1
|
Tan S, Ding Y, Wang W, Rao J, Cheng F, Zhang Q, Xu T, Hu T, Hu Q, Ye Z, Yan X, Wang X, Li M, Xie P, Chen Z, Liang G, Pu Y, Zhang J, Gu Z. Development of an AI model for DILI-level prediction using liver organoid brightfield images. Commun Biol 2025; 8:886. [PMID: 40483291 PMCID: PMC12145446 DOI: 10.1038/s42003-025-08205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 05/12/2025] [Indexed: 06/11/2025] Open
Abstract
AI image processing techniques hold promise for clinical applications by enabling analysis of complex status information from cells. Importantly, real-time brightfield imaging has advantages of informativeness, non-destructive nature, and low cost over fluorescence imaging. Currently, human liver organoids (HLOs) offer an alternative to animal models due to their excellent physiological recapitulation including basic functions and drug metabolism. Here we show a drug-induced liver injury (DILI) level prediction model using HLO brightfield images (DILITracer) considering that DILI is the major causes of drug withdrawals. Specifically, we utilize BEiT-V2 model, pretrained on 700,000 cell images, to enhance 3D feature extraction. A total of 30 compounds from FDA DILIrank are selected (classified into Most-, Less-, and No-DILI) to activate HLOs and corresponding brightfield images are collected at different time series and z-axis. Our computer vision model based on image-spatial-temporal coding layer excavates fully spatiotemporal information of continuously captured images, links HLO morphology with DILI severity, and final output DILI level of compounds. DILITracer achieves an overall accuracy of 82.34%. To our knowledge, this is the first model to output ternary classification of hepatotoxicity. Overall, DILITracer, using clinical data as an endpoint categorization label, offers a rapid and effective approach for screening hepatotoxic compounds.
Collapse
Affiliation(s)
- Shiyi Tan
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Yan Ding
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Wei Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Jianhua Rao
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Feng Cheng
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Qiuyin Zhang
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Tingting Xu
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Tianmu Hu
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Qinyi Hu
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Ziliang Ye
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xiaopeng Yan
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Xiaowei Wang
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China
| | - Mingyue Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Peng Xie
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zaozao Chen
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China.
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China.
| | - Zhongze Gu
- Jiangsu Institute for Sport and Health (JISH), Nanjing, China.
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Watanabe M, Salvadori A, Markovic M, Sudo R, Ovsianikov A. Advanced liver-on-chip model mimicking hepatic lobule with continuous microvascular network via high-definition laser patterning. Mater Today Bio 2025; 32:101643. [PMID: 40206147 PMCID: PMC11979415 DOI: 10.1016/j.mtbio.2025.101643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/09/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
There is a great demand for development of advanced in vitro liver models to predict the efficacy and safety of drug candidates accurately in the preclinical drug development. Despite the great efforts to develop biomimetic models, it remains challenging to precisely mimic a functional unit of the liver (i.e., hepatic lobule) with a continuous microvascular network. Recent progress in laser patterning has allowed us to create arbitrary biomimetic structures with high resolution. Here, we propose an advanced liver-on-chip model mimicking the hepatic lobule with a continuous microvascular network, ranging from the microvessels to the central vein of the liver, utilizing femtosecond laser patterning. Firstly, we optimize the laser power to pattern microchannels mimicking the microvessel and central vein of the hepatic lobule by using a femtosecond laser within a collagen-based hydrogel containing hepatic cells. Secondly, we construct continuous microvessels with luminal structures by comparing different microchannel sizes in diameter. Finally, we assemble a millimeter-scale hepatic lobule-like structure with multiple layers of microvascular networks in the liver-on-chip. Furthermore, our liver-on-chip model exhibits major liver functions and drug-induced hepatotoxicity, as evidenced by albumin and urea productions and by a toxic response to acetaminophen, respectively. Our approach provides valuable strategies for the development of advanced physiological and pathological liver-on-chip models for pharmaceutical and toxicological studies.
Collapse
Affiliation(s)
- Masafumi Watanabe
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
- Japan Society for the Promotion of Science (JSPS) Overseas Research Fellow, Japan
| | - Alice Salvadori
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| | - Marica Markovic
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| | - Ryo Sudo
- Department of System Design Engineering, Keio University, 223-8522 Yokohama, Japan
| | - Aleksandr Ovsianikov
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| |
Collapse
|
3
|
Skat-Rørdam J, Lykkesfeldt J, Gluud LL, Tveden-Nyborg P. Mechanisms of drug induced liver injury. Cell Mol Life Sci 2025; 82:213. [PMID: 40418327 DOI: 10.1007/s00018-025-05744-3] [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: 01/28/2025] [Revised: 04/23/2025] [Accepted: 05/07/2025] [Indexed: 05/27/2025]
Abstract
Drug induced liver injury (DILI) is a serious and potentially life-threatening condition resulting from an adverse drug reaction. Both the clinical manifestations and pathological mechanisms of DILI vary depending on drug characteristics, dose, duration of exposure as well as host specific factors. Disease onset can occur within days or months after the introduction of a drug. This has challenged identification of disease specific biomarkers and resulted in delayed and even erroneous diagnosis of patients. Apart from discontinuation of current pharmacotherapy, options for DILI patients are scarce and the condition can sometimes continue or worsen after drugs are discontinued or result in irreversible liver damage such as cirrhosis. This illustrates the need to uncover relevant pathological pathways that will pave the road for targeted interventions. In an effort to accommodate these needs, novel insights from preclinical and cellular disease modeling have allowed coupling of specific drugs to potential mechanisms of toxicity. This review outlines three signaling pathways of DILI: organelle stress, cholestasis, and immune responses, discusses their interplay with oxidative stress, and provides examples of drugs specifically targeting one or more steps in these pathways. A systematic approach identifying specific mechanisms of DILI could allow for the assembly of large databases, in turn enabling advanced computational modelling to provide accurate predictions of the DILI potential of both known drugs and future drug candidates.
Collapse
Affiliation(s)
- J Skat-Rørdam
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - J Lykkesfeldt
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - L L Gluud
- Gastro Unit, Hvidovre Hospital, Hvidovre, Denmark
| | - P Tveden-Nyborg
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Chen Y, Jiang G, Huang Y, Song S, Fu H, Du B, Xiao Y, Li P, Shi K, Huang Y, Song Q, Gao X, Xie Q. Lipoprotein-Based Nanocatalyst Enables Targeted Treatment of APAP-Induced Liver Injury via Enhanced Macropinocytosis. Adv Healthc Mater 2025:e2500507. [PMID: 40411876 DOI: 10.1002/adhm.202500507] [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: 01/29/2025] [Revised: 05/09/2025] [Indexed: 05/26/2025]
Abstract
Drug-induced liver injury (DILI), predominantly caused by acetaminophen (APAP) overdose, is characterized by excessive reactive oxygen species (ROS) production and subsequent hepatocyte necrosis. Although N-acetylcysteine (NAC) remains the only approved treatment, its effectiveness is limited by a narrow therapeutic time window and reduced efficacy in advanced cases. To address these limitations, an innovative therapeutic approach is developed utilizing ceria's antioxidant properties. In this study, a reconstituted high-density lipoprotein-encapsulated ceria nanocatalyst (CeO2-rHDL) is engineered to overcome the aggregation tendency and targeting limitation of naked ceria nanoparticles. These findings revealed that CeO2-rHDL enters hepatocytes through macropinocytosis, a process synergistically enhanced by both APAP and NAC, facilitating efficient liver targeting. The nanocatalyst demonstrated remarkable therapeutic efficacy by restoring mitochondrial function through ROS reduction. When combined with NAC, CeO2-rHDL significantly improved survival outcomes in DILI mice. This lipoprotein-based nanocatalyst system represents a promising therapeutic strategy for DILI treatment, offering enhanced targeting capabilities and improved therapeutic efficacy.
