livers after APAP challenge. Taken together, our results demonstrate that MsrA protects the liver from APAP-induced toxicity. The data provided herein constitute the first in vivo evidence of the involvement of MsrA in hepatic function under APAP challenge.
Collapse
Affiliation(s)
- Mahendra Pratap Singh
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu, 42415, Republic of Korea; School of Bioengineering and Biosciences, Department of Zoology, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Ki Young Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu, 42415, Republic of Korea
| | - Hwa-Young Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu, 42415, Republic of Korea.
| |
Collapse
|
30
|
González-Ponce HA, Martínez-Saldaña MC, Rincón-Sánchez AR, Sumaya-Martínez MT, Buist-Homan M, Faber KN, Moshage H, Jaramillo-Juárez F. Hepatoprotective Effect of Opuntia robusta and Opuntia streptacantha Fruits against Acetaminophen-Induced Acute Liver Damage. Nutrients 2016; 8:E607. [PMID: 27782042 PMCID: PMC5083995 DOI: 10.3390/nu8100607] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/20/2016] [Indexed: 12/25/2022] Open
Abstract
Acetaminophen (APAP)-induced acute liver failure (ALF) is a serious health problem in developed countries. N-acetyl-L-cysteine (NAC), the current therapy for APAP-induced ALF, is not always effective, and liver transplantation is often needed. Opuntia spp. fruits are an important source of nutrients and contain high levels of bioactive compounds, including antioxidants. The aim of this study was to evaluate the hepatoprotective effect of Opuntia robusta and Opuntia streptacantha extracts against APAP-induced ALF. In addition, we analyzed the antioxidant activities of these extracts. Fruit extracts (800mg/kg/day, orally) were given prophylactically to male Wistar rats before intoxication with APAP (500 mg/kg, intraperitoneally). Rat hepatocyte cultures were exposed to 20mmol/LAPAP, and necrosis was assessed by LDH leakage. Opuntia robusta had significantly higher levels of antioxidants than Opuntia streptacantha. Both extracts significantly attenuated APAP-induced injury markers AST, ALT and ALP and improved liver histology. The Opuntia extracts reversed APAP-induced depletion of liver GSH and glycogen stores. In cultured hepatocytes, Opuntia extracts significantly reduced leakage of LDH and cell necrosis, both prophylactically and therapeutically. Both extracts appeared to be superior to NAC when used therapeutically. We conclude that Opuntia extracts are hepatoprotective and can be used as a nutraceutical to prevent ALF.
Collapse
Affiliation(s)
- Herson Antonio González-Ponce
- Department of Physiology and Pharmacology, Basic Science Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20131, Mexico;
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands; (M.B.-H.); (K.N.F.); (H.M.)
| | | | - Ana Rosa Rincón-Sánchez
- Department of Physiology, University Center of Health Sciences, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - María Teresa Sumaya-Martínez
- Food Technology Unit, Secretary of Research and Graduate Studies, Universidad Autónoma de Nayarit, Tepic 63160, Mexico;
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands; (M.B.-H.); (K.N.F.); (H.M.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands; (M.B.-H.); (K.N.F.); (H.M.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands; (M.B.-H.); (K.N.F.); (H.M.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Fernando Jaramillo-Juárez
- Department of Physiology and Pharmacology, Basic Science Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20131, Mexico;
| |
Collapse
|
31
|
Yoon E, Babar A, Choudhary M, Kutner M, Pyrsopoulos N. Acetaminophen-Induced Hepatotoxicity: a Comprehensive Update. J Clin Transl Hepatol 2016; 4:131-142. [PMID: 27350943 PMCID: PMC4913076 DOI: 10.14218/jcth.2015.00052] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/07/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatic injury and subsequent hepatic failure due to both intentional and non-intentional overdose of acetaminophen (APAP) has affected patients for decades, and involves the cornerstone metabolic pathways which take place in the microsomes within hepatocytes. APAP hepatotoxicity remains a global issue; in the United States, in particular, it accounts for more than 50% of overdose-related acute liver failure and approximately 20% of the liver transplant cases. The pathophysiology, disease course and management of acute liver failure secondary to APAP toxicity remain to be precisely elucidated, and adverse patient outcomes with increased morbidity and mortality continue to occur. Although APAP hepatotoxicity follows a predictable timeline of hepatic failure, its clinical presentation might vary. N-acetylcysteine (NAC) therapy is considered as the mainstay therapy, but liver transplantation might represent a life-saving procedure for selected patients. Future research focus in this field may benefit from shifting towards obtaining antidotal knowledge at the molecular level, with focus on the underlying molecular signaling pathways.
Collapse
Affiliation(s)
- Eric Yoon
- Rutgers New Jersey Medical School, University Hospital, Newark, New Jersey, USA
| | - Arooj Babar
- Rutgers New Jersey Medical School, University Hospital, Newark, New Jersey, USA
| | - Moaz Choudhary
- Rutgers New Jersey Medical School, University Hospital, Newark, New Jersey, USA
| | - Matthew Kutner
- Rutgers New Jersey Medical School, University Hospital, Newark, New Jersey, USA
| | | |
Collapse
|
32
|
Badr H, Kozai D, Sakaguchi R, Numata T, Mori Y. Different Contribution of Redox-Sensitive Transient Receptor Potential Channels to Acetaminophen-Induced Death of Human Hepatoma Cell Line. Front Pharmacol 2016; 7:19. [PMID: 26903865 PMCID: PMC4746322 DOI: 10.3389/fphar.2016.00019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/21/2016] [Indexed: 01/30/2023] Open
Abstract
Acetaminophen (APAP) is a safe analgesic antipyretic drug at prescribed doses. Its overdose, however, can cause life-threatening liver damage. Though, involvement of oxidative stress is widely acknowledged in APAP-induced hepatocellular death, the mechanism of this increased oxidative stress and the associated alterations in Ca2+ homeostasis are still unclear. Among members of transient receptor potential (TRP) channels activated in response to oxidative stress, we here identify that redox-sensitive TRPV1, TRPC1, TRPM2, and TRPM7 channels underlie Ca2+ entry and downstream cellular damages induced by APAP in human hepatoma (HepG2) cells. Our data indicate that APAP treatment of HepG2 cells resulted in increased reactive oxygen species (ROS) production, glutathione (GSH) depletion, and Ca2+ entry leading to increased apoptotic cell death. These responses were significantly suppressed by pretreatment with the ROS scavengers N-acetyl-L-cysteine (NAC) and 4,5-dihydroxy-1,3-benzene disulfonic acid disodium salt monohydrate (Tiron), and also by preincubation of cells with the glutathione inducer Dimethylfumarate (DMF). TRP subtype-targeted pharmacological blockers and siRNAs strategy revealed that suppression of either TRPV1, TRPC1, TRPM2, or TRPM7 reduced APAP-induced ROS formation, Ca2+ influx, and cell death; the effects of suppression of TRPV1 or TRPC1, known to be activated by oxidative cysteine modifications, were stronger than those of TRPM2 or TRPM7. Interestingly, TRPV1 and TRPC1 were labeled by the cysteine-selective modification reagent, 5,5′-dithiobis (2-nitrobenzoic acid)-2biotin (DTNB-2Bio), and this was attenuated by pretreatment with APAP, suggesting that APAP and/or its oxidized metabolites act directly on the modification target cysteine residues of TRPV1 and TRPC1 proteins. In human liver tissue, TRPV1, TRPC1, TRPM2, and TRPM7 channels transcripts were localized mainly to hepatocytes and Kupffer cells. Our findings strongly suggest that APAP-induced Ca2+ entry and subsequent hepatocellular death are regulated by multiple redox-activated cation channels, among which TRPV1 and TRPC1 play a prominent role.
Collapse
Affiliation(s)
- Heba Badr
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University Kyoto, Japan
| | - Daisuke Kozai
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University Kyoto, Japan
| | - Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyoto, Japan; World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto UniversityKyoto, Japan
| | - Tomohiro Numata
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyoto, Japan; Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto UniversityKyoto, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyoto, Japan; World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto UniversityKyoto, Japan; Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto UniversityKyoto, Japan
| |
Collapse
|
33
|
Abstract
More than one in four American adults consume alcohol in quantities exceeding recommended limits. One in 12 have an alcohol use disorder marked by harmful consequences. Both types of alcohol misuse contribute to acute injury and chronic disease, making alcohol the third largest cause of preventable death in the United States. Alcohol misuse alters the management of common conditions from insomnia to anemia. Primary care providers should screen adult patients to identify the full spectrum of alcohol misuse. A range of effective treatments are available - from brief counselling interventions and mutual help groups to medications and behavioral therapies.
Collapse
Affiliation(s)
- Douglas Berger
- General Medicine Service, VA Puget Sound, Seattle, WA 98108, USA; Department of Medicine, University of Washington, Seattle, WA 98101, USA.
| | - Katharine A Bradley
- Department of Medicine, University of Washington, Seattle, WA 98101, USA; Department of Health Services, University of Washington, Seattle, WA 98101, USA; Group Health Research Institute, Seattle, WA, USA; VA Health Services Research & Development (HSR&D) and Center of Excellence in Substance Abuse Treatment and Education (CESATE), Seattle, WA, USA
| |
Collapse
|
34
|
Chen PJ, Cai SP, Huang C, Meng XM, Li J. Protein tyrosine phosphatase 1B (PTP1B): A key regulator and therapeutic target in liver diseases. Toxicology 2015; 337:10-20. [PMID: 26299811 DOI: 10.1016/j.tox.2015.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/12/2015] [Accepted: 08/15/2015] [Indexed: 12/11/2022]
Abstract
Phosphorylation of tyrosine residues within proteins, which is controlled by the reciprocal action of protein tyrosine kinases and protein tyrosine phosphatases, plays a key role in regulating almost all physiological responses. Therefore, it comes as no surprise that once the balance of tyrosine phosphorylation is disturbed, drastic effects can occur. Protein tyrosine phosphatase 1B (PTP1B), a classical non-transmembrane tyrosine phosphatase, is a pivotal regulator and promising drug target in type 2 diabetes and obesity. Recently it has received renewed attention in liver diseases and represents an intriguing opportunity as a drug target by modulating hepatocyte death and survival, hepatic lipogenesis and so on. Here, the multiple roles of PTP1B in liver diseases will be presented, with respect to liver regeneration, drug-induced liver disease, non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Pei-Jie Chen
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Shuang-Peng Cai
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Cheng Huang
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Xiao-Ming Meng
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Jun Li
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
35
|
Zhou X, Cheung CM, Yang JM, Or PMY, Lee WYW, Yeung JHK. Danshen (Salvia miltiorrhiza) water extract inhibits paracetamol-induced toxicity in primary rat hepatocytes via reducing CYP2E1 activity and oxidative stress. J Pharm Pharmacol 2015; 67:980-989. [PMID: 25645193 DOI: 10.1111/jphp.12381] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 12/07/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVES This study aimed to investigate the protective effects of Danshen (Salvia miltiorrhiza) water extract (DSE) and its major phenolic acid components against CYP2E1-mediated paracetamol (APAP)-induced hepatic toxicity. METHODS The protection and underlying mechanisms were detected in CYP2E1 overexpression primary rat hepatocytes by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, alamar blue assay, CYP2E1 inhibition assay and glutathione assay. KEY FINDINGS After APAP treatment, DSE (0.06-1 mg/ml) significantly increased cell viability in MTT assay. Two major components danshensu (8.2-130.5 μm) and salvianolic acid B (Sal B; 3.3-53.5 μm) mainly contributed to this protection, but rosmarinic acid, protocatechuic aldehyde and Sal A did not. Alamar blue assay showed that DSE, danshensu and Sal B maintained mitochondrial metabolic activity. DSE inhibited CYP2E1 (Ki = 1.46 mg/ml) in a mixed mode in rat liver microsomes in vitro; DSE decreased APAP-induced total glutathione depletion and preserved redox status (GSH/GSSG ratio) in hepatocytes. Danshensu and Sal B did not inhibit CYP2E1 or decrease total glutathione depletion, but preserved redox status. CONCLUSIONS DSE protected hepatocytes against APAP-induced injury via maintenance of mitochondrial metabolic activity, CYP2E1 inhibition, reduction of total glutathione depletion and preservation of redox status. Danshensu and Sal B were mainly responsible for this protection.
Collapse
Affiliation(s)
- Xuelin Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ching Mei Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jia-Ming Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Penelope M Y Or
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wayne Y W Lee
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - John H K Yeung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
36
|
Yiang GT, Yu YL, Lin KT, Chen JN, Chang WJ, Wei CW. Acetaminophen induces JNK/p38 signaling and activates the caspase-9-3-dependent cell death pathway in human mesenchymal stem cells. Int J Mol Med 2015; 36:485-92. [PMID: 26096646 PMCID: PMC4501662 DOI: 10.3892/ijmm.2015.2254] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 06/05/2015] [Indexed: 02/05/2023] Open
Abstract
Acetaminophen (APAP) is a widely used analgesic and antipyretic drug. Generally, the therapeutic dose of APAP is clinically safe, however, high doses of APAP can cause acute liver and kidney injury. Therefore, the majority of previous studies have focussed on elucidating the mechanisms of APAP-induced hepatotoxicity and nephrotoxicity, in addition to examining ways to treat these conditions in clinical cases. However, few studies have reported APAP-induced intoxication in human stem cells. Stem cells are important in cell proliferation, differentiation and repair during human development, particularly during fetal and child development. At present, whether APAP causes cytotoxic effects in human stem cells remains to be elucidated, therefore, the present study aimed to investigate the cellular effects of APAP treatment in human stem cells. The results of the present study revealed that high-dose APAP induced more marked cytotoxic effects in human mesenchymal stem cells (hMSCs) than in renal tubular cells. In addition, increased levels of hydrogen peroxide (H2O2), phosphorylation of c-Jun N-terminal kinase and p38, and activation of caspase-9/-3 cascade were observed in the APAP-treated hMSCs. By contrast, antioxidants, including vitamin C reduced APAP-induced augmentations in H2O2 levels, but did not inhibit the APAP-induced cytotoxic effects in the hMSCs. These results suggested that high doses of APAP may cause serious damage towards hMSCs.
Collapse
Affiliation(s)
- Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Yung-Lung Yu
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Ko-Ting Lin
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Shalu, Taichung 433, Taiwan, R.O.C
| | - Jen-Ni Chen
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Shalu, Taichung 433, Taiwan, R.O.C
| | - Wei-Jung Chang
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Chyou-Wei Wei
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Shalu, Taichung 433, Taiwan, R.O.C
| |
Collapse
|
37
|
Jiang Y, Fan X, Wang Y, Chen P, Zeng H, Tan H, Gonzalez FJ, Huang M, Bi H. Schisandrol B protects against acetaminophen-induced hepatotoxicity by inhibition of CYP-mediated bioactivation and regulation of liver regeneration. Toxicol Sci 2014; 143:107-15. [PMID: 25319358 DOI: 10.1093/toxsci/kfu216] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (APAP) overdose is the most frequent cause of drug-induced acute liver failure. Schisandra sphenanthera is a traditional hepato-protective Chinese medicine and Schisandrol B (SolB) is one of its major active constituents. In this study, the protective effect of SolB against APAP-induced acute hepatotoxicity in mice and the involved mechanisms were investigated. Morphological and biochemical assessments clearly demonstrated a protective effect of SolB against APAP-induced liver injury. SolB pretreatment significantly attenuated the increases in alanine aminotransferase and aspartate aminotransferase activity, and prevented elevated hepatic malondialdehyde formation and the depletion of mitochondrial glutathione (GSH) in a dose-dependent manner. SolB also dramatically altered APAP metabolic activation by inhibiting the activities of CYP2E1 and CYP3A11, which was evidenced by significant inhibition of the formation of the oxidized APAP metabolite NAPQI-GSH. A molecular docking model also predicted that SolB had potential to interact with the CYP2E1 and CYP3A4 active sites. In addition, SolB abrogated APAP-induced activation of p53 and p21, and increased expression of liver regeneration and antiapoptotic-related proteins such as cyclin D1 (CCND1), PCNA, and BCL-2. This study demonstrated that SolB exhibited a significant protective effect toward APAP-induced liver injury, potentially through inhibition of CYP-mediated APAP bioactivation and regulation of the p53, p21, CCND1, PCNA, and BCL-2 to promote liver regeneration.
Collapse
Affiliation(s)
- Yiming Jiang
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xiaomei Fan
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ying Wang
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Pan Chen
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Hang Zeng
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Huasen Tan
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Frank J Gonzalez
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Min Huang
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Huichang Bi
- *School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
38
|
Mobasher MA, de Toro-Martín J, González-Rodríguez Á, Ramos S, Letzig LG, James LP, Muntané J, Álvarez C, Valverde ÁM. Essential role of protein-tyrosine phosphatase 1B in the modulation of insulin signaling by acetaminophen in hepatocytes. J Biol Chem 2014; 289:29406-19. [PMID: 25204659 DOI: 10.1074/jbc.m113.539189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many drugs are associated with the development of glucose intolerance or deterioration in glycemic control in patients with pre-existing diabetes. We have evaluated the cross-talk between signaling pathways activated by acetaminophen (APAP) and insulin signaling in hepatocytes with or without expression of the protein-tyrosine phosphatase 1B (PTP1B) and in wild-type and PTP1B-deficient mice chronically treated with APAP. Human primary hepatocytes, Huh7 hepatoma cells with silenced PTP1B, mouse hepatocytes from wild-type and PTP1B-deficient mice, and a mouse model of chronic APAP treatment were used to examine the mechanisms involving PTP1B in the effects of APAP on glucose homeostasis and hepatic insulin signaling. In APAP-treated human hepatocytes at concentrations that did not induce death, phosphorylation of JNK and PTP1B expression and enzymatic activity were increased. APAP pretreatment inhibited activation of the early steps of insulin signaling and decreased Akt phosphorylation. The effects of APAP in insulin signaling were prevented by suramin, a PTP1B inhibitor, or rosiglitazone that decreased PTP1B levels. Likewise, PTP1B deficiency in human or mouse hepatocytes protected against APAP-mediated impairment in insulin signaling. These signaling pathways were modulated in mice with chronic APAP treatment, resulting in protection against APAP-mediated hepatic insulin resistance and alterations in islet alpha/beta cell ratio in PTP1B(-/-) mice. Our results demonstrate negative cross-talk between signaling pathways triggered by APAP and insulin signaling in hepatocytes, which is in part mediated by PTP1B. Moreover, our in vivo data suggest that chronic use of APAP may be associated with insulin resistance in the liver.
Collapse
Affiliation(s)
- Maysa Ahmed Mobasher
- From the Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain, the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salid Carlos III, 28029 Madrid
| | - Juan de Toro-Martín
- the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salid Carlos III, 28029 Madrid, the Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense de Madrid, 28034 Madrid, Spain
| | - Águeda González-Rodríguez
- From the Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain, the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salid Carlos III, 28029 Madrid
| | - Sonia Ramos
- the Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN-CSIC), 28040 Madrid, Spain
| | - Lynda G Letzig
- the Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, and
| | - Laura P James
- the Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, and
| | - Jordi Muntané
- the Department of General Surgery, Institute of Biomedicine of Seville, Hospital Universitary "Virgen del Rocío"/CSIC/University of Seville, 41013 Seville, Spain
| | - Carmen Álvarez
- the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salid Carlos III, 28029 Madrid, the Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense de Madrid, 28034 Madrid, Spain
| | - Ángela M Valverde
- From the Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain, the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salid Carlos III, 28029 Madrid,
| |
Collapse
|
39
|
Truong VL, Bak MJ, Jun M, Kong ANT, Ho CT, Jeong WS. Antioxidant defense and hepatoprotection by procyanidins from almond (Prunus amygdalus) skins. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:8668-8678. [PMID: 25119859 DOI: 10.1021/jf5027247] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
UNLABELLED Procyanidins, polymeric flavan-3-ols, are known to possess antioxidant, antiatherogenic, and anticarcinogenic properties. In the present study, we investigated the role of almond (Prunus amygdalus) skin procyanidins (ASP) in regulating the protein expression of phase II detoxifying and antioxidant enzymes in HepG2 cells and acetaminophen (APAP)-treated hepatotoxic mice. Treatments of ASP significantly induced the expression of phase II enzymes including NAD(P)H quinoneoxidoreductase 1, catalase, glutathione peroxidase, and superoxide dismutase in the cells and mice. ASP also potently enhanced the expression of nuclear factor-E2-related factor 2 (Nrf2) and antioxidant response element (ARE)-reporter gene activity in vitro. APAP-induced hepatotoxic markers including AST and ALT in mice were inhibited by ASP administration. However, regulation of upstream kinases by ASP was different between in vitro and in vivo models. Collectively, ASP could induce the activation of Nrf2/ARE-mediated phase II detoxifying/antioxidant enzymes but with differential regulation on upstream kinases between in vitro and in vivo.
Collapse
Affiliation(s)
- Van-Long Truong
- Department of Smart Foods and Drugs, Department of Food and Life Sciences, College of Biomedical Science and Engineering, Inje University , Gimhae 621-749, Korea
| | | | | | | | | | | |
Collapse
|
40
|
Yu YL, Yiang GT, Chou PL, Tseng HH, Wu TK, Hung YT, Lin PS, Lin SY, Liu HC, Chang WJ, Wei CW. Dual role of acetaminophen in promoting hepatoma cell apoptosis and kidney fibroblast proliferation. Mol Med Rep 2014; 9:2077-84. [PMID: 24682227 PMCID: PMC4055434 DOI: 10.3892/mmr.2014.2085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/28/2014] [Indexed: 12/19/2022] Open
Abstract
Acetaminophen (APAP), is a safe analgesic and antipyretic drug at therapeutic dose, and is widely used in the clinic. However, high doses of APAP can induce hepatotoxicity and nephrotoxicity. Most studies have focused on high‑dose APAP‑induced acute liver and kidney injury. So far, few studies have investigated the effects of the therapeutic dose (1/10 of the high dose) or of the low dose (1/100 of the high dose) of APAP on the cells. The aim of this study was to investigate the cellular effects of therapeutic- or low‑dose APAP treatment on hepatoma cells and kidney fibroblasts. As expected, high‑dose APAP treatment inhibited while therapeutic and low‑dose treatment did not inhibit cell survival of kidney tubular epithelial cells. In addition, therapeutic-dose treatment induced an increase in the H2O2 level, activated the caspase‑9/‑3 cascade, and induced cell apoptosis of hepatoma cells. Notably, APAP promoted fibroblast proliferation, even at low doses. This study demonstrates that different cellular effects are exerted upon treatment with different APAP concentrations. Our results indicate that treatment with the therapeutic dose of APAP may exert an antitumor activity on hepatoma, while low‑dose treatment may be harmful for patients with fibrosis, since it may cause proliferation of fibroblasts.
Collapse
Affiliation(s)
- Yung-Luen Yu
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Pei-Lun Chou
- Division of Allergy‑Immunology‑Rheumatology, Department of Internal Medicine, Saint Mary's Hospital Luodong, Yilan 265, Taiwan, R.O.C
| | - Hsu-Hung Tseng
- Division of General Surgery, Taichung Hospital, Ministry of Health and Welfare, Taichung 403, Taiwan, R.O.C
| | - Tsai-Kun Wu
- 2The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Yu-Ting Hung
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Pei-Shiuan Lin
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Shu-Yu Lin
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Hsiao-Chun Liu
- Department of Nursing, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Wei-Jung Chang
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Chyou-Wei Wei
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| |
Collapse
|
41
|
Sugiyama A, Sun J. Immunochemical detection of lipid hydroperoxide- and aldehyde-modified proteins in diseases. Subcell Biochem 2014; 77:115-25. [PMID: 24374923 DOI: 10.1007/978-94-007-7920-4_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Polyunsaturated fatty acid (PUFA) is easily peroxidized by free radicals and enzymes. When this occurs, it results in the compromised integrity of cellular membranes and leads to lipid hydroperoxide as a major reaction product, which is decomposed into aldehyde. Lipid hydroperoxide-modified lysine is known to be an early product of the lipid peroxidation process, suggesting that it might be a PUFA-oxidative stress marker during the initial stage of oxidative stress. Lipid hydroperoxides cause or enhance ROS-mediated DNA fragmentation. The α,β-unsaturated aldehydes are end products of PUFA peroxidation. They are highly reactive and readily attack and modify the protein amino acid residues into aldehyde-modified proteins. Lipid peroxidation-derived α,β-unsaturated aldehydes are capable of inducing cellular stress-responsive processes such as cell signaling and apoptosis. The lipid hydroperoxide- and aldehyde-modified proteins have been immunohistochemically detected in diverse pathological situations such as atherosclerosis, Alzheimer's disease, Parkinson's disease, and chemical material-induced liver injury and renal tubular injury in humans and experimental animals. These findings suggest that the expression of the lipid hydroperoxide- and aldehyde-modified proteins is closely associated with the pathogenesis of these diseases in humans and experimental animals.
Collapse
Affiliation(s)
- Akihiko Sugiyama
- Course of Veterinary Laboratory Medicine, School of Veterinary Medicine, Faculty of Agriculture, Tottori University, Minami 4-101 m Koyama-cho, Tottori, 680-8553, Japan,
| | | |
Collapse
|
42
|
Schunke KJ, Coyle L, Merrill GF, Denhardt DT. Acetaminophen attenuates doxorubicin-induced cardiac fibrosis via osteopontin and GATA4 regulation: reduction of oxidant levels. J Cell Physiol 2013; 228:2006-14. [PMID: 23526585 PMCID: PMC3739938 DOI: 10.1002/jcp.24367] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/14/2013] [Indexed: 01/07/2023]
Abstract
It is well documented in animal and human studies that therapy with the anti-cancer drug doxorubicin (DOX) induces fibrosis, cardiac dysfunction, and cell death. The most widely accepted mechanism of cardiac injury is through production of reactive oxygen species (ROS), which cause mitochondrial damage, sarcomere structural alterations, and altered gene expression in myocytes and fibroblasts. Here we investigated the effects of acetaminophen (APAP, N-acetyl-para-aminophenol) on DOX-induced cardiac injury and fibrosis in the presence or absence of osteopontin (OPN). H9c2 rat heart-derived embryonic myoblasts were exposed to increasing concentrations of DOX ± APAP; cell viability, oxidative stress, and OPN transcript levels were analyzed. We found a dose-dependent decrease in cell viability and a corresponding increase in intracellular oxidants at the tested concentrations of DOX. These effects were attenuated in the presence of APAP. RT-PCR analysis revealed a small increase in OPN transcript levels in response to DOX, which was suppressed by APAP. When male 10-12-week-old mice (OPN(+/+) or OPN(-/-)) were given weekly injections of DOX ± APAP for 4 weeks there was substantial cardiac fibrosis in OPN(+/+) and, to a lesser extent, in OPN(-/-) mice. In both groups, APAP decreased fibrosis to near baseline levels. Activity of the pro-survival GATA4 transcription factor was diminished by DOX in both mouse genotypes, but retained baseline activity in the presence of APAP. These effects were mediated, in part, by the ability of APAP, acting as an anti-inflammatory agent, to decrease intracellular ROS levels, consequently diminishing the injury-induced increase in OPN levels.
Collapse
Affiliation(s)
- Kathryn J Schunke
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Acetaminophen (APAP) is the leading worldwide cause of drug overdose and acute liver failure (ALF). Single overdose ingestion and therapeutic misadventure may cause hepatotoxicity. Several factors, such as concomitant alcohol use or abuse, concurrent medications, genetic factors, and nutritional status, can influence the susceptibility and severity of APAP hepatotoxicity. Early manifestations of APAP hepatotoxicity are nonspecific, but require prompt recognition by physicians. Patients with repeated overdose tend to present late, and in such hepatotoxicity may have already evolved. N-acetylcysteine is a very effective antidote when giving within 8 hours, and is also recommended after a presentation of hepatotoxicity and ALF. The prognosis of patients with APAP-induced ALF is better than other causes of ALF. Liver transplantation should be offered to those who are unlikely to survive.
Collapse
Affiliation(s)
- Chalermrat Bunchorntavakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania, 2 Dulles, 3400 Spruce Street, Philadelphia, PA 19104, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Rajavithi Hospital, College of Medicine, Rangsit University, Rajavithi Road, Ratchathewi, Bangkok 10400, Thailand
| | | |
Collapse
|
44
|
Acetaminophen-induced acute liver failure cured by integrative therapy of chinese and western medicine—a case report. Chin J Integr Med 2013; 19:712-5. [DOI: 10.1007/s11655-013-1570-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Indexed: 02/05/2023]
|
45
|
Park JH, Seo KS, Tadi S, Ahn BH, Lee JU, Heo JY, Han J, Song MS, Kim SH, Yim YH, Choi HS, Shong M, Kweon G. An indole derivative protects against acetaminophen-induced liver injury by directly binding to N-acetyl-p-benzoquinone imine in mice. Antioxid Redox Signal 2013; 18:1713-22. [PMID: 23121402 PMCID: PMC3619205 DOI: 10.1089/ars.2012.4677] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
AIMS Acetaminophen (APAP)-induced liver injury is mainly due to the excessive formation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) through the formation of a reactive intermediate, N-acetyl-p-benzoquinone imine (NAPQI), in both humans and rodents. Here, we show that the indole-derived synthetic compound has a protective effect against APAP-induced liver injury in C57Bl/6 mice model. RESULTS NecroX-7 decreased tert-butylhydroperoxide (t-BHP)- and APAP-induced cell death and ROS/RNS formation in HepG2 human hepatocarcinoma and primary mouse hepatocytes. In mice, NecroX-7 decreased APAP-induced phosphorylation of c-Jun N-terminal kinase (JNK) and 3-nitrotyrosine (3-NT) formation, and also protected mice from APAP-induced liver injury and lethality by binding directly to NAPQI. The binding of NecroX-7 to NAPQI did not require any of cofactors or proteins. NecroX-7 could only scavenge NAPQI when hepatocellular GSH levels were very low. INNOVATION NecroX-7 is an indole-derived potent antioxidant molecule, which can be bound to some types of radicals and especially NAPQI. It is well known that the NAPQI is a major intermediate of APAP, which causes necrosis of hepatocytes in rodents and humans. Thus, blocking NAPQI formation or eliminating NAPQI are novel strategies for the treatment or prevention of APAP-induced liver injury instead of GSH replenishment. CONCLUSION Our data suggest that the indole-derivative, NecroX-7, directly binds to NAPQI when hepatic GSH levels are very low and the NAPQI-NecroX-7 complex is secreted to the blood from the liver. NecroX-7 shows more preventive and similar therapeutic effects against APAP-induced liver injury when compared to the effect of N-acetylcysteine in C57Bl/6 mice.
Collapse
Affiliation(s)
- Ji-Hoon Park
- Department of Biochemistry, School of Medicine, Chungnam National University, Daejeon, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Protein tyrosine phosphatase 1B modulates GSK3β/Nrf2 and IGFIR signaling pathways in acetaminophen-induced hepatotoxicity. Cell Death Dis 2013; 4:e626. [PMID: 23661004 PMCID: PMC3674359 DOI: 10.1038/cddis.2013.150] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute hepatic failure secondary to acetaminophen (APAP) poisoning is associated with high mortality. Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of tyrosine kinase growth factor signaling. In the liver, this pathway confers protection against injury. However, the involvement of PTP1B in the intracellular networks activated by APAP is unknown. We have assessed PTP1B expression in APAP-induced liver failure in humans and its role in the molecular mechanisms that regulate the balance between cell death and survival in human and mouse hepatocytes, as well as in a mouse model of APAP-induced hepatotoxicity. PTP1B expression was increased in human liver tissue removed during liver transplant from patients for APAP overdose. PTP1B was upregulated by APAP in primary human and mouse hepatocytes together with the activation of c-jun (NH2) terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK), resulting in cell death. Conversely, Akt phosphorylation and the antiapoptotic Bcl2 family members BclxL and Mcl1 were decreased. PTP1B deficiency in mouse protects hepatocytes against APAP-induced cell death, preventing glutathione depletion, reactive oxygen species (ROS) generation and activation of JNK and p38 MAPK. APAP-treated PTP1B−/− hepatocytes showed enhanced antioxidant defense through the glycogen synthase kinase 3 (GSK3)β/Src kinase family (SKF) axis, delaying tyrosine phosphorylation of the transcription factor nuclear factor-erythroid 2-related factor (Nrf2) and its nuclear exclusion, ubiquitination and degradation. Insulin-like growth factor-I receptor-mediated signaling decreased in APAP-treated wild-type hepatocytes, but was maintained in PTP1B−/− cells or in wild-type hepatocytes with reduced PTP1B levels by RNA interference. Likewise, both signaling cascades were modulated in mice, resulting in less severe APAP hepatotoxicity in PTP1B−/− mice. Our results demonstrated that PTP1B is a central player of the mechanisms triggered by APAP in hepatotoxicity, suggesting a novel therapeutic target against APAP-induced liver failure.
Collapse
|
47
|
Wang K, Yuan Y, Li H, Cho JH, Huang D, Gray L, Qin S, Galas DJ. The spectrum of circulating RNA: a window into systems toxicology. Toxicol Sci 2013; 132:478-92. [PMID: 23358195 DOI: 10.1093/toxsci/kft014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Adverse effects caused by therapeutic drugs are a serious and costly health concern. Despite the body's systemic responses to therapeutics, the liver is often the focus of damage and is usually the focus of studies of toxic effects due to its active roles in the metabolism of xenobiotics. It is extremely difficult, however, to assess systemic responses with currently available methods. Comprehensive cataloging of cell-free circulating RNAs using next-generation sequencing technology may open a window to assess drug-associated adverse effects at the systems level. To explore this potential, we conducted an RNA profiling study using the well-characterized acetaminophen overdose mouse model on liver and plasma with microarray and next-generation sequencing platforms, respectively. After drug treatment, the levels of a number of transcripts, both endogenous and exogenous RNAs, showed significant changes in plasma, reflecting not only the classical liver injury induced by acetaminophen overdose but also damage in tissues other than the liver. The changes in exogenous RNAs also reflect alteration on dieting behavior after acetaminophen overdose. Besides reporting an extensive list of circulating RNA-based biomarker candidates, this study illustrates the possibility of using circulating RNAs to assess global effects of therapeutics. This could also lead to a new approach for a more comprehensive assessment of the efficacy and safety of therapeutics.
Collapse
Affiliation(s)
- Kai Wang
- Institute for Systems Biology, Seattle, Washington 98109, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Badmann A, Langsch S, Keogh A, Brunner T, Kaufmann T, Corazza N. TRAIL enhances paracetamol-induced liver sinusoidal endothelial cell death in a Bim- and Bid-dependent manner. Cell Death Dis 2012; 3:e447. [PMID: 23254290 PMCID: PMC3542621 DOI: 10.1038/cddis.2012.185] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Paracetamol (acetaminophen, APAP) is a universally used analgesic and antipyretic agent. Considered safe at therapeutic doses, overdoses cause acute liver damage characterized by centrilobular hepatic necrosis. One of the major clinical problems of paracetamol-induced liver disease is the development of hemorrhagic alterations. Although hepatocytes represent the main target of the cytotoxic effect of paracetamol overdose, perturbations within the endothelium involving morphological changes of liver sinusoidal endothelial cells (LSECs) have also been described in paracetamol-induced liver disease. Recently, we have shown that paracetamol-induced liver damage is synergistically enhanced by the TRAIL signaling pathway. As LSECs are constantly exposed to activated immune cells expressing death ligands, including TRAIL, we investigated the effect of TRAIL on paracetamol-induced LSEC death. We here demonstrate for the first time that TRAIL strongly enhances paracetamol-mediated LSEC death with typical features of apoptosis. Inhibition of caspases using specific inhibitors resulted in a strong reduction of cell death. TRAIL appears to enhance paracetamol-induced LSEC death via the activation of the pro-apoptotic BH3-only proteins Bid and Bim, which initiate the mitochondrial apoptotic pathway. Taken together this study shows that the liver endothelial layer, mainly LSECs, represent a direct target of the cytotoxic effect of paracetamol and that activation of TRAIL receptor synergistically enhances paracetamol-induced LSEC death via the mitochondrial apoptotic pathway. TRAIL-mediated acceleration of paracetamol-induced cell death may thus contribute to the pathogenesis of paracetamol-induced liver damage.
Collapse
Affiliation(s)
- A Badmann
- Division of Immunopathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
49
|
Rodrigues D, Silveira TRD, Matte U. Freshly isolated hepatocyte transplantation in acetaminophen-induced hepatotoxicity model in rats. ARQUIVOS DE GASTROENTEROLOGIA 2012; 49:291-5. [DOI: 10.1590/s0004-28032012000400011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 08/24/2012] [Indexed: 11/22/2022]
Abstract
CONTEXT: Hepatocyte transplantation is an attractive therapeutic modality for liver disease as an alternative for orthotopic liver transplantation. OBJECTIVE: The aim of the current study was to investigate the feasibility of freshly isolated rat hepatocyte transplantation in acetaminophen-induced hepatotoxicity model. METHODS: Hepatocytes were isolated from male Wistar rats and transplanted 24 hours after acetaminophen administration in female recipients. Female rats received either 1x10(7) hepatocytes or phosphate buffered saline through the portal vein or into the spleen and were sacrificed after 48 hours. RESULTS: Alanine aminotransferase levels measured within the experiment did not differ between groups at any time point. Molecular analysis and histology showed presence of hepatocytes in liver of transplanted animals injected either through portal vein or spleen. CONCLUSION: These data demonstrate the feasibility and efficacy of hepatocyte transplantation in the liver or spleen in a mild acetaminophen-induced hepatotoxicity model.
Collapse
Affiliation(s)
- Daniela Rodrigues
- Hospital de Clínicas de Porto Alegre; Universidade Federal do Rio Grande do Sul
| | | | - Ursula Matte
- UFRGS; UFRGS; Hospital de Clínicas de Porto Alegre, Brazil
| |
Collapse
|
50
|
Kjartansdottir I, Bergmann OM, Arnadottir RS, Björnsson ES. Paracetamol intoxications: a retrospective population-based study in Iceland. Scand J Gastroenterol 2012; 47:1344-52. [PMID: 22827594 DOI: 10.3109/00365521.2012.703236] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Paracetamol is the most common cause of acute liver failure (ALF) in many countries. Much data on paracetamol toxicity originate from liver transplant centers and tertiary referral institutions. The authors analyzed the population-based annual incidence of paracetamol overdoses and ALF, and described the risk factors for hepatotoxicity. METHODS A search was undertaken for the diagnosis of paracetamol overdoses in the diagnoses registry of the National University Hospital of Iceland from 2004 to 2009 serving a population of 219,249 inhabitants. Relevant information was collected from medical records. RESULTS A total of 1913 drug-related poisoning episodes were identified and reviewed, 352 (18%) involved paracetamol overdoses. The annual incidence of paracetamol overdoses declined from 30.0 (2004) to 16.0/100,000 per year (2009) (p < 0.05). The female/male ratio was 3.0 and the largest age group was 16-25 years. After the initial examination, 26% were discharged home. Hospitalized index visits were 182 with accidental overdoses constituting 16 (9%) with no gender difference. Compared with intentional overdose the accidental group had higher aminotransferases (p < 0.005). ALF occurred in 3.8% (7/182) of the index visits and the incidence was 0.7/100,000 per year. In the intentional group, 1.2% (2/163) developed ALF versus 25% (4/16) of the accidental group (p = 0.001). Only one patient died from ALF and none underwent liver transplantation. CONCLUSION The annual incidence of paracetamol overdoses was high in this population-based study but declined. Young females with intentional overdose accounted for most of the cases, whereas accidental overdoses were more common in older patients. The occurrence of ALF was low and mostly associated with accidental overdose.
Collapse
|