1
|
Delaporte F, Roger E, Bejaud J, Loyer P, Lagarce F, Savary CC. Internalization and mechanisms of toxicity of lipid nanocapsules in HepG2 and HepaRG hepatoma cells upon acute and chronic exposures. Int J Pharm 2024; 667:124815. [PMID: 39424085 DOI: 10.1016/j.ijpharm.2024.124815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Lipid nanocapsules (LNCs) used as nanomedicine have been developed to enhance pharmacokinetics and decrease side effects of drugs, particularly for cancer therapies. After intravenous administration, LNCs possess an important hepatic tropism however, few data exist about their toxicity and even less after repeated exposure. This study aimed to assess the in vitro toxicity and internalization of unloaded LNCs, of 50 and 100 nm size, on HepG2 and HepaRG liver cell lines. Internalization of the 50 nm LNCs was slower compared to the 100 nm LNCs and both LNCs exhibited a higher toxicity on cancerous HepG2 cells compared to differentiated HepaRG cells. LNCs were mainly internalized via caveolin-mediated endocytosis in both cell lines. Upon chronic exposure, the toxicity of LNCs on HepaRG cells increased, although the pathways of internalization remained unchanged. Cell death studies have demonstrated an involvement of ferroptosis, but not of apoptosis. After acute and repeated exposures on HepaRG cells, the 100 nm LNCs showed a good safety profile. Finally, LNCs induced a more significant toxicity associated with faster internalization in the HepG2 cancerous model than in the differentiated HepaRG cells. This provides good evidence for LNCs to potentialize the cytotoxic effects of an active drug on liver cancer cells.
Collapse
Affiliation(s)
- Flavien Delaporte
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France; CHU Angers, 4 rue Larrey, 49033 Angers, France.
| | - Emilie Roger
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Jérome Bejaud
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Pascal Loyer
- Inserm, University of Rennes, INRAE, NuMeCan Institute (Nutrition, Metabolisms and Cancer), Rennes, France
| | - Frédéric Lagarce
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France; CHU Angers, 4 rue Larrey, 49033 Angers, France
| | - Camille C Savary
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
2
|
Lim JC, Jiang L, Lust NG, Donaldson PJ. Minimizing Oxidative Stress in the Lens: Alternative Measures for Elevating Glutathione in the Lens to Protect against Cataract. Antioxidants (Basel) 2024; 13:1193. [PMID: 39456447 PMCID: PMC11505578 DOI: 10.3390/antiox13101193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Oxidative stress plays a major role in the formation of the cataract that is the result of advancing age, diabetes or which follows vitrectomy surgery. Glutathione (GSH) is the principal antioxidant in the lens, and so supplementation with GSH would seem like an intuitive strategy to counteract oxidative stress there. However, the delivery of glutathione to the lens is fraught with difficulties, including the limited bioavailability of GSH caused by its rapid degradation, anatomical barriers of the anterior eye that result in insufficient delivery of GSH to the lens, and intracellular barriers within the lens that limit delivery of GSH to its different regions. Hence, more attention should be focused on alternative methods by which to enhance GSH levels in the lens. In this review, we focus on the following three strategies, which utilize the natural molecular machinery of the lens to enhance GSH and/or antioxidant potential in its different regions: the NRF2 pathway, which regulates the transcription of genes involved in GSH homeostasis; the use of lipid permeable cysteine-based analogues to increase the availability of cysteine for GSH synthesis; and the upregulation of the lens's internal microcirculation system, which is a circulating current of Na+ ions that drives water transport in the lens and with it the potential delivery of cysteine or GSH. The first two strategies have the potential to restore GSH levels in the epithelium and cortex, while the ability to harness the lens's internal microcirculation system offers the exciting potential to deliver and elevate antioxidant levels in its nucleus. This is an important distinction, as the damage phenotypes for age-related (nuclear) and diabetic (cortical) cataract indicate that antioxidant delivery must be targeted to different regions of the lens in order to alleviate oxidative stress. Given our increasing aging and diabetic populations it has become increasingly important to consider how the natural machinery of the lens can be utilized to restore GSH levels in its different regions and to afford protection from cataract.
Collapse
Affiliation(s)
- Julie C. Lim
- Department Physiology, University of Auckland, Auckland 1023, New Zealand; (L.J.); (N.G.L.); (P.J.D.)
- Aotearoa New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| | - Lanpeng Jiang
- Department Physiology, University of Auckland, Auckland 1023, New Zealand; (L.J.); (N.G.L.); (P.J.D.)
- Aotearoa New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| | - Natasha G. Lust
- Department Physiology, University of Auckland, Auckland 1023, New Zealand; (L.J.); (N.G.L.); (P.J.D.)
- Aotearoa New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| | - Paul J. Donaldson
- Department Physiology, University of Auckland, Auckland 1023, New Zealand; (L.J.); (N.G.L.); (P.J.D.)
- Aotearoa New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
3
|
Rodrigues K, Hussain R, Cooke S, Zhang G, Zhang D, Yin L, Tong X. Fructose as a novel nutraceutical for acetaminophen (APAP)-induced hepatotoxicity. METABOLISM AND TARGET ORGAN DAMAGE 2023; 3:20. [PMID: 39193224 PMCID: PMC11349303 DOI: 10.20517/mtod.2023.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Acetaminophen (APAP) is the most widely used analgesic in the world. APAP overdose can cause severe hepatotoxicity and therefore is the most common cause of drug-induced liver injury. The only approved treatment for APAP overdose is N-acetyl-cysteine (NAC) supplementation. However, the narrow efficacy window of the drug severely limits its clinical use, prompting the search for other therapeutic options to counteract APAP toxicity. Recent research has pointed to fructose as a novel nutraceutical for APAP-induced liver injury. This review summarizes the current understanding of the molecular mechanisms underlying APAP-induced liver injury, introduces how fructose supplementation could prevent and treat APAP liver toxicity with a focus on the ChREBPα-FGF21 pathway, and proposes possible future directions of study.
Collapse
Affiliation(s)
- Kyle Rodrigues
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Rawdat Hussain
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Sarah Cooke
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Gary Zhang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Deqiang Zhang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Lei Yin
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Xin Tong
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
4
|
Li J, Lu Q, Peng M, Liao J, Zhang B, Yang D, Huang P, Yang Y, Zhao Q, Han B, Li J. Water extract from Herpetospermum pedunculosum attenuates oxidative stress and ferroptosis induced by acetaminophen via regulating Nrf2 and NF-κB pathways. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116069. [PMID: 36572326 DOI: 10.1016/j.jep.2022.116069] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/22/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The seeds of Herpetospermum pedunculosum seeds is a traditional Tibetan medicine possessing hepatoprotective effect, but their protective effect on APAP-induced liver injury has not yet been explored. AIM OF THE STUDY This study aimed at exploring the protective effect and mechanism of the water extract from the seeds of Herpetospermum pedunculosum (HPWE) on APAP-induced liver injury in vitro and in vivo. MATERIALS AND METHODS In vitro and in vivo models of liver injury were established by APAP treatment of BRL-3A cells or mice. The effect and mechanism of action of HPWE were explored by using cell viability assay, ELISA, immunofluorescence assay, RT-qPCR, histological observation and immunohistochemistry staining, western blotting and high-content imaging system. RESULTS In vitro experiments showed that HPWE treatment significantly promoted the cell viability, decreased ALT/AST level, and inhibited the ROS accumulation induced by APAP. Furthermore, HPWE and Fer-1 alleviated erastin-induced cell ferroptosis, upregulated GPX4 and SLC7A11 expression, and reduced lipid peroxides production. Further study showed that APAP could also downregulate the expression of GPX4 and SLC7A11, causing cell ferroptosis, and HPWE and Fer-1 counteracted this process. Our in vivo experiments showed that pretreatment with HPWE in APAP-treated mice significantly alleviated the serum ALT/AST level, decreased necrotic cells and inflammatory cell infiltration, upregulated the expression of GPX4 and SLC7A11. Further, it was demonstrated that HPWE treatment downregulated Nrf2 and its downstream target genes, i.e. HO-1 and NQO1 expression at the mRNA and protein levels. HPWE treatment also inhibited the activation of NF-κB p65 and downregulated its target genes, i.e. TNF-α and IL-1β, expression. CONCLUSION The present study showed that HPWE could relieve oxidative stress and ferroptosis via activating Nrf2 signaling pathway and inhibiting NF-κB mediated pathway.
Collapse
Affiliation(s)
- Jintao Li
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Pharmacy, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Qiuxia Lu
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Meihao Peng
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Pharmacy, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Jiaqing Liao
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Pharmacy, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Bowen Zhang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Di Yang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Peng Huang
- Tibet Rhodiola Pharmaceutical Holding Company, Lhasa, Tibet, 850000, China
| | - Yixi Yang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Qi Zhao
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jian Li
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu, 610106, China; School of Basic Medical Sciences, Chengdu University, Chengdu, 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu, 610106, China.
| |
Collapse
|
5
|
Zhang B, Yu L, Zhu R, Wei X, Fan X, Hu H, Yang D, Du H, Zhao M, Li L, Oh Y, Feng Y, Gu N. Malting barley carbon dots-mediated oxidative stress promotes insulin resistance in mice via NF-κB pathway and MAPK cascade. J Nanobiotechnology 2022; 20:331. [PMID: 35842638 PMCID: PMC9288084 DOI: 10.1186/s12951-022-01543-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022] Open
Abstract
Background Food-borne carbon dots (CDs) are widely generated during food processing and are inevitably ingested by humans causing toxicity. However, the toxic effects of food-borne CDs on the blood glucose metabolism are unknown. Results In this study, we brewed beer via a representative strategy and extracted the melting-barley CDs (MBCDs) to explore the toxic effects on blood glucose in mice. We found the accumulation of fluorescent labeled MBCDs in various organs and oral administration of MBCDs can cause visceral toxicity, manifested as liver damage. Mice were orally administered MBCDs (5 and 25 mg/kg) for 16 weeks, and increased levels of fasting blood glucose were observed in both MBCDs-treated groups. Transcriptomic analyses revealed that MBCDs activate oxidative stress, inflammatory responses, the MAPK cascade, and PI3K/Akt signaling in mice livers. Mechanistically, MBCDs exposure-induced reactive oxygen species (ROS) overproduction activates the nuclear factor-κB (NF-κB) signaling pathway and MAPK cascade, thereby promoting phosphorylated insulin receptor substrate (IRS)-1 at Ser307 and inducing insulin resistance (IR). Meanwhile, the IR promoted gluconeogenesis, which enhanced MBCDs-induced hyperglycemia of mice. Importantly, inhibition of the ROS significantly attenuated the MBCDs-induced inflammatory response and MAPK cascade, thereby alleviating IR and hyperglycemia in mice. Conclusion In summary, this study revealed that MBCDs promote ROS overproduction and thus induced IR, resulting in imbalance of glucose homeostasis in mice. More importantly, this study was further assessed to reveal an imperative emphasis on the reevaluation of dietary and environmental CDs exposure, and has important implications for T2DM prevention research. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01543-1.
Collapse
Affiliation(s)
- Boya Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.,State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, China
| | - Lidong Yu
- School of Physics, Harbin Institute of Technology, Harbin, 150001, China
| | - Ruijiao Zhu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xiangjuan Wei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xingpei Fan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Hailong Hu
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19019, USA
| | - Daqian Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Haining Du
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Meimei Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Li Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yuri Oh
- Faculty of Education, Wakayama University, Wakayama, Japan
| | - Yujie Feng
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, China
| | - Ning Gu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China. .,State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, China.
| |
Collapse
|
6
|
Cao YJ, Huang ZR, You SZ, Guo WL, Zhang F, Liu B, Lv XC, Lin ZX, Liu PH. The Protective Effects of Ganoderic Acids from Ganoderma lucidum Fruiting Body on Alcoholic Liver Injury and Intestinal Microflora Disturbance in Mice with Excessive Alcohol Intake. Foods 2022; 11:949. [PMID: 35407036 PMCID: PMC8997615 DOI: 10.3390/foods11070949] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/09/2022] [Accepted: 03/18/2022] [Indexed: 02/05/2023] Open
Abstract
This study aimed to investigate the protective effects of ganoderic acids (GA) from Ganoderma lucidum against liver injury and intestinal microbial disorder in mice with excessive alcohol intake. Results showed GA supplement significantly inhibited the abnormal elevation of the liver index, serum lipid parameters, aspartate aminotransferase and alanine aminotransferase in mice exposed to alcohol intake, and also significantly protected the excessive lipid accumulation and pathological changes. Alcohol-induced oxidative stress in the liver was significantly ameliorated by GA intervention through reducing the levels of maleic dialdehyde and lactate dehydrogenase and increasing the levels of glutathione, catalase, superoxide dismutase and alcohol dehydrogenase. Intestinal microbiota profiling demonstrated GA intervention modulated the composition of intestinal microflora by increasing the levels of Lactobacillus, Faecalibaculum, Romboutsia, Bifidobacterium and decreasing the Helicobacter level. Furthermore, liver metabolomic profiling suggested GA intervention had a remarkable regulatory effect on liver metabolism with excessive alcohol consumption. Moreover, GA intervention regulated mRNA levels of alcohol metabolism, fatty lipid metabolism, oxidative stress, bile acid biosynthesis and metabolism-related genes in the liver. Conclusively, these findings demonstrate GA intervention can significantly relieve alcoholic liver injury and it is hopeful to become a new functional food ingredient for the prevention of alcoholic liver injury.
Collapse
Affiliation(s)
- Ying-Jia Cao
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.-J.C.); (Z.-R.H.); (B.L.); (Z.-X.L.)
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (W.-L.G.); (F.Z.)
| | - Zi-Rui Huang
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.-J.C.); (Z.-R.H.); (B.L.); (Z.-X.L.)
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (W.-L.G.); (F.Z.)
| | - Shi-Ze You
- School of Clinical Medicine, Fujian Medical University, Fuzhou 350122, China;
| | - Wei-Ling Guo
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (W.-L.G.); (F.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Fang Zhang
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (W.-L.G.); (F.Z.)
| | - Bin Liu
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.-J.C.); (Z.-R.H.); (B.L.); (Z.-X.L.)
| | - Xu-Cong Lv
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (W.-L.G.); (F.Z.)
| | - Zhan-Xi Lin
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.-J.C.); (Z.-R.H.); (B.L.); (Z.-X.L.)
| | - Peng-Hu Liu
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.-J.C.); (Z.-R.H.); (B.L.); (Z.-X.L.)
| |
Collapse
|
7
|
Pfaff A, Chernatynskaya A, Vineyard H, Ercal N. Thiol antioxidants protect human lens epithelial (HLE B-3) cells against tert-butyl hydroperoxide-induced oxidative damage and cytotoxicity. Biochem Biophys Rep 2022; 29:101213. [PMID: 35128081 PMCID: PMC8808075 DOI: 10.1016/j.bbrep.2022.101213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 12/20/2022] Open
Abstract
Oxidative damage to lens epithelial cells plays an important role in the development of age-related cataract, and the health of the lens has important implications for overall ocular health. As a result, there is a need for effective therapeutic agents that prevent oxidative damage to the lens. Thiol antioxidants such as tiopronin or N-(2-mercaptopropionyl)glycine (MPG), N-acetylcysteine amide (NACA), N-acetylcysteine (NAC), and exogenous glutathione (GSH) may be promising candidates for this purpose, but their ability to protect lens epithelial cells is not well understood. The effectiveness of these compounds was compared by exposing human lens epithelial cells (HLE B-3) to the chemical oxidant tert-butyl hydroperoxide (tBHP) and treating the cells with each of the antioxidant compounds. MTT cell viability, apoptosis, reactive oxygen species (ROS), and levels of intracellular GSH, the most important antioxidant in the lens, were measured after treatment. All four compounds provided some degree of protection against tBHP-induced oxidative stress and cytotoxicity. Cells treated with NACA exhibited the highest viability after exposure to tBHP, as well as decreased ROS and increased intracellular GSH. Exogenous GSH also preserved viability and increased intracellular GSH levels. MPG scavenged significant amounts of ROS, and NAC increased intracellular GSH levels. Our results suggest that both scavenging ROS and increasing GSH may be necessary for effective protection of lens epithelial cells. Further, the compounds tested may be useful for the development of therapeutic strategies that aim to prevent oxidative damage to the lens.
Collapse
Key Words
- 7-AAD, 7-aminoactinomycin D
- ATCC, American Type Culture Collection
- Antioxidant
- Carboxy-H2DCFDA, 6-carboxy-2′,7′-dichlorodihydrofluorescein diacetate
- Cataract
- EMEM, Eagle's minimum essential medium
- FBS, fetal bovine serum
- FDA, United States Food and Drug Administration
- GSH, glutathione
- GSSG, glutathione disulfide
- Glutathione
- H2O2, hydrogen peroxide
- HLE B-3, human (eye) lens epithelial cell line B-3
- Lens
- MPG, N-(2-mercaptopropionyl)glycine
- MTT, (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)
- NAC, N-acetylcysteine
- NACA, N-acetylcysteine amide
- OH•, hydroxyl radical
- Oxidative stress
- PBS, phosphate-buffered saline
- ROS, reactive oxygen species
- Thiol
- tBHP, tert-butyl hydroperoxide
Collapse
Affiliation(s)
| | | | - Hannah Vineyard
- Department of Chemistry, Missouri University of Science & Technology, 104 Schrenk Hall, 400 W. 11th Street, Rolla, MO, 65409, USA
| | - Nuran Ercal
- Department of Chemistry, Missouri University of Science & Technology, 104 Schrenk Hall, 400 W. 11th Street, Rolla, MO, 65409, USA
| |
Collapse
|
8
|
Benić MS, Nežić L, Vujić-Aleksić V, Mititelu-Tartau L. Novel Therapies for the Treatment of Drug-Induced Liver Injury: A Systematic Review. Front Pharmacol 2022; 12:785790. [PMID: 35185538 PMCID: PMC8847672 DOI: 10.3389/fphar.2021.785790] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/30/2021] [Indexed: 12/15/2022] Open
Abstract
Many drugs with different mechanisms of action and indications available on the market today are capable of inducing hepatotoxicity. Drug-induced liver injury (DILI) has been a treatment challenge nowadays as it was in the past. We searched Medline (via PubMed), CENTRAL, Science Citation Index Expanded, clinical trials registries and databases of DILI and hepatotoxicity up to 2021 for novel therapies for the management of adult patients with DILI based on the combination of three main search terms: 1) treatment, 2) novel, and 3) drug-induced liver injury. The mechanism of action of novel therapies, the potential of their benefit in clinical settings, and adverse drug reactions related to novel therapies were extracted. Cochrane Risk of bias tool and Grading of Recommendations Assessment, Development and Evaluation (GRADE) assessment approach was involved in the assessment of the certainty of the evidence for primary outcomes of included studies. One thousand three hundred seventy-two articles were identified. Twenty-eight articles were included in the final analysis. Eight randomized controlled trials (RCTs) were detected and for six the available data were sufficient for analysis. In abstract form only we found six studies which were also anaylzed. Investigated agents included: bicyclol, calmangafodipir, cytisin amidophospate, fomepizole, livina-polyherbal preparation, magnesium isoglycyrrhizinate (MgIG), picroliv, plasma exchange, radix Paeoniae Rubra, and S-adenosylmethionine. The primary outcomes of included trials mainly included laboratory markers improvement. Based on the moderate-certainty evidence, more patients treated with MgIG experienced alanine aminotransferase (ALT) normalization compared to placebo. Low-certainty evidence suggests that bicyclol treatment leads to a reduction of ALT levels compared to phosphatidylcholine. For the remaining eight interventions, the certainty of the evidence for primary outcomes was assessed as very low and we are very uncertain in any estimate of effect. More effort should be involved to investigate the novel treatment of DILI. Well-designed RCTs with appropriate sample sizes, comparable groups and precise, not only surrogate outcomes are urgently welcome.
Collapse
Affiliation(s)
- Mirjana Stanić Benić
- Department of Clinical Pharmacology, Clinical Hospital Centre Rijeka, Rijeka, Croatia
| | - Lana Nežić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Vesna Vujić-Aleksić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- The Republic of Srpska Agency for Certification, Accreditation and Quality Improvement in Health Care, Banja Luka, Bosnia and Herzegovina
| | | |
Collapse
|
9
|
Atlas D. Emerging therapeutic opportunities of novel thiol-amides, NAC-amide (AD4/NACA) and thioredoxin mimetics (TXM-Peptides) for neurodegenerative-related disorders. Free Radic Biol Med 2021; 176:120-141. [PMID: 34481041 DOI: 10.1016/j.freeradbiomed.2021.08.239] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/17/2021] [Accepted: 08/29/2021] [Indexed: 12/23/2022]
Abstract
Understanding neurodegenerative diseases have challenged scientists for decades. It has become apparent that a decrease in life span is often correlated with the development of neurodegenerative disorders. Oxidative stress and the subsequent inflammatory damages appear to contribute to the different molecular and biochemical mechanisms associated with neurodegeneration. In this review, I examine the protective properties of novel amino acid based compounds, comprising the AD series (AD1-AD7) in particular N-acetylcysteine amide, AD4, also called NACA, and the series of thioredoxin mimetic (TXM) peptides, TXM-CB3-TXM-CB16. Designed to cross the blood-brain-barrier (BBB) and permeate the cell membrane, these antioxidant/anti-inflammatory compounds may enable effective treatment of neurodegenerative related disorders. The review addresses the molecular mechanism of cellular protection exhibited by these new reagents, focusing on the reversal of oxidative stress, mitochondrial stress, inflammatory damages, and prevention of premature cell death. In addition, it will cover the outlook of the clinical prospects of AD4/NACA and the thioredoxin-mimetic peptides, which are currently in development.
Collapse
Affiliation(s)
- Daphne Atlas
- Professor of Neurochemistry, Dept. of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel.
| |
Collapse
|
10
|
Eshrati R, Jafari M, Gudarzi S, Nazari A, Samizadeh E, Ghafourian Hesami M. Comparison of ameliorative effects of Taraxacum syriacum and N-acetylcysteine against acetaminophen-induced oxidative stress in rat liver and kidney. J Biochem 2021; 169:337-350. [PMID: 32970799 DOI: 10.1093/jb/mvaa107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/08/2020] [Indexed: 11/13/2022] Open
Abstract
Taraxacum syriacum (TS) with natural antioxidant and pharmacological activities may be considered for treatment of oxidative stress induced by acetaminophen (APAP). The aim of this study was to evaluate the ameliorative effects of the ethanol extract of TS root against hepatorenal toxicity induced by APAP in comparison to N-acetylcysteine (NAC) as a standard drug. Thirty male Wistar rats were randomly divided into five groups. Control group; APAP (1 g/kg) group; APAP-NAC (160 mg/kg) group and APAP-TS100 and APAP-TS200 groups: APAP plus 100 and 200 mg/kg of TS extract, respectively. After 7 days treatment, serum and liver and kidney tissues were prepared and evaluated. TS extract ameliorated the increased lipid peroxidation level and decreased antioxidant enzymes activities and glutathione level in liver and kidney of APAP-treated rats. Moreover, treatment with the TS extract caused significant reduction in the histopathological damages and high levels of serum biochemical markers of hepatic and renal functions after APAP treatment. This study suggests that the extract of TS roots has dose-dependent ameliorative effect against APAP-induced oxidative damage in liver and kidney due to its free radical scavenging and antioxidant properties. The overall efficacy of the extract at 200 mg/kg dose is comparable with NAC.
Collapse
Affiliation(s)
- Reza Eshrati
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahvash Jafari
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Saeed Gudarzi
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Afshen Nazari
- Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | | |
Collapse
|
11
|
Dang XL, Yang LF, Shi L, Li LF, He P, Chen J, Zheng BJ, Yang P, Wen AD. Post-treatment with glycyrrhizin can attenuate hepatic mitochondrial damage induced by acetaminophen in mice. Exp Biol Med (Maywood) 2021; 246:1219-1227. [PMID: 33342284 PMCID: PMC8142107 DOI: 10.1177/1535370220977823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/03/2020] [Indexed: 11/16/2022] Open
Abstract
Overdose of acetaminophen (APAP) is responsible for the most cases of acute liver failure worldwide. Hepatic mitochondrial damage mediated by neuronal nitric oxide synthase- (nNOS) induced liver protein tyrosine nitration plays a critical role in the pathophysiology of APAP hepatotoxicity. It has been reported that pre-treatment or co-treatment with glycyrrhizin can protect against hepatotoxicity through prevention of hepatocellular apoptosis. However, the majority of APAP-induced acute liver failure cases are people intentionally taking the drug to commit suicide. Any preventive treatment is of little value in practice. In addition, the hepatocellular damage induced by APAP is considered to be oncotic necrosis rather than apoptosis. In the present study, our aim is to investigate if glycyrrhizin can be used therapeutically and the underlying mechanisms of APAP hepatotoxicity protection. Hepatic damage was induced by 300 mg/kg APAP in balb/c mice, followed with administration of 40, 80, or 160 mg/kg glycyrrhizin 90 min later. Mice were euthanized and harvested at 6 h post-APAP. Compared with model controls, glycyrrhizin post-treatment attenuated hepatic mitochondrial and hepatocellular damages, as indicated by decreased serum glutamate dehydrogenase, alanine aminotransferase, and aspartate aminotransferase activities as well as ameliorated mitochondrial swollen, distortion, and hepatocellular necrosis. Notably, 80 mg/kg glycyrrhizin inhibited hepatic nNOS activity and its mRNA and protein expression levels by 16.9, 14.9, and 28.3%, respectively. These results were consistent with the decreased liver nitric oxide content and liver protein tyrosine nitration indicated by 3-nitrotyrosine staining. Moreover, glycyrrhizin did not affect the APAP metabolic activation, and the survival rate of ALF mice was increased by glycyrrhizin. The present study indicates that post-treatment with glycyrrhizin can dose-dependently attenuate hepatic mitochondrial damage and inhibit the up-regulation of hepatic nNOS induced by APAP. Glycyrrhizin shows promise as drug for the treatment of APAP hepatotoxicity.
Collapse
Affiliation(s)
- Xue-Liang Dang
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, China
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Long-Fei Yang
- Departments of Medical Laboratory and Research Center, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Lei Shi
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Long-Fei Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Ping He
- Renji Hospital, Medical School of Shanghai Jiaotong University, Shanghai 200127, China
| | - Jie Chen
- Renji Hospital, Medical School of Shanghai Jiaotong University, Shanghai 200127, China
| | - Bei-Jie Zheng
- Renji Hospital, Medical School of Shanghai Jiaotong University, Shanghai 200127, China
| | - Peng Yang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Ai-Dong Wen
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
12
|
Tan YL, Tey SM, Ho HK. Moderate Hypothermia Effectively Alleviates Acetaminophen-Induced Liver Injury With Prolonged Action Beyond Cooling. Dose Response 2020; 18:1559325820970846. [PMID: 33239997 PMCID: PMC7675884 DOI: 10.1177/1559325820970846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022] Open
Abstract
Acetaminophen (APAP) overdose accounts for the highest incidence of acute liver failure, despite the availability of an antidote i.e. N-acetylcysteine. This calls for alternative strategies to manage APAP-induced liver injury (AILI). Therapeutic hypothermia has been explored in past studies for hepatoprotection, but these phenomenal reports lack clarification of its optimal window for application, and mechanistic effects in specific AILI. Hence, we conducted an in vitro study with transforming growth factor-α transgenic mouse hepatocytes cell line, TAMH, and human liver hepatocytes cell line, L-02, where cells were conditioned with deep (25°C) or moderate (32°C) hypothermia before, during or after APAP toxicity. Cell viability was evaluated as a hallmark of cytoprotection, along with cell death. Simultaneously, cold shock proteins (CSPs) and heat shock proteins expressions were monitored; key liver functions including drug-metabolizing ability and hepatic clearance were also investigated. Herein, we demonstrated significant hepatoprotection with 24-hour moderate hypothermic conditioning during AILI and this effect sustained for at least 24 hours of rewarming. Such liver preservation was associated with a CSP—RNA-binding motif protein 3 (RBM3) as its knockdown promptly abolished the cytoprotective effects of hypothermia. With mild and reversible liver perturbations, hypothermic therapy appears promising and its RBM3 involvement deserves future exploration.
Collapse
Affiliation(s)
- Yeong Lan Tan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences & Engineering, Centre for Life Sciences, National University of Singapore, Singapore
| | - Siew Min Tey
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences & Engineering, Centre for Life Sciences, National University of Singapore, Singapore
| |
Collapse
|
13
|
Pfaff AR, Beltz J, King E, Ercal N. Medicinal Thiols: Current Status and New Perspectives. Mini Rev Med Chem 2020; 20:513-529. [PMID: 31746294 PMCID: PMC7286615 DOI: 10.2174/1389557519666191119144100] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 02/08/2023]
Abstract
The thiol (-SH) functional group is found in a number of drug compounds and confers a unique combination of useful properties. Thiol-containing drugs can reduce radicals and other toxic electrophiles, restore cellular thiol pools, and form stable complexes with heavy metals such as lead, arsenic, and copper. Thus, thiols can treat a variety of conditions by serving as radical scavengers, GSH prodrugs, or metal chelators. Many of the compounds discussed here have been in use for decades, yet continued exploration of their properties has yielded new understanding in recent years, which can be used to optimize their clinical application and provide insights into the development of new treatments. The purpose of this narrative review is to highlight the biochemistry of currently used thiol drugs within the context of developments reported in the last five years. More specifically, this review focuses on thiol drugs that represent the standard of care for their associated conditions, including N-acetylcysteine, 2,3-meso-dimercaptosuccinic acid, British anti-Lewisite, D-penicillamine, amifostine, and others. Reports of novel dosing regimens, delivery strategies, and clinical applications for these compounds were examined with an eye toward emerging approaches to address a wide range of medical conditions in the future.
Collapse
Affiliation(s)
- Annalise R. Pfaff
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri, U.S.A
| | - Justin Beltz
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri, U.S.A
| | - Emily King
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri, U.S.A
| | - Nuran Ercal
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri, U.S.A
| |
Collapse
|
14
|
Hu H, Fan X, Guo Q, Wei X, Yang D, Zhang B, Liu J, Wu Q, Oh Y, Feng Y, Chen K, Hou L, Gu N. Silicon dioxide nanoparticles induce insulin resistance through endoplasmic reticulum stress and generation of reactive oxygen species. Part Fibre Toxicol 2019; 16:41. [PMID: 31699096 PMCID: PMC6836410 DOI: 10.1186/s12989-019-0327-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/23/2019] [Indexed: 12/18/2022] Open
Abstract
Background Silicon dioxide nanoparticles (SiO2 NPs) are one of the most widely utilized NPs in various food sectors. However, the potential endocrine toxicity of SiO2 NPs has not been characterized. Results In the present study, mice were orally administered a series of doses of SiO2 NPs. All doses of SiO2 NPs were absorbed into the blood, liver, and pancreas of the mice. Administration of 100 mg/kg bw (body weight) of SiO2 NPs significantly increased blood glucose levels in mice. However, the same dose of SiO2 fine-particles (FPs) did not result in altered blood glucose. Whole-genome analysis showed that SiO2 NPs affected the expression of genes associated with reactive oxygen species (ROS) production and endoplasmic reticulum (ER) stress. In addition, we showed that SiO2 NPs activated xenobiotic metabolism, resulting in ER stress. Endoplasmic reticulum stress resulted in increased ROS production, which activated the NF-κB pathway leading to expression of inflammatory cytokines. Increased inflammatory cytokine expression resulted in serine phosphorylation of IRS1, which induced insulin resistance (IR). Furthermore these inflammatory cytokines activated the MAPK pathway, which further promoted the serine phosphorylation of IRS1. Insulin resistance resulted in elevated blood glucose. The ER stress inhibitor 4-phenylbutyric acid (4-PBA) inhibited SiO2 NP-induced ROS production. The ROS scavenger N-acetylcysteine (NAC) did not affect SiO2 NP-induced ER stress, but inhibited SiO2 NP-induced activation of the NF-κB and MAPK pathways, expression of inflammatory cytokines, SiO2 NP-induced serine phosphorylation of IRS1, and SiO2 NP-induced elevations of blood glucose. Conclusion Silicon dioxide NPs induced IR through ER stress and generation of ROS, but SiO2 FPs did not. Therefore, lifelong exposure of humans to SiO2 NPs may result in detrimental effects on blood glucose. The results of this study strongly suggested that non-nanoformed SiO2 should be used as food additives.
Collapse
Affiliation(s)
- Hailong Hu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Xingpei Fan
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Qian Guo
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Xiangjuan Wei
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Daqian Yang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Boya Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Jing Liu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China
| | - Yuri Oh
- Faculty of Education, Wakayama University, Wakayama, Japan
| | - Yujie Feng
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Kun Chen
- The Joint Research Center of Guangzhou University and Keele University for Gene Interference and Application, School of Life Science, Guangzhou University, Guangzhou, China
| | - Liping Hou
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Ning Gu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin, Heilongjiang, 150001, China.
| |
Collapse
|
15
|
Yang Y, Li L, Hang Q, Fang Y, Dong X, Cao P, Yin Z, Luo L. γ-glutamylcysteine exhibits anti-inflammatory effects by increasing cellular glutathione level. Redox Biol 2018; 20:157-166. [PMID: 30326393 PMCID: PMC6197438 DOI: 10.1016/j.redox.2018.09.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to infection and characterized by redox imbalance and severe oxidative stress. Glutathione (GSH) serves several vital functions, including scavenging free radicals and maintaining intracellular redox balance. Extracellular GSH is unable to be taken into the majority of human cells, and the GSH prodrug N-acetyl-l-cysteine (NAC) does not exhibit promising clinical effects. γ-glutamylcysteine (γ-GC), an intermediate dipeptide of the GSH-synthesis pathway and harboring anti-inflammatory properties, represents a relatively unexplored option for sepsis treatment. The anti-inflammatory efficiency of γ-GC and the associated molecular mechanism need to be explored. In vivo investigation showed that γ-GC reduced sepsis lethality and attenuated systemic inflammatory responses in mice, as well as inhibited lipopolysaccharide (LPS)-stimulated production of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), high-mobility group box 1 (HMGB1), and nitric oxide (NO) and the expression of inducible NO synthase and cyclooxygenase 2 in RAW264.7 cells. Moreover, both in vivo and in vitro experiments demonstrated that γ-GC exhibited better therapeutic effects against inflammation compared with N-acetyl-L-cysteine (NAC) and GSH. Mechanistically, γ-GC suppressed LPS-induced reactive oxygen species accumulation and GSH depletion. Inflammatory stimuli, such as LPS treatment, upregulated the expression of glutathione synthetase via activating nuclear factor-erythroid 2-related factor (Nrf2) and nuclear factor kappa B (NF-κB) pathways, thereby promoting synthesis of GSH from γ-GC. These findings suggested that γ-GC might represent a potential therapeutic agent for sepsis treatment. γ-GC reduces sepsis lethality and attenuates inflammatory responses in BALB/c mice. γ-GC suppresses LPS-induced inflammation, ROS accumulation, and GSH depletion. Nrf2 and NF-κB pathways are essential for upregulating GSS level to promote GSH synthesis from γ-GC. γ-GC is more effective in attenuation inflammation than NAC and GSH.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Ling Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu, China
| | - Qiyun Hang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu, China
| | - Yuan Fang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Xiaoliang Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu, China
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, China.
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
16
|
Rharass T, Lantow M, Gbankoto A, Weiss DG, Panáková D, Lucas S. Ascorbic acid alters cell fate commitment of human neural progenitors in a WNT/β-catenin/ROS signaling dependent manner. J Biomed Sci 2017; 24:78. [PMID: 29037191 PMCID: PMC5641995 DOI: 10.1186/s12929-017-0385-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 10/06/2017] [Indexed: 12/22/2022] Open
Abstract
Background Improving the neuronal yield from in vitro cultivated neural progenitor cells (NPCs) is an essential challenge in transplantation therapy in neurological disorders. In this regard, Ascorbic acid (AA) is widely used to expand neurogenesis from NPCs in cultures although the mechanisms of its action remain unclear. Neurogenesis from NPCs is regulated by the redox-sensitive WNT/β-catenin signaling pathway. We therefore aimed to investigate how AA interacts with this pathway and potentiates neurogenesis. Methods Effects of 200 μM AA were compared with the pro-neurogenic reagent and WNT/β-catenin signaling agonist lithium chloride (LiCl), and molecules with antioxidant activities i.e. N-acetyl-L-cysteine (NAC) and ruthenium red (RuR), in differentiating neural progenitor ReNcell VM cells. Cells were supplemented with reagents for two periods of treatment: a full period encompassing the whole differentiation process versus an early short period that is restricted to the cell fate commitment stage. Intracellular redox balance and reactive oxygen species (ROS) metabolism were examined by flow cytometry using redox and ROS sensors. Confocal microscopy was performed to assess cell viability, neuronal yield, and levels of two proteins: Nucleoredoxin (NXN) and the WNT/β-catenin signaling component Dishevelled 2 (DVL2). TUBB3 and MYC gene responses were evaluated by quantitative real-time PCR. DVL2-NXN complex dissociation was measured by fluorescence resonance energy transfer (FRET). Results In contrast to NAC which predictably exhibited an antioxidant effect, AA treatment enhanced ROS metabolism with no cytotoxic induction. Both drugs altered ROS levels only at the early stage of the differentiation as no changes were held beyond the neuronal fate commitment stage. FRET studies showed that AA treatment accelerated the redox-dependent release of the initial pool of DVL2 from its sequestration by NXN, while RuR treatment hampered the dissociation of the two proteins. Accordingly, AA increased WNT/β-catenin signaling output i.e. MYC mRNA level, whereas RuR attenuated it. Moreover, AA improved neurogenesis as much as LiCl as both TUBB3-positive cell yield and TUBB3 mRNA level increased, while NAC or RuR attenuated neurogenesis. Markedly, the neurogenesis outputs between the short and the full treatment with either NAC or AA were found unchanged, supporting our model that neuronal yield is altered by events taking place at the early phase of differentiation. Conclusions Our findings demonstrate that AA treatment elevates ROS metabolism in a non-lethal manner prior to the NPCs commitment to their neuronal fate. Such effect stimulates the redox-sensitive DVL2 activation and WNT/β-catenin signaling response that would enhance the ensuing neuronal cell differentiation. Electronic supplementary material The online version of this article (10.1186/s12929-017-0385-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tareck Rharass
- Physiopathology of Inflammatory Bone Diseases, University of the Littoral Opal Coast, F-62327, Boulogne sur Mer, France. .,Electrochemical Signaling in Development and Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, D-13125, Berlin, Germany. .,Physiopathology of Inflammatory Bone Diseases, University of the Littoral Opal Coast, Boulevard Bassin Napoléon - Quai Masset, B.P. 120, F-62327, Boulogne sur Mer, Cédex, France.
| | - Margareta Lantow
- Cell Biology and Biosystems Technology, Institute of Biological Sciences, University of Rostock, D-18059, Rostock, Germany
| | - Adam Gbankoto
- Department of Animal Physiology, Faculty of Sciences and Technics, University of Abomey-Calavi, 01, BP, 526, Cotonou, Benin
| | - Dieter G Weiss
- Cell Biology and Biosystems Technology, Institute of Biological Sciences, University of Rostock, D-18059, Rostock, Germany
| | - Daniela Panáková
- Electrochemical Signaling in Development and Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, D-13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, D-13125, Berlin, Germany
| | - Stéphanie Lucas
- Physiopathology of Inflammatory Bone Diseases, University of the Littoral Opal Coast, F-62327, Boulogne sur Mer, France
| |
Collapse
|
17
|
Gill P, Bhattacharyya S, McCullough S, Letzig L, Mishra PJ, Luo C, Dweep H, James L. MicroRNA regulation of CYP 1A2, CYP3A4 and CYP2E1 expression in acetaminophen toxicity. Sci Rep 2017; 7:12331. [PMID: 28951593 PMCID: PMC5614957 DOI: 10.1038/s41598-017-11811-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/30/2017] [Indexed: 01/29/2023] Open
Abstract
MicroRNAs (miRNAs) that regulate the cytochrome P-450 isoforms involved in acetaminophen (APAP) toxicity were examined in HepaRG cells treated with APAP (20 mM). In-vitro studies found that APAP protein adducts were increased at 1 h, followed by ALT increases at 12 and 24 h. CYP1A2, CYP3A4 and CYP2E1 mRNA levels were decreased, while miRNAs were increased for miR-122-5p, miR-378a-5p, miR-27b-3p at 6 h and miR-125b-5p at 12 h and miR-27b-3p at 24 h. Putative miRNA binding sites on the 3′UTRs of the CYPs were identified in-silico. Overexpression of miR-122-5p and miR-378a-5p in cells suppressed protein expression of CYP1A2, CYP3A4 and CYP2E1. Luciferase reporter assays confirmed the interaction between miR-122 and the 3′UTR of the CYP1A2 and CYP3A4. Thus, the in-vitro experiments showed that miR-122-5p and miR-378a-5p upregulation were associated with translational repression of CYPs. Serum samples of children with APAP overdose had significant elevation of miR-122-5p, miR-378a-5p, miR-125b-5p and miR-27b-3p, compared to healthy controls and receiver operator curves of the miRNAs had AUCs of 91 to 100%. Collectively, the data suggest that miRNA elevations in APAP toxicity represent a regulatory response to modify CYP1A2, CYP3A4 and CYP2E1 translation due to cellular stress and injury.
Collapse
Affiliation(s)
- Pritmohinder Gill
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA. .,Arkansas Children's Research Institute, Little Rock, AR, 72202, USA.
| | - Sudeepa Bhattacharyya
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.,Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Sandra McCullough
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.,Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Lynda Letzig
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.,Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Prasun J Mishra
- Department of Biochemical and Cellular Pharmacology, Genentech, 1, DNA Way, South San Francisco, California, 94080, USA
| | - Chunqiao Luo
- Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Harsh Dweep
- The Wistar Institute, 3601 Spruce St, Philadelphia, Pennsylvania, 19104, USA
| | - Laura James
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.,Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| |
Collapse
|
18
|
Maddirala Y, Tobwala S, Karacal H, Ercal N. Prevention and reversal of selenite-induced cataracts by N-acetylcysteine amide in Wistar rats. BMC Ophthalmol 2017; 17:54. [PMID: 28446133 PMCID: PMC5405552 DOI: 10.1186/s12886-017-0443-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 04/20/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The present study sought to evaluate the efficacy of N-acetylcysteine amide (NACA) eye drops in reversing the cataract formation induced by sodium selenite in male Wistar rat pups. METHODS Forty male Wistar rat pups were randomly divided into a control group, an N-acetylcysteine amide-only group, a sodium selenite-induced cataract group, and a NACA-treated sodium selenite-induced cataract group. Sodium selenite was injected intraperitoneally on postpartum day 10, whereas N-acetylcysteine amide was injected intraperitoneally on postpartum days 9, 11, and 13 in the respective groups. Cataracts were evaluated at the end of week 2 (postpartum day 14) when the rat pups opened their eyes. N-acetylcysteine amide eye drops were administered beginning on week 3 until the end of week 4 (postpartum days 15 to 30), and the rats were sacrificed at the end of week 4. Lenses were isolated and examined for oxidative stress parameters such as glutathione, lipid peroxidation, and calcium levels along with the glutathione reductase and thioltransferase enzyme activities. Casein zymography and Western blot of m-calpain were performed using the water soluble fraction of lens proteins. RESULTS Morphological examination of the lenses in the NACA-treated group indicated that NACA was able to reverse the cataract grade. In addition, glutathione level, thioltransferase activity, m-calpain activity, and m-calpain level (as assessed by Western blot) were all significantly higher in the NACA-treated group than in the sodium selenite-induced cataract group. Furthermore, sodium selenite- injected rat pups had significantly higher levels of malondialdehyde, glutathione reductase enzyme activity, and calcium levels, which were reduced to control levels upon treatment with NACA. CONCLUSIONS The data suggest that NACA has the potential to significantly improve vision and decrease the burden of cataract-related loss of function. Prevention and reversal of cataract formation could have a global impact. Development of pharmacological agents like NACA may eventually prevent cataract formation in high-risk populations and may prevent progression of early-stage cataracts. This brings a paradigm shift from expensive surgical treatment of cataracts to relatively inexpensive prevention of vision loss.
Collapse
Affiliation(s)
- Yasaswi Maddirala
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409 USA
| | - Shakila Tobwala
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409 USA
| | - Humeyra Karacal
- Department of Ophthalmology, Washington University, St. Louis, MO 63110 USA
| | - Nuran Ercal
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409 USA
| |
Collapse
|
19
|
Benterud T, Ystgaard MB, Manueldas S, Pankratov L, Alfaro-Cervello C, Florholmen G, Ahmed MS, Sandvik L, Norgren S, Bjørås M, Baumbusch LO, Solberg R, Saugstad OD. N-Acetylcysteine Amide Exerts Possible Neuroprotective Effects in Newborn Pigs after Perinatal Asphyxia. Neonatology 2017; 111:12-21. [PMID: 27497671 DOI: 10.1159/000447255] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/29/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Perinatal asphyxia and ensuing reoxygenation change the antioxidant capacity of cells and organs. OBJECTIVES To analyze the neuroprotective effect of the antioxidant N-acetylcysteine amide (NACA) after perinatal hypoxia-reoxygenation with an emphasis on proinflammatory cytokines and the transcription factor NF-κB in the prefrontal cortex of neonatal pigs. METHODS Twenty-nine newborn pigs, aged 12-36 h, were subjected to global hypoxia and hypercapnia. One sham-operated group (n = 5) and 2 experimental groups (n = 12) were exposed to 8% oxygen, until the base excess was -20 mmol/l or the mean arterial blood pressure fell to <20 mm Hg (asphyxia with NACA or saline). The pigs were observed for 9.5 h after hypoxia. Samples of prefrontal cortex and plasma were analyzed. RESULTS Cortex: there was no significant difference in mRNA expression between the intervention groups regarding IL-1β, IL6, TNFα, MMP2, MMP9 or IL18. Pigs exposed to hypoxia-reoxygenation and treatment with NACA (NACA-pigs) had a significantly lower protein concentration of IL-1β than pigs treated with saline (placebo controls), i.e. 8.8 ± 3.9 versus 16.8 ± 10.5 pg/mg protein (p = 0.02). The activation of the transcription factor NF-κB (measured as the fold-change of phosphorylated p65Ser 536), was reduced in the NACA-pigs when compared to the placebo controls (5.2 ± 4.3 vs. 16.0 ± 13.5; p = 0.02). No difference between the intervention groups regarding brain histopathology or in the levels of 8-oxoguanine measured in the prefrontal cortex were observed. Plasma: the NACA-pigs had a stronger reduction of TNFα in the first 30 min following asphyxia compared with the placebo controls, i.e. 36 (30-44) versus 24 (14-32)% (p = 0.01). CONCLUSION The reduced levels of the pivotal inflammatory markers IL-1β and TNFα and the transcription factor NF-κB may indicate that NACA has possible neuroprotective effects after perinatal asphyxia.
Collapse
Affiliation(s)
- Torkil Benterud
- Department of Pediatric Research, University of Oslo, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Carver KA, Yang D. N-Acetylcysteine Amide Protects Against Oxidative Stress-Induced Microparticle Release From Human Retinal Pigment Epithelial Cells. Invest Ophthalmol Vis Sci 2016; 57:360-71. [PMID: 26842754 PMCID: PMC4736743 DOI: 10.1167/iovs.15-17117] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Oxidative stress is a major factor involved in retinal pigment epithelium (RPE) apoptosis that underlies AMD. Drusen, extracellular lipid- and protein-containing deposits, are strongly associated with the development of AMD. Cell-derived microparticles (MPs) are small membrane-bound vesicles shed from cells. The purpose of this study was to determine if oxidative stress drives MP release from RPE cells, to assess whether these MPs carry membrane complement regulatory proteins (mCRPs: CD46, CD55, and CD59), and to evaluate the effects of a thiol antioxidant on oxidative stress–induced MP release. Methods Retinal pigment epithelium cells isolated from human donor eyes were cultured and treated with hydrogen peroxide (H2O2) to induce oxidative stress. Isolated MPs were fixed for transmission electron microscopy or processed for component analysis by flow cytometry, Western blot analysis, and confocal microscopy. Results Transmission electron microscopy showed that MPs ranged in diameter from 100 to 1000 nm. H2O2 treatment led to time- and dose-dependent elevations in MPs with externalized phosphatidylserine and phosphatidylethanolamine, known markers of MPs. These increases were strongly correlated to RPE apoptosis. Oxidative stress significantly increased the release of mCRP-positive MPs, which were prevented by a thiol antioxidant, N-acetylcysteine amide (NACA). Conclusions This is the first evidence that oxidative stress induces cultured human RPE cells to release MPs that carry mCRPs on their surface. The levels of released MPs are strongly correlated with RPE apoptosis. N-acetylcysteine amide prevents oxidative stress–induced effects. Our findings indicate that oxidative stress reduces mCRPs on the RPE surface through releasing MPs.
Collapse
|
21
|
N-acetylcysteine amid reduces pancreatic damage in a rat model of acute necrotizing pancreatitis. J Surg Res 2016; 203:383-9. [DOI: 10.1016/j.jss.2016.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/11/2016] [Accepted: 03/02/2016] [Indexed: 02/07/2023]
|
22
|
Wang J, Chen F, Liu L, Qi C, Wang B, Yan X, Huang C, Hou W, Zhang MQ, Chen Y, Du Y. Engineering EMT using 3D micro-scaffold to promote hepatic functions for drug hepatotoxicity evaluation. Biomaterials 2016; 91:11-22. [PMID: 26994875 DOI: 10.1016/j.biomaterials.2016.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 03/01/2016] [Indexed: 12/25/2022]
|
23
|
Maes M, Vinken M, Jaeschke H. Experimental models of hepatotoxicity related to acute liver failure. Toxicol Appl Pharmacol 2016; 290:86-97. [PMID: 26631581 PMCID: PMC4691574 DOI: 10.1016/j.taap.2015.11.016] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/19/2015] [Accepted: 11/24/2015] [Indexed: 12/13/2022]
Abstract
Acute liver failure can be the consequence of various etiologies, with most cases arising from drug-induced hepatotoxicity in Western countries. Despite advances in this field, the management of acute liver failure continues to be one of the most challenging problems in clinical medicine. The availability of adequate experimental models is of crucial importance to provide a better understanding of this condition and to allow identification of novel drug targets, testing the efficacy of new therapeutic interventions and acting as models for assessing mechanisms of toxicity. Experimental models of hepatotoxicity related to acute liver failure rely on surgical procedures, chemical exposure or viral infection. Each of these models has a number of strengths and weaknesses. This paper specifically reviews commonly used chemical in vivo and in vitro models of hepatotoxicity associated with acute liver failure.
Collapse
Affiliation(s)
- Michaël Maes
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, United States
| |
Collapse
|
24
|
Michaut A, Le Guillou D, Moreau C, Bucher S, McGill MR, Martinais S, Gicquel T, Morel I, Robin MA, Jaeschke H, Fromenty B. A cellular model to study drug-induced liver injury in nonalcoholic fatty liver disease: Application to acetaminophen. Toxicol Appl Pharmacol 2015; 292:40-55. [PMID: 26739624 DOI: 10.1016/j.taap.2015.12.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022]
Abstract
Obesity and nonalcoholic fatty liver disease (NAFLD) can increase susceptibility to hepatotoxicity induced by some xenobiotics including drugs, but the involved mechanisms are poorly understood. For acetaminophen (APAP), a role of hepatic cytochrome P450 2E1 (CYP2E1) is suspected since the activity of this enzyme is consistently enhanced during NAFLD. The first aim of our study was to set up a cellular model of NAFLD characterized not only by triglyceride accumulation but also by higher CYP2E1 activity. To this end, human HepaRG cells were incubated for one week with stearic acid or oleic acid, in the presence of different concentrations of insulin. Although cellular triglycerides and the expression of lipid-responsive genes were similar with both fatty acids, CYP2E1 activity was significantly increased only by stearic acid. CYP2E1 activity was reduced by insulin and this effect was reproduced in cultured primary human hepatocytes. Next, APAP cytotoxicity was assessed in HepaRG cells with or without lipid accretion and CYP2E1 induction. Experiments with a large range of APAP concentrations showed that the loss of ATP and glutathione was almost always greater in the presence of stearic acid. In cells pretreated with the CYP2E1 inhibitor chlormethiazole, recovery of ATP was significantly higher in the presence of stearate with low (2.5mM) or high (20mM) concentrations of APAP. Levels of APAP-glucuronide were significantly enhanced by insulin. Hence, HepaRG cells can be used as a valuable model of NAFLD to unveil important metabolic and hormonal factors which can increase susceptibility to drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Anaïs Michaut
- INSERM, U991, Université de Rennes 1, Rennes, France
| | | | - Caroline Moreau
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | - Simon Bucher
- INSERM, U991, Université de Rennes 1, Rennes, France
| | - Mitchell R McGill
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Thomas Gicquel
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | - Isabelle Morel
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | | | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | | |
Collapse
|
25
|
Oxidative Stress and Inflammation in Hepatic Diseases: Therapeutic Possibilities of N-Acetylcysteine. Int J Mol Sci 2015; 16:30269-308. [PMID: 26694382 PMCID: PMC4691167 DOI: 10.3390/ijms161226225] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 12/02/2015] [Accepted: 12/04/2015] [Indexed: 12/12/2022] Open
Abstract
Liver disease is highly prevalent in the world. Oxidative stress (OS) and inflammation are the most important pathogenetic events in liver diseases, regardless the different etiology and natural course. N-acetyl-l-cysteine (the active form) (NAC) is being studied in diseases characterized by increased OS or decreased glutathione (GSH) level. NAC acts mainly on the supply of cysteine for GSH synthesis. The objective of this review is to examine experimental and clinical studies that evaluate the antioxidant and anti-inflammatory roles of NAC in attenuating markers of inflammation and OS in hepatic damage. The results related to the supplementation of NAC in any form of administration and type of study are satisfactory in 85.5% (n = 59) of the cases evaluated (n = 69, 100%). Within this percentage, the dosage of NAC utilized in studies in vivo varied from 0.204 up to 2 g/kg/day. A standard experimental design of protection and treatment as well as the choice of the route of administration, with a broader evaluation of OS and inflammation markers in the serum or other biological matrixes, in animal models, are necessary. Clinical studies are urgently required, to have a clear view, so that, the professionals can be sure about the effectiveness and safety of NAC prescription.
Collapse
|
26
|
Khayyat A, Tobwala S, Hart M, Ercal N. N-acetylcysteine amide, a promising antidote for acetaminophen toxicity. Toxicol Lett 2015; 241:133-42. [PMID: 26602168 DOI: 10.1016/j.toxlet.2015.11.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/27/2015] [Accepted: 11/08/2015] [Indexed: 12/20/2022]
Abstract
Acetaminophen (N-acetyl-p-aminophenol, APAP) is one of the most widely used over the counter antipyretic and analgesic medications. It is safe at therapeutic doses, but its overdose can result in severe hepatotoxicity, a leading cause of drug-induced acute liver failure in the USA. Depletion of glutathione (GSH) is one of the initiating steps in APAP-induced hepatotoxicity; therefore, one strategy for restricting organ damage is to restore GSH levels by using GSH prodrugs. N-acetylcysteine (NAC), a GSH precursor, is the only currently approved antidote for an acetaminophen overdose. Unfortunately, fairly high doses and longer treatment times are required due to its poor bioavailability. In addition, oral and I.V. administration of NAC in a hospital setting are laborious and costly. Therefore, we studied the protective effects of N-acetylcysteine amide (NACA), a novel antioxidant with higher bioavailability, and compared it with NAC in APAP-induced hepatotoxicity in C57BL/6 mice. Our results showed that NACA is better than NAC at a low dose (106mg/kg) in preventing oxidative stress and protecting against APAP-induced damage. NACA significantly increased GSH levels and the GSH/GSSG ratio in the liver to 66.5% and 60.5% of the control, respectively; and it reduced the level of ALT by 30%. However, at the dose used, NAC was not effective in combating the oxidative stress induced by APAP. Thus, NACA appears to be better than NAC in reducing the oxidative stress induced by APAP. It would be of great value in the health care field to develop drugs like NACA as more effective and safer options for the prevention and therapeutic intervention in APAP-induced toxicity.
Collapse
Affiliation(s)
- Ahdab Khayyat
- Chemistry Department, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - Shakila Tobwala
- Chemistry Department, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - Marcia Hart
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Nuran Ercal
- Chemistry Department, Missouri University of Science and Technology, Rolla, MO 65409, USA.
| |
Collapse
|
27
|
Mohammed NEM, Messiha BAS, Abo-Saif AA. Effect of amlodipine, lisinopril and allopurinol on acetaminophen-induced hepatotoxicity in rats. Saudi Pharm J 2015; 24:635-644. [PMID: 27829805 PMCID: PMC5094429 DOI: 10.1016/j.jsps.2015.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 04/30/2015] [Indexed: 12/09/2022] Open
Abstract
Background Exposure to chemotherapeutic agents such as acetaminophen may lead to serious liver injury. Calcium deregulation, angiotensin II production and xanthine oxidase activity are suggested to play mechanistic roles in such injury. Objective This study evaluates the possible protective effects of the calcium channel blocker amlodipine, the angiotensin converting enzyme inhibitor lisinopril, and the xanthine oxidase inhibitor allopurinol against experimental acetaminophen-induced hepatotoxicity, aiming to understand its underlying hepatotoxic mechanisms. Material and methods Animals were allocated into a normal control group, a acetaminophen hepatotoxicity control group (receiving a single oral dose of acetaminophen; 750 mg/kg/day), and four treatment groups receive N-acetylcysteine (300 mg/kg/day; a reference standard), amlodipine (10 mg/kg/day), lisinopril (20 mg/kg/day) and allopurinol (50 mg/kg/day) orally for 14 consecutive days prior to acetaminophen administration. Evaluation of hepatotoxicity was performed by the assessment of hepatocyte integrity markers (serum transaminases), oxidative stress markers (hepatic malondialdehyde, glutathione and catalase), and inflammatory markers (hepatic myeloperoxidase and nitrate/nitrite), in addition to a histopathological study. Results Rats pre-treated with amlodipine, lisinopril or allopurinol showed significantly lower serum transaminases, significantly lower hepatic malondialdehyde, myeloperoxidase and nitrate/nitrite, as well as significantly higher hepatic glutathione and catalase levels, compared with acetaminophen control rats. Serum transaminases were normalized in the lisinopril treatment group, while hepatic myeloperoxidase was normalized in the all treatment groups. Histopathological evaluation strongly supported the results of biochemical estimations. Conclusion Amlodipine, lisinopril or allopurinol can protect against acetaminophen-induced hepatotoxicity, showing mechanistic roles of calcium channels, angiotensin converting enzyme and xanthine oxidase enzyme in the pathogenesis of hepatotoxicity induced by acetaminophen.
Collapse
Affiliation(s)
- Nesreen E M Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Sueif, Egypt
| | - Basim A S Messiha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni Sueif University, Beni-Sueif, Egypt
| | - Ali A Abo-Saif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Sueif, Egypt
| |
Collapse
|