Collapse
Affiliation(s)
- Yaoxing Chen
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road Number Two, Shanghai, 200025, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yan Huang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road Number Two, Shanghai, 200025, China
| | - Shuying Song
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road Number Two, Shanghai, 200025, China
| | - Haoshuang Fu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road Number Two, Shanghai, 200025, China
| | - Bingying Du
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road Number Two, Shanghai, 200025, China
| | - Yuelin Xiao
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road Number Two, Shanghai, 200025, China
| | - Peiying Li
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Kexin Shi
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yukun Huang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road Number Two, Shanghai, 200025, China
| |
Collapse
|
5
|
Elblová P, Anthi J, Liu M, Lunova M, Jirsa M, Stephanopoulos N, Lunov O. DNA Nanostructures for Rational Regulation of Cellular Organelles. JACS AU 2025; 5:1591-1616. [PMID: 40313805 PMCID: PMC12042030 DOI: 10.1021/jacsau.5c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 05/03/2025]
Abstract
DNA nanotechnology has revolutionized materials science and biomedicine by enabling precise manipulation of matter at the nanoscale. DNA nanostructures (DNs) in particular represent a promising frontier for targeted therapeutics. Engineered DNs offer unprecedented molecular programmability, biocompatibility, and structural versatility, making them ideal candidates for advanced drug delivery, organelle regulation, and cellular function modulation. This Perspective explores the emerging role of DNs in modulating cellular behavior through organelle-targeted interventions. We highlight current advances in nuclear, mitochondrial, and lysosomal targeting, showcasing applications ranging from gene delivery to cancer therapeutics. For instance, DNs have enabled precision mitochondrial disruption in cancer cells, lysosomal pH modulation to enhance gene silencing, and nuclear delivery of gene-editing templates. While DNs hold immense promise for advancing nanomedicine, outstanding challenges include optimizing biological interactions and addressing safety concerns. This Perspective highlights the current potential of DNs for rational control of targeted organelles, which could lead to novel therapeutic strategies and advancement of precision nanomedicines in the future.
Collapse
Affiliation(s)
- Petra Elblová
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- Faculty
of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16 Prague 2, Czech Republic
| | - Judita Anthi
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Minghui Liu
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Mariia Lunova
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- Institute
for Clinical & Experimental Medicine (IKEM), 14 021 Prague, Czech Republic
| | - Milan Jirsa
- Institute
for Clinical & Experimental Medicine (IKEM), 14 021 Prague, Czech Republic
| | - Nicholas Stephanopoulos
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Oleg Lunov
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
| |
Collapse
|
6
|
Liao Y, Chai D, Wang Q, Wang X, Yong Q, Cheng Z, Zhang C, Zhang D, Liu B, Liu R, Li Z. Sensor-combined organ-on-a-chip for pharmaceutical and medical sciences: from design and materials to typical biomedical applications. MATERIALS HORIZONS 2025; 12:2161-2178. [PMID: 39801302 DOI: 10.1039/d4mh01174k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Organ-on-a-chip (OoC) is a breakthrough technology in biomedicine. As microphysiological systems constructed in vitro, OoCs can simulate the main structures and functions of human organs, thereby providing a powerful tool for drug screening and disease model construction. Furthermore, the coupling of OoCs and sensors has been an innovative discovery in the field of biomedical and electronic engineering in recent years. The integration of sensors into OoCs allows the real-time monitoring of the changes in the microenvironmental parameters within the chip, reflecting the physiological responses of cells or tissues in the OoC and providing more accurate data support for drug development and disease treatment. In this work, we briefly outline the design ideas of OoCs, summarize the commonly used materials for OoCs and their advantages and disadvantages, and provide the most recent practical examples of the combination of OoCs and sensors in pharmaceutical and medical sciences. Furthermore, perspectives, challenges and their solutions in the future development of this technology are provided, with the aim to inspire the researchers to work toward the subsequent development of OoCs having improved reliability.
Collapse
Affiliation(s)
- Yumeng Liao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Danni Chai
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Quan Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xueqi Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qian Yong
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhaoming Cheng
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Chuanjun Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Di Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Boshi Liu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Rui Liu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| |
Collapse
|
7
|
Li J, Yang X, Lian X, Li B, Ma Q, Yang L, Gao G, Deng Y, Yang Z. Licorice attenuates cisplatin-induced hepatotoxicity by alleviating endoplasmic reticulum stress and apoptosis. Front Pharmacol 2025; 16:1557125. [PMID: 40183095 PMCID: PMC11965583 DOI: 10.3389/fphar.2025.1557125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
Cisplatin (CP), a widely used antineoplastic drug, could induce hepatotoxicity and is also one of the most common reasons for drug-induced liver injury (DILI). Licorice (Chinese name GanCao, GC) is a commonly used herbal drug in traditional Chinese medicine (TCM) that has been shown to treat liver diseases and DILI. CP has been documented to induce apoptosis through the promotion of endoplasmic reticulum (ER) stress. However, the exact role of ER stress in the pathogenesis of CP-induced hepatotoxicity remains unclear. A rat DILI model was constructed through intraperitoneal injection of CP, and the anti-DILI effect of GC was detected by liver coefficients, liver function tests, pathological staining, and oxidative stress indices. Additionally, the ER stress and apoptosis indices were investigated by quantitative real-time PCR (qRT-PCR), Western blotting, and immunofluorescence (IF) on CP-induced toxicity in rat liver tissues and LO2 cells. In the model group, liver function indicators significantly elevated, liver lesions more pronounced, and the reactive oxygen species (ROS) level in the liver increased, the expression of ER stress markers, apoptosis factors, and indicators related to the protein kinase RNA-like ER kinase/activating transcription factor 4/C/EBP homologous protein (PERK/ATF4/CHOP) pathway significantly elevated. Treatment of the CP-induced toxicity in the rat model with GC significantly improved liver function, reduced liver lesions, decreased liver ROS. In addition, GC significantly inhibited the expression of ER stress markers, apoptosis factors, and indicators related to PERK/ATF4/CHOP pathway, demonstrating the anti-CP-induced hepatotoxicity effect of GC. In this study, we verified the protective effect of GC in CP-induced hepatotoxicity in rats and clarified its mechanisms related to ER stress and apoptosis.
Collapse
Affiliation(s)
- Jie Li
- School of Pharmaceutical Science, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Xiujuan Yang
- School of Pharmaceutical Science, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaolong Lian
- Department of pharmacy, Qinghai University Medical College, Xining, China
| | - Baojian Li
- Shaanxi University of Chinese Medicine, Xi’an, China
| | - Quhuan Ma
- School of Pharmaceutical Science, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Lingling Yang
- School of Pharmaceutical Science, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Guangmiao Gao
- School of Pharmaceutical Science, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Yi Deng
- School of Pharmaceutical Science, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Zhijun Yang
- School of Pharmaceutical Science, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
8
|
Lai S, Ye Y, Ding Q, Hu X, Fu A, Wu L, Cao W, Liu Q, Dou X, Qi X. Thonningianin A ameliorates acetaminophen-induced liver injury by activating GPX4 and modulating endoplasmic reticulum stress. Front Pharmacol 2025; 16:1531277. [PMID: 40051561 PMCID: PMC11882853 DOI: 10.3389/fphar.2025.1531277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
Introduction Acetaminophen (APAP) is widely used as an analgesic and antipyretic. However overdose APAP can lead to acute liver injury (ALI), representing a significant challenge for public health due to limited treatment options. Current research highlights the need for safer and more effective therapies for APAP-induced liver injury, especially those that target oxidative and endoplasmic reticulum (ER) stress pathways. This study investigates the protective effects of Thonningianin A (TA), a flavonoid compound derived from Penthorum chinense Pursh, in mitigating APAP-induced hepatotoxicity. Methods The experimental design involved administering TA at doses of 20 mg/kg and 40 mg/kg to C57BL/6 mice prior to inducing hepatotoxicity with APAP. Results and discussion TA treatment significantly lowered plasma ALT and AST levels, inhibited the production of inflammatory cytokines, and reduced oxidative stress markers in liver tissues. Furthermore, TA modulated apoptosis-related proteins by increasing BCL-2 expression while decreasing CHOP and BAX levels. It alleviated endoplasmic reticulum (ER) stress by downregulating GRP78, p-PERK, and ATF4. Notably, liver-specific GPX4 knockdown, achieved through AAV-8-mediated shRNA delivery, abolished the hepatoprotective effects of TA, underscoring GPX4's essential role in mediating TA-induced hepatoprotection. These findings suggest TA as a promising therapeutic agent in managing APAP-induced liver injury, with its unique action on both oxidative and ER stress pathways contributing to its hepatoprotective efficacy.
Collapse
Affiliation(s)
- Shanglei Lai
- Department of Medical Research Center, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Yingyan Ye
- Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, China
| | - Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaokai Hu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ai Fu
- Institute of Hepatology and Epidemiology, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lan Wu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wenjing Cao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qingsheng Liu
- Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
9
|
Li J, Liu H, Jia Y, Tuniyazi X, Liao X, Zhao J, Du Y, Fang Z, Lü G. SW033291 promotes liver regeneration after acetaminophen-induced liver injury in mice. Biochem Biophys Res Commun 2025; 749:151365. [PMID: 39855045 DOI: 10.1016/j.bbrc.2025.151365] [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: 12/25/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025]
Abstract
Acetaminophen (APAP) is a commonly utilized antipyretic and analgesic drug. Overdose of APAP is a primary contributor to drug-induced liver injury and acute liver failure (ALF). SW033291 has been shown to play a role in tissue regeneration in various diseases; however, its potential to facilitate liver regeneration following APAP-induced hepatic injury remains unexamined. Thus, this study focused on exploring the therapeutic impacts and mechanisms of SW033291 on liver damage by establishing models of APAP-induced acute liver injury in mice. The results showed that treatment with SW033291 reduces serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, decreases the area of hepatic necrosis, increases glutathione (GSH) levels, and decreases tissue malondialdehyde (MDA) content, as well as the expression levels of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in mice with liver injury. It could also promote hepatocyte proliferation and inhibit apoptosis by increasing tissue prostaglandin E2 (PGE2) levels. In conclusion, SW033291 demonstrates the capacity to ameliorate APAP-induced hepatic injury in mice by fostering liver regeneration, attenuating oxidative stress, and modulating inflammatory responses, thereby presenting itself as a promising candidate for the development of therapeutic interventions targeting acute liver failure.
Collapse
Affiliation(s)
- Jing Li
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Hui Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Yutong Jia
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Xiayidanmu Tuniyazi
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Xia Liao
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Jinlong Zhao
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Yun Du
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Ziyi Fang
- College of Life Sciences and Technology, Xinjiang University, Urumqi, 830054, Xinjiang, China
| | - Guodong Lü
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
10
|
Elblová P, Andělová H, Lunova M, Anthi J, Henry SJW, Tu X, Dejneka A, Jirsa M, Stephanopoulos N, Lunov O. Geometrically constrained cytoskeletal reorganisation modulates DNA nanostructures uptake. J Mater Chem B 2025; 13:2335-2351. [PMID: 39835937 PMCID: PMC11749194 DOI: 10.1039/d5tb00074b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
DNA nanostructures (DNs) have gained popularity in various biomedical applications due to their unique properties, including structural programmability, ease of synthesis and functionalization, and low cytotoxicity. Effective utilization of DNs in biomedical applications requires a fundamental understanding of their interactions with living cells and the mechanics of cellular uptake. Current knowledge primarily focuses on how the physicochemical properties of DNs, such as mass, shape, size, and surface functionalization, affect uptake efficacy. However, the role of cellular mechanics and morphology in DN uptake remains largely unexplored. In this work, we show that cells subjected to geometric constraints remodel their actin cytoskeleton, resulting in differential mechanical force generation that facilitates DN uptake. The length, number, and orientation of F-actin fibers are influenced by these constraints, leading to distinct mechanophenotypes. Overall, DN uptake is governed by F-actin forces arising from filament reorganisation under geometric constraints. These results underscore the importance of actin dynamics in the cellular uptake of DNs and suggest that leveraging geometric constraints to induce specific cell morphology adaptations could enhance the uptake of therapeutically designed DNs.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18200, Czech Republic.
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, CZ-121 16 Prague 2, Czech Republic
| | - Hana Andělová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18200, Czech Republic.
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18200, Czech Republic.
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Judita Anthi
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18200, Czech Republic.
| | - Skylar J W Henry
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, USA.
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, USA
| | - Xinyi Tu
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, USA.
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, USA
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18200, Czech Republic.
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Nicholas Stephanopoulos
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, USA.
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, USA
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18200, Czech Republic.
| |
Collapse
|
11
|
Yang Y, Chen Q, Liu Z, Huang T, Hong Y, Li N, Ai K, Huang Q. Novel reduced heteropolyacid nanoparticles for effective treatment of drug-induced liver injury by manipulating reactive oxygen and nitrogen species and inflammatory signals. J Colloid Interface Sci 2025; 678:174-187. [PMID: 39243718 DOI: 10.1016/j.jcis.2024.08.239] [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: 06/14/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
With the rapid advancements in biomedicine, the use of clinical drugs has surged sharply. However, potential hepatotoxicity limits drug exploitation and widespread usage, posing serious threats to patient health. Hepatotoxic drugs disrupt liver enzyme levels and cause refractory pathological damage, creating a challenge in the application of diverse first-line drugs. The activation and deterioration of reactive oxygen and nitrogen species (RONS) and inflammatory signals are key pathological mechanisms of drug-induced liver injury (DILI). Herein, a novel reduced heteropolyacid nanoparticle (RNP) has been developed, possessing high RONS-scavenging ability, strong anti-inflammatory activity, and excellent biosafety. These features enable it to swiftly restore the redox and immune balance of the liver. Intravenous administration of RNP effectively scavenged RONS storm, reversing liver oxidative stress and restoring normal mitochondrial membrane potential and function. Furthermore, by inhibiting c-Jun-N-terminal kinase phosphorylation, RNP facilitated the restoration of nuclear factor erythroid 2-related factor 2-mediated endogenous antioxidant signaling, ultimately rescuing the liver function and tissue morphology in acetaminophen-induced DILI mice. Crucially, the high biocompatible RNP exhibited superior efficacy in the DILI mouse model compared to the clinical antioxidant N-acetylcysteine. This targeted therapeutic approach, tailored to address the onset and progression of DILI, offers valuable new insights into controlling the condition and restoring liver structure and function.
Collapse
Affiliation(s)
- Yongqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Zerun Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ting Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ying Hong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Niansheng Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
12
|
Livoti LA, Sison-Young R, Reddyhoff D, Fisher CP, Gardner I, Diaz-Nieto R, Goldring CE, Copple IM. Limitations of acetaminophen as a reference hepatotoxin for the evaluation of in vitro liver models. Toxicol Sci 2025; 203:35-40. [PMID: 39509272 DOI: 10.1093/toxsci/kfae133] [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] [Indexed: 11/15/2024] Open
Abstract
Acetaminophen is commonly used as a reference hepatotoxin to demonstrate that in vitro human liver platforms can emulate features of clinical drug-induced liver injury. However, the induction of substantial cell death in these models typically requires acetaminophen concentrations (∼10 mM) far higher than blood concentrations of the drug associated with clinical hepatotoxicity (∼1 mM). Using the cytochrome P450 inhibitor 1-aminobenzotriazole, we show that acetaminophen toxicity in cultured human, mouse, and rat hepatocytes is not dependent on N-acetyl-p-benzoquinonimine formation, unlike the in vivo setting. This finding highlights the limitation of using acetaminophen as a reference hepatotoxin for the evaluation of in vitro liver models. Hence, we make recommendations on the selection of reference hepatotoxins for this purpose.
Collapse
Affiliation(s)
- Lucia A Livoti
- Department of Pharmacology & Therapeutics, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| | - Rowena Sison-Young
- Department of Pharmacology & Therapeutics, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| | - Dennis Reddyhoff
- Certara Predictive Technologies, Sheffield, S1 2BJ, United Kingdom
| | - Ciarán P Fisher
- Certara Predictive Technologies, Sheffield, S1 2BJ, United Kingdom
- GSK Research and Development Ltd, Stevenage, SG1 2NY, United Kingdom
| | - Iain Gardner
- Certara Predictive Technologies, Sheffield, S1 2BJ, United Kingdom
| | - Rafael Diaz-Nieto
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, L7 8YE, United Kingdom
| | - Christopher E Goldring
- Department of Pharmacology & Therapeutics, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| | - Ian M Copple
- Department of Pharmacology & Therapeutics, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| |
Collapse
|
13
|
Lei P, Li X, Jiang L, Yu H, Zhang P, Han L, Jiang M. Alisma plantago-aquatica polysaccharides ameliorate acetaminophen-induced acute liver injury by regulating hepatic metabolic profiles and modulating gut microbiota. Int J Biol Macromol 2024; 285:138345. [PMID: 39631232 DOI: 10.1016/j.ijbiomac.2024.138345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Acetaminophen (APAP) has emerged as a predominant contributor to acute liver failure (ALF) in United States. Alismatis rhizoma, a commonly used traditional herbal medicine, contains small molecular components with extensive hepatoprotective activity. However, the specific role of Alismatis rhizoma polysaccharide (ARP) in liver protection remains unclear. ARP50 and ARP70, derived through graded alcohol precipitation and refinement, predominantly consisted of varying proportions of glucose, galactose, and arabinose. In vitro experiments on free radical scavenging demonstrated notable antioxidant capabilities of ARP50 and ARP70. To investigate the hepatoprotective effects, an APAP-induced acute liver injury (ALI) model was established in mice. ARP50 and ARP70 exerted dose-dependent therapeutic effects on APAP-induced liver injury. Further analysis of liver metabolites revealed that ARPs facilitated the reconstruction of the liver antioxidant system by modulating the metabolism network centered on l-glutamine. In addition, the abundance of gut microbiota was altered under the influence of ARPs. ARP50 significantly reduced the levels of Pseudarthrobacter and markedly increased the levels of Faecalibacterium,At the same time, ARP50 could increase the levels of acetic acid in the liver and serum. Meanwhile, ARP70 significantly increased the abundance of Dubosiella, Muribaculum, Ileibacterium, and Prevotellaceae UCG 001, while reducing the abundance of Escherichia Shigella and Pseudarthrobacter. The results indicated that ARPs could exert a protective effect against APAP-induced acute liver injury by reshaping the liver metabolic profile and modulating the gut microbiota.
Collapse
Affiliation(s)
- Peng Lei
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoge Li
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lei Jiang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Heshui Yu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peng Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lifeng Han
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miaomiao Jiang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
14
|
Luo Q, Li X, Huang J, Zhao L, Liu L, Huang S, Xu Y, Qiu P, Li C. Shenqi Pill alleviates acetaminophen-induced liver injury: a comprehensive strategy of network pharmacology and spectrum-effect relationship reveals mechanisms and active components. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156050. [PMID: 39303509 DOI: 10.1016/j.phymed.2024.156050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Acetaminophen (APAP), commonly used for its antipyretic and analgesic properties, can cause severe liver injury or even acute liver failure when overdosed. However, the options for treating APAP-induced liver toxicity are limited. Shenqi Pill (SQP), a traditional Chinese herbal formula, has shown effectiveness in treating various liver ailments. SQP consists of cinnamon, aconite, rehmannia, cornus, peony bark, Chinese yam, poria, and alisma in a ratio of 1:1:8:4:3:4:3:3. However, the mechanisms and active components of SQP that counteract drug-induced liver injury (DILI) are not well understood. PURPOSE This study aimed to explore the protective effects of SQP against APAP-induced liver injury in both laboratory and animal settings. It seeks to identify the active components and potential mechanisms by which SQP targets mitochondria to alleviate liver damage. METHODS A mouse model with APAP-induced liver injury was established to assess SQP's therapeutic impact. This study then analyzed the components of SQP using UPLC-Q-TOF-MS in both in vivo and in vitro environments. Network pharmacology and the GEO database helped predict potential pathways and targets. Potential active components were identified through spectrum-effect relationship analysis and validated their efficacy using Seahorse assays and molecular docking. RESULTS Treatment with SQP significantly reduced liver dysfunction, tissue damage, lipid metabolic disruptions, and inflammation caused by APAP in mice. In cellular tests, SQP-treated serum notably enhanced mitochondrial function, maintained membrane potential, decreased ROS levels, and prevented mitochondrial permeability transition pore opening. Biochemically, SQP reversed the suppression of p-AMPK, p-ACC, CPT1, and ACADM expression caused by APAP overdose. This study identified 97 in vitro and 24 in vivo components of SQP, with eight showing significant mitochondrial benefits. Molecular docking studies suggest that fuziline and paeoniflorin could activate AMPK. CONCLUSION SQP effectively mitigates APAP-induced liver injury by enhancing mitochondrial function via the AMPK-ACC-CPT1-ACADM pathway. Moreover, this study introduces a novel strategy for analyzing the relationship between the chemical and pharmacological properties of drug-containing serum, successfully identifying compounds with mitochondrial activity. Fuziline and paeoniflorin, in particular, emerge as promising mitochondrial protectants and warrant further investigation. This research underpins the development of innovative treatments for DILI using SQP and its components.
Collapse
Affiliation(s)
- Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Junhao Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lisha Zhao
- Analytical Testing Center, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Liu Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueling Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Qiu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
15
|
Kong D, Huang Y, Song B, Zhang X, Yuan J. Novel Endoplasmic Reticulum-Targeted Luminescent Probe for Visualization of Carbon Monoxide in Drug-Induced Liver Injury. Anal Chem 2024; 96:18246-18253. [PMID: 39491487 DOI: 10.1021/acs.analchem.4c04528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Drug-induced liver injury (DILI) is a major hepatic dysfunction commonly caused by hepatotoxic drug overdose, resulting in a considerable number of fatalities worldwide. Recent studies have highlighted the regulatory and hepatoprotective effects of carbon monoxide (CO) during the liver injury process. However, precisely tracking the dynamic changes in the composition of CO in DILI is still a great challenge. In this work, leveraging the innovative "quencher-insertion" strategy, a unique endoplasmic reticulum (ER)-targetable lanthanide complex-based luminescence probe, ER-ANBTTA-Eu3+/Tb3+, has been developed for the selective and accurate monitoring of CO fluxes in live cells and laboratory animals. The new probe is composed of three covalently linked functional moieties: the terpyridine polyacid-Eu3+/Tb3+-mixed chelates as the long-lived luminophore, a p-toluenesulfonamide moiety as the ER-anchoring motif, and an allyloxy-nitrobenzyl ether moiety as the CO-specific recognition unit. Upon reaction with CO in the presence of Pd2+ ions, the Tsuji-Trost reaction leads to the cleavage of the allyloxy-nitrobenzyl group from the Eu3+/Tb3+-mixed chelates, which results in the restoration of Tb3+ emission at 538 nm and the attenuation of Eu3+ emission at 688 nm, leading to a dramatic increase of the I538/I688 ratio. In addition to the exceptional response sensitivity and selectivity toward CO, ER-ANBTTA-Eu3+/Tb3+ also exhibits the outstanding ER-locating capability, which allows the probe to be used for imaging of CO in the ER of live cells. Using this probe, combined with the time-gated luminescence imaging mode, the exogenous and endogenous CO in ER of live cells were monitored without the interference of background autofluorescence. Moreover, the upregulation of hepatic CO in DILI mice was successfully visualized. The results suggested the potential of ER-ANBTTA-Eu3+/Tb3+ for deeply exploring the functions of CO in DILI pathogenesis.
Collapse
Affiliation(s)
- Deshu Kong
- School of Chemistry, Dalian University of Technology, Dalian 116024, China
| | - Yundi Huang
- School of Chemistry, Dalian University of Technology, Dalian 116024, China
| | - Bo Song
- School of Chemistry, Dalian University of Technology, Dalian 116024, China
| | - Xinyue Zhang
- School of Chemistry, Dalian University of Technology, Dalian 116024, China
| | - Jingli Yuan
- College of Life Science, Dalian Minzu University, Dalian 116600, China
| |
Collapse
|
16
|
Guo K, van den Beucken T. Advances in drug-induced liver injury research: in vitro models, mechanisms, omics and gene modulation techniques. Cell Biosci 2024; 14:134. [PMID: 39488681 PMCID: PMC11531151 DOI: 10.1186/s13578-024-01317-2] [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: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Drug-induced liver injury (DILI) refers to drug-mediated damage to the structure and function of the liver, ranging from mild elevation of liver enzymes to severe hepatic insufficiency, and in some cases, progressing to liver failure. The mechanisms and clinical symptoms of DILI are diverse due to the varying combination of drugs, making clinical treatment and prevention complex. DILI has significant public health implications and is the primary reason for post-marketing drug withdrawals. The search for reliable preclinical models and validated biomarkers to predict and investigate DILI can contribute to a more comprehensive understanding of adverse effects and drug safety. In this review, we examine the progress of research on DILI, enumerate in vitro models with potential benefits, and highlight cellular molecular perturbations that may serve as biomarkers. Additionally, we discuss omics approaches frequently used to gather comprehensive datasets on molecular events in response to drug exposure. Finally, three commonly used gene modulation techniques are described, highlighting their application in identifying causal relationships in DILI. Altogether, this review provides a thorough overview of ongoing work and approaches in the field of DILI.
Collapse
Affiliation(s)
- Kaidi Guo
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands.
| | - Twan van den Beucken
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
17
|
Yang JW, Khorsandi D, Trabucco L, Ahmed M, Khademhosseini A, Dokmeci MR, Ye JY, Jucaud V. Liver-on-a-Chip Integrated with Label-Free Optical Biosensors for Rapid and Continuous Monitoring of Drug-Induced Toxicity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403560. [PMID: 39212623 PMCID: PMC11602353 DOI: 10.1002/smll.202403560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Drug toxicity assays using conventional 2D static cultures and animal studies have limitations preventing the translation of potential drugs to the clinic. The recent development of organs-on-a-chip platforms provides promising alternatives for drug toxicity/screening assays. However, most studies conducted with these platforms only utilize single endpoint results, which do not provide real-time/ near real-time information. Here, a versatile technology is presented that integrates a 3D liver-on-a-chip with a label-free photonic crystal-total internal reflection (PC-TIR) biosensor for rapid and continuous monitoring of the status of cells. This technology can detect drug-induced liver toxicity by continuously monitoring the secretion rates and levels of albumin and glutathione S-transferase α (GST-α) of a 3D liver on-a-chip model treated with Doxorubicin. The PC-TIR biosensor is based on a one-step antibody functionalization with high specificity and a detection range of 21.7 ng mL-1 to 7.83 x 103 ng mL-1 for albumin and 2.20 ng mL-1 to 7.94 x 102 ng mL-1 for GST-α. This approach provides critical advantages for the early detection of drug toxicity and improved temporal resolution to capture transient drug effects. The proposed proof-of-concept study introduces a scalable and efficient plug-in solution for organ-on-a-chip technologies, advancing drug development and in vitro testing methods by enabling timely and accurate toxicity assessments.
Collapse
Affiliation(s)
- Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA 91367, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA 91367, USA
| | - Luis Trabucco
- The University of Texas at San Antonio, Department of Biomedical Engineering and Chemical Engineering, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Maisha Ahmed
- The University of Texas at San Antonio, Department of Biomedical Engineering and Chemical Engineering, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA 91367, USA
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA 91367, USA
| | - Jing Yong Ye
- The University of Texas at San Antonio, Department of Biomedical Engineering and Chemical Engineering, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA 91367, USA
| |
Collapse
|
18
|
Dey S, Bhat A, Janani G, Shandilya V, Gupta R, Mandal BB. Microfluidic human physiomimetic liver model as a screening platform for drug induced liver injury. Biomaterials 2024; 310:122627. [PMID: 38823194 DOI: 10.1016/j.biomaterials.2024.122627] [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: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The pre-clinical animal models often fail to predict intrinsic and idiosyncratic drug induced liver injury (DILI), thus contributing to drug failures in clinical trials, black box warnings and withdrawal of marketed drugs. This suggests a critical need for human-relevant in vitro models to predict diverse DILI phenotypes. In this study, a porcine liver extracellular matrix (ECM) based biomaterial ink with high printing fidelity, biocompatibility and tunable rheological and mechanical properties is formulated for supporting both parenchymal and non-parenchymal cells. Further, we applied 3D printing and microfluidic technology to bioengineer a human physiomimetic liver acinus model (HPLAM), recapitulating the radial hepatic cord-like structure with functional sinusoidal microvasculature network, biochemical and biophysical properties of native liver acinus. Intriguingly, the human derived hepatic cells incorporated HPLAM cultured under physiologically relevant microenvironment, acts as metabolic biofactories manifesting enhanced hepatic functionality, secretome levels and biomarkers expression over several weeks. We also report that the matured HPLAM reproduces dose- and time-dependent hepatotoxic response of human clinical relevance to drugs typically recognized for inducing diverse DILI phenotypes as compared to conventional static culture. Overall, the developed HPLAM emulates in vivo like functions and may provide a useful platform for DILI risk assessment to better determine safety and human risk.
Collapse
Affiliation(s)
- Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Amritha Bhat
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - G Janani
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Vartik Shandilya
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Raghvendra Gupta
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Biman B Mandal
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
19
|
Segovia-Zafra A, Villanueva-Paz M, Serras AS, Matilla-Cabello G, Bodoque-García A, Di Zeo-Sánchez DE, Niu H, Álvarez-Álvarez I, Sanz-Villanueva L, Godec S, Milisav I, Bagnaninchi P, Andrade RJ, Lucena MI, Fernández-Checa JC, Cubero FJ, Miranda JP, Nelson LJ. Control compounds for preclinical drug-induced liver injury assessment: Consensus-driven systematic review by the ProEuroDILI network. J Hepatol 2024; 81:630-640. [PMID: 38703829 DOI: 10.1016/j.jhep.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/10/2024] [Accepted: 04/21/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND & AIMS Idiosyncratic drug-induced liver injury (DILI) is a complex and unpredictable event caused by drugs, and herbal or dietary supplements. Early identification of human hepatotoxicity at preclinical stages remains a major challenge, in which the selection of validated in vitro systems and test drugs has a significant impact. In this systematic review, we analyzed the compounds used in hepatotoxicity assays and established a list of DILI-positive and -negative control drugs for validation of in vitro models of DILI, supported by literature and clinical evidence and endorsed by an expert committee from the COST Action ProEuroDILI Network (CA17112). METHODS Following 2020 PRISMA guidelines, original research articles focusing on DILI which used in vitro human models and performed at least one hepatotoxicity assay with positive and negative control compounds, were included. Bias of the studies was assessed by a modified 'Toxicological Data Reliability Assessment Tool'. RESULTS A total of 51 studies (out of 2,936) met the inclusion criteria, with 30 categorized as reliable without restrictions. Although there was a broad consensus on positive compounds, the selection of negative compounds lacked clarity. 2D monoculture, short exposure times and cytotoxicity endpoints were the most tested, although there was no consensus on drug concentrations. CONCLUSIONS Extensive analysis highlighted the lack of agreement on control compounds for in vitro DILI assessment. Following comprehensive in vitro and clinical data analysis together with input from the expert committee, an evidence-based consensus-driven list of 10 positive and negative control drugs for validation of in vitro models of DILI is proposed. IMPACT AND IMPLICATIONS Prediction of human toxicity early in the drug development process remains a major challenge, necessitating the development of more physiologically relevant liver models and careful selection of drug-induced liver injury (DILI)-positive and -negative control drugs to better predict the risk of DILI associated with new drug candidates. Thus, this systematic study has crucial implications for standardizing the validation of new in vitro models of DILI. By establishing a consensus-driven list of positive and negative control drugs, the study provides a scientifically justified framework for enhancing the consistency of preclinical testing, thereby addressing a significant challenge in early hepatotoxicity identification. Practically, these findings can guide researchers in evaluating safety profiles of new drugs, refining in vitro models, and informing regulatory agencies on potential improvements to regulatory guidelines, ensuring a more systematic and efficient approach to drug safety assessment.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Marina Villanueva-Paz
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Ana Sofia Serras
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Gonzalo Matilla-Cabello
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Ana Bodoque-García
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - Daniel E Di Zeo-Sánchez
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Hao Niu
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - Ismael Álvarez-Álvarez
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Laura Sanz-Villanueva
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy VIC, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - Sergej Godec
- Department of Anaesthesiology and Surgical Intensive Care, University Medical Centre Ljubljana, Ljubljana, Slovenia; Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of oxidative stress research, Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Pierre Bagnaninchi
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Raúl J Andrade
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Plataforma de Investigación Clínica y Ensayos Clínicos UICEC-IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain
| | - M Isabel Lucena
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Plataforma de Investigación Clínica y Ensayos Clínicos UICEC-IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain.
| | - José C Fernández-Checa
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Medicine, Keck School of Division of Gastrointestinal and Liver disease, University of Southern California, Los Angeles, CA, United States.
| | - Francisco Javier Cubero
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Joana Paiva Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Leonard J Nelson
- Institute for Bioengineering, School of Engineering, Faraday Building, The University of Edinburgh, Scotland, United Kingdom
| |
Collapse
|
20
|
Xiao K, Li H, Li Y, Zhan B, Fang X, Zhao B, Zhang X, Wu Y, Wang F, Jia Y. Protective effects and mechanism of Sangyu granule on acetaminophen-induced liver injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118282. [PMID: 38701935 DOI: 10.1016/j.jep.2024.118282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/23/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Sang Yu granule (SY), a traditional Chinese medicine prescription of Xijing Hospital, was developed based on the Guanyin powder in the classical prescription "Hong's Collection of Proven Prescriptions" and the new theory of modern Chinese medicine. It has been proved to have a certain therapeutic effect on drug-induced liver injury (DILI), but the specific mechanism of action is still unclear. AIM OF STUDY Aim of the study was to explore the effect of SangYu granule on treating drug-induced liver injury induced by acetaminophen in mice. MATERIALS AND METHODS The chemical composition of SY, serum, and liver tissue was analyzed using ultrahigh-performance liquid chromatography quadrupole time-of-flight mass spectrometry. To assess hepatic function, measurements were taken using kits for total bile acids, as well as serum AST, ALT, and ALP activity. Concentrations of IL-1β and TNF-α in serum were quantified using ELISA kits. Transcriptome Sequencing Analysis and 2bRAD-M microbial diversity analysis were employed to evaluate gene expression variance in liver tissue and fecal microbiota diversity among different groups, respectively. Western blotting was performed to observe differences in the activation levels of FXR, SHP, CYP7A1 and PPARα in the liver, and the levels of FXR and FGF-15 genes and proteins in the ileum of mice. Additionally, fecal microbiota transplantation (FMT) experiments were conducted to investigate the potential therapeutic effect of administering the intestinal microbial suspension from mice treated with SY on drug-induced liver injury. RESULTS SY treatment exhibited significant hepatoprotective effects in mice, effectively ameliorating drug-induced liver injury while concurrently restoring intestinal microbial dysbiosis. Furthermore, SY administration demonstrated a reduction in the concentration of total bile acids, the expression of FXR and SHP proteins in the liver was up-regulated, CYP7A1 protein was down-regulated, and the expressions of FXR and FGF-15 proteins in the ileum were up-regulated. However, no notable impact on PPARα was observed. Furthermore, results from FMT experiments indicated that the administration of fecal suspensions derived from mice treated with SY did not yield any therapeutic benefits in the context of drug-induced liver injury. CONCLUSION The aforementioned findings strongly suggest that SY exerts a pronounced ameliorative effect on drug-induced liver injury through its ability to modulate the expression of key proteins involved in bile acid secretion, thereby preserving hepato-enteric circulation homeostasis.
Collapse
Affiliation(s)
- Kexin Xiao
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China; College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi Province, China
| | - Hongyu Li
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China; College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi Province, China
| | - Yuening Li
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China; College of life sciences, Northwestern University, Xi'an, 710069, China
| | - Bo Zhan
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China; College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi Province, China
| | - Xiaohua Fang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi Province, China
| | - Bingjie Zhao
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China; College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi Province, China
| | - Xiaofei Zhang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi Province, China.
| | - Yumei Wu
- Department of Pharmacology, Air Force Medical University, Xi'an, 710032, China.
| | - Fan Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Yanyan Jia
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China; College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi Province, China.
| |
Collapse
|
21
|
Khan MZI, Ren JN, Cao C, Ye HYX, Wang H, Guo YM, Yang JR, Chen JZ. Comprehensive hepatotoxicity prediction: ensemble model integrating machine learning and deep learning. Front Pharmacol 2024; 15:1441587. [PMID: 39234116 PMCID: PMC11373136 DOI: 10.3389/fphar.2024.1441587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/24/2024] [Indexed: 09/06/2024] Open
Abstract
Background Chemicals may lead to acute liver injuries, posing a serious threat to human health. Achieving the precise safety profile of a compound is challenging due to the complex and expensive testing procedures. In silico approaches will aid in identifying the potential risk of drug candidates in the initial stage of drug development and thus mitigating the developmental cost. Methods In current studies, QSAR models were developed for hepatotoxicity predictions using the ensemble strategy to integrate machine learning (ML) and deep learning (DL) algorithms using various molecular features. A large dataset of 2588 chemicals and drugs was randomly divided into training (80%) and test (20%) sets, followed by the training of individual base models using diverse machine learning or deep learning based on three different kinds of descriptors and fingerprints. Feature selection approaches were employed to proceed with model optimizations based on the model performance. Hybrid ensemble approaches were further utilized to determine the method with the best performance. Results The voting ensemble classifier emerged as the optimal model, achieving an excellent prediction accuracy of 80.26%, AUC of 82.84%, and recall of over 93% followed by bagging and stacking ensemble classifiers method. The model was further verified by an external test set, internal 10-fold cross-validation, and rigorous benchmark training, exhibiting much better reliability than the published models. Conclusion The proposed ensemble model offers a dependable assessment with a good performance for the prediction regarding the risk of chemicals and drugs to induce liver damage.
Collapse
Affiliation(s)
| | - Jia-Nan Ren
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Cheng Cao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Polytechnic Institute, Zhejiang University, Hangzhou, China
| | - Hong-Yu-Xiang Ye
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hao Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ya-Min Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jin-Rong Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Polytechnic Institute, Zhejiang University, Hangzhou, China
| | - Jian-Zhong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
22
|
Wang J, Huang D, Chen H, Zhao Y. Biomimetic hepatic lobules from three-dimensional imprinted cell sheets. Sci Bull (Beijing) 2024; 69:1448-1457. [PMID: 38490890 DOI: 10.1016/j.scib.2024.02.030] [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: 01/07/2024] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 03/17/2024]
Abstract
Liver-tissue engineering has proven valuable in treating liver diseases, but the construction of liver tissues with high fidelity remains challenging. Here, we present a novel three-dimensional (3D)-imprinted cell-sheet strategy for the synchronous construction of biomimetic hepatic microtissues with high accuracy in terms of cell type, density, and distribution. To achieve this, the specific composition of hepatic cells in a normal human liver was determined using a spatial proteogenomics dataset. The data and biomimetic hepatic micro-tissues with hexagonal hollow cross-sections indicate that cell information was successfully generated using a homemade 3D-imprinted device for layer-by-layer imprinting and assembling the hepatic cell sheets. By infiltrating vascular endothelial cells into the hollow section of the assembly, biomimetic hepatic microtissues with vascularized channels for nutrient diffusion and drug perfusion can be obtained. We demonstrate that the resultant vascularized biomimetic hepatic micro-tissues can not only be integrated into a microfluidic drug-screening liver-on-a-chip but also assembled into an enlarged physiological structure to promote liver regeneration. We believe that our 3D-imprinted cell sheets strategy will open new avenues for biomimetic microtissue construction.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Danqing Huang
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Hanxu Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yuanjin Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
23
|
Ortega-Vallbona R, Méndez R, Tolosa L, Escher SE, Castell JV, Gozalbes R, Serrano-Candelas E. Uncovering the toxicity mechanisms of a series of carboxylic acids in liver cells through computational and experimental approaches. Toxicology 2024; 504:153764. [PMID: 38428665 DOI: 10.1016/j.tox.2024.153764] [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: 12/22/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Hepatotoxicity poses a significant concern in drug design due to the potential liver damage that can be caused by new drugs. Among common manifestations of hepatotoxic damage is lipid accumulation in hepatic tissue, resulting in liver steatosis or phospholipidosis. Carboxylic derivatives are prone to interfere with fatty acid metabolism and cause lipid accumulation in hepatocytes. This study investigates the toxic behaviour of 24 structurally related carboxylic acids in hepatocytes, specifically their ability to cause accumulation of fatty acids and phospholipids. Using high-content screening (HCS) assays, we identified two distinct lipid accumulation patterns. Subsequently, we developed structure-activity relationship (SAR) and quantitative structure-activity relationship (QSAR) models to determine relevant molecular substructures and descriptors contributing to these adverse effects. Additionally, we calculated physicochemical properties associated with lipid accumulation in hepatocytes and examined their correlation with our chemical structure characteristics. To assess the applicability of our findings to a wide range of chemical compounds, we employed two external datasets to evaluate the distribution of our QSAR descriptors. Our study highlights the significance of subtle molecular structural variations in triggering hepatotoxicity, such as the presence of nitrogen or the specific arrangement of substitutions within the carbon chain. By employing our comprehensive approach, we pinpointed specific molecules and elucidated their mechanisms of toxicity, thus offering valuable insights to guide future toxicology investigations.
Collapse
Affiliation(s)
- Rita Ortega-Vallbona
- ProtoQSAR SL., Centro Europeo de Empresas e Innovación (CEEI), Parque Tecnológico de Valencia, Av. Benjamín Franklin, 12, Valencia, Paterna 46980, Spain
| | - Rebeca Méndez
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, Valencia 46026, Spain
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, Valencia 46026, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), ISCIII, C/ Monforte de Lemos, Madrid 28029, Spain
| | - Sylvia E Escher
- Fraunhofer ITEM, Chemical Safety and Toxicology, Nikolai-Fuchs-Straße 1, Hannover 30625, Germany
| | - José V Castell
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, Valencia 46026, Spain; Departamento de Bioquímica y Biología Molecular. Facultad de Medicina, Universidad de Valencia, Av. de Blasco Ibáñez, 15, Valencia 46010, Spain; CIBEREHD, ISCIII, C/ Monforte de Lemos, Madrid 28029, Spain.
| | - Rafael Gozalbes
- ProtoQSAR SL., Centro Europeo de Empresas e Innovación (CEEI), Parque Tecnológico de Valencia, Av. Benjamín Franklin, 12, Valencia, Paterna 46980, Spain; Moldrug AI Systems SL, c/Olimpia Arozena Torres 45, Valencia 46018, Spain
| | - Eva Serrano-Candelas
- ProtoQSAR SL., Centro Europeo de Empresas e Innovación (CEEI), Parque Tecnológico de Valencia, Av. Benjamín Franklin, 12, Valencia, Paterna 46980, Spain
| |
Collapse
|
24
|
Zhao S, Feng Y, Zhang J, Zhang Q, Wang J, Cui S. Comparative analysis of gene expression between mice and humans in acetaminophen-induced liver injury by integrating bioinformatics analysis. BMC Med Genomics 2024; 17:80. [PMID: 38549107 PMCID: PMC10976682 DOI: 10.1186/s12920-024-01848-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
OBJECTIVE Mice are routinely utilized as animal models of drug-induced liver injury (DILI), however, there are significant differences in the pathogenesis between mice and humans. This study aimed to compare gene expression between humans and mice in acetaminophen (APAP)-induced liver injury (AILI), and investigate the similarities and differences in biological processes between the two species. METHODS A pair of public datasets (GSE218879 and GSE120652) obtained from GEO were analyzed using "Limma" package in R language, and differentially expressed genes (DEGs) were identified, including co-expressed DEGs (co-DEGs) and specific-expressed DEGS (specific-DEGs). Analysis of Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed analyses for specific-DEGs and co-DEGs. The co-DEGs were also used to construct transcription factor (TF)-gene network, gene-miRNA interactions network and protein-protein interaction (PPI) network for analyzing hub genes. RESULTS Mouse samples contained 1052 up-regulated genes and 1064 down-regulated genes, while human samples contained 1156 up-regulated genes and 1557 down-regulated genes. After taking the intersection between the DEGs, only 154 co-down-regulated and 89 co-up-regulated DEGs were identified, with a proportion of less than 10%. It was suggested that significant differences in gene expression between mice and humans in drug-induced liver injury. Mouse-specific-DEGs predominantly engaged in processes related to apoptosis and endoplasmic reticulum stress, while human-specific-DEGs were concentrated around catabolic process. Analysis of co-regulated genes reveals showed that they were mainly enriched in biosynthetic and metabolism-related processes. Then a PPI network which contains 189 nodes and 380 edges was constructed from the co-DEGs and two modules were obtained by Mcode. We screened out 10 hub genes by three algorithms of Degree, MCC and MNC, including CYP7A1, LSS, SREBF1, FASN, CD44, SPP1, ITGAV, ANXA5, LGALS3 and PDGFRA. Besides, TFs such as FOXC1, HINFP, NFKB1, miRNAs like mir-744-5p, mir-335-5p, mir-149-3p, mir-218-5p, mir-10a-5p may be the key regulatory factors of hub genes. CONCLUSIONS The DEGs of AILI mice models and those of patients were compared, and common biological processes were identified. The signaling pathways and hub genes in co-expression were identified between mice and humans through a series of bioinformatics analyses, which may be more valuable to reveal molecular mechanisms of AILI.
Collapse
Affiliation(s)
- Shanmin Zhao
- Department of Laboratory Animal Sciences, School of Basic Medicine, Naval Medical University, NO. 800 Xiangyin Road, 200433, Shanghai, China
| | - Yan Feng
- Department of Laboratory Animal Sciences, School of Basic Medicine, Naval Medical University, NO. 800 Xiangyin Road, 200433, Shanghai, China
| | - Jingyuan Zhang
- Department of Laboratory Animal Sciences, School of Basic Medicine, Naval Medical University, NO. 800 Xiangyin Road, 200433, Shanghai, China
| | - Qianqian Zhang
- Department of Laboratory Animal Sciences, School of Basic Medicine, Naval Medical University, NO. 800 Xiangyin Road, 200433, Shanghai, China
| | - Junyang Wang
- Department of Laboratory Animal Sciences, School of Basic Medicine, Naval Medical University, NO. 800 Xiangyin Road, 200433, Shanghai, China
| | - Shufang Cui
- Department of Laboratory Animal Sciences, School of Basic Medicine, Naval Medical University, NO. 800 Xiangyin Road, 200433, Shanghai, China.
| |
Collapse
|
25
|
Barazesh M, Jalili S, Akhzari M, Faraji F, Khorramdin E. Recent Progresses on Pathophysiology, Diagnosis, Therapeutic Modalities,
and Management of Non-alcoholic Fatty Liver Disorder. CURRENT DRUG THERAPY 2024; 19:20-48. [DOI: 10.2174/1574885518666230417111247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 01/03/2025]
Abstract
Abstract:
Non-alcoholic fatty liver disease (NAFLD) is currently the utmost common chronic liver
disorder that happens through all age groups and is identified to occur in 14%-30% of the general
population, demonstrating a critical and grossing clinical issue because of the growing incidence of
obesity and overweight. From the histological aspect, it looks like alcoholic liver damage, but it happens in patients who avoid remarkable alcohol usage. NAFLD comprises a broad spectrum, ranging
from benign hepatocellular steatosis to inflammatory nonalcoholic steatohepatitis (NASH), different
levels of fibrosis, and cirrhosis. Patients with NASH are more susceptible to more rapid progression to
cirrhosis and hepatocellular carcinoma. There is no single factor that drives proceeding from simple
steatosis to NASH. However, a combination of multi parameters such as genetic background, gut microflora, intake of high fat/ fructose dietary contents or methionine/choline-deficient diet, and consequently accumulated hepatocellular lipids mainly including triglycerides and also other bio-analytes,
such as free fatty acids, cholesterol, and phospholipids display a crucial role in disease promotion.
NAFLD is related to overweight and insulin resistance (IR) and is regarded as the hepatic presentation
of the metabolic syndrome, an amalgamation of medical statuses such as hyperlipidemia, hypertension, type 2 diabetes, and visceral obesity. Despite the increasing prevalence of this disease, which
imposes a remarkable clinical burden, most affected patients remain undiagnosed in a timely manner,
largely related to the asymptomatic entity of NAFLD patients and the unavailability of accurate and
efficient noninvasive diagnostic tests. However, liver biopsy is considered a gold standard for NAFLD
diagnosis, but due to being expensive and invasiveness is inappropriate for periodic disease screening.
Some noninvasive monitoring approaches have been established recently for NAFLD assessment. In
addition to the problem of correct disease course prediction, no effective therapeutic modalities are
approved for disease treatment. Imaging techniques can commonly validate the screening and discrimination of NAFLD; nevertheless, staging the disease needs a liver biopsy. The present therapeutic approaches depend on weight loss, sports activities, and dietary modifications, although different insulin-sensitizing drugs, antioxidants, and therapeutic agents seem hopeful. This review aims to focus on
the current knowledge concerning epidemiology, pathogenesis, and different biochemical experiments
and imaging modalities applied to diagnose the different grades of NAFLD and its management, as
well as new data about pharmacological therapies for this disorder.
Collapse
Affiliation(s)
- Mahdi Barazesh
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of
Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of
Medical Sciences, Larestan, Iran
| | - Fouzieyeh Faraji
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Ebrahim Khorramdin
- Department of Orthopedics, School of
Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
26
|
Lu J, Gong Y, Gao Y, Yang Y, Zhang Y, Zhang Z, Shi X. Wolfberry, Yam, and Chrysanthemum polysaccharides increased intestinal Akkermansia muciniphila abundance and hepatic YAP1 expression to alleviate DILI. FASEB J 2023; 37:e23286. [PMID: 37950623 DOI: 10.1096/fj.202301388r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/27/2023] [Accepted: 10/18/2023] [Indexed: 11/13/2023]
Abstract
Drug-induced liver injury (DILI) is frequently induced by high dose of acetaminophen (APAP) and is concomitant with disturbances of gut flora. Akkermansia muciniphila is beneficial for the repair of liver injury. Lycium barbarum polysaccharide, yam polysaccharide, and chrysanthemum polysaccharide all have anti-inflammatory and antioxidation effects. The objective of this study is to investigate the potential of lycium barbarum polysaccharide, yam polysaccharide, and chrysanthemum polysaccharide (LYC) in improving DILI by increasing the abundance of A. muciniphila. Initially, screening for the optimal concentrations of wolfberry, yam, and chrysanthemum (WYC) or LYC to promote A. muciniphila proliferation in vitro and validated in antibiotic (ATB)-treated KM mice. Subsequently, APAP-induced DILI model in BALB/c mice were constructed to examine the treatment effects of LYC. Our findings indicate that the optimal concentration ratio of WYC was 2:3:2, and LYC was 1:1:1. WYC increased A. muciniphila proliferation in vitro and in ATB-treated mice under this ratio. Meanwhile, LYC increased A. muciniphila abundance in vitro and the combination LYC with A. muciniphila promoted the proliferation of A. muciniphila in ATB-treated mice. The overdose of APAP resulted in the impairment of the intestinal barrier function and subsequent leakage of lipopolysaccharide (LPS). Moreover, LYC increased A. muciniphila abundance, reduced intestinal inflammation and permeability, and upregulated the expression of the tight junction protein zonula occludens protein 1 (ZO-1) and occludin contents in the gut. Lastly, LYC inhibited LPS leakage and upregulated hepatic YAP1 expression, ultimately leading to the repair of DILI.
Collapse
Affiliation(s)
- Junlan Lu
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yi Gong
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuting Gao
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yanguang Yang
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuman Zhang
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhiqin Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xinli Shi
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
27
|
Nayak S, Gowda J, Abbas SA, Kim H, Han SB. Recent Advances in the Development of Sulfamoyl-Based Hepatitis B Virus Nucleocapsid Assembly Modulators. Viruses 2023; 15:2367. [PMID: 38140607 PMCID: PMC10747759 DOI: 10.3390/v15122367] [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: 10/15/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Hepatitis B virus (HBV) is the primary contributor to severe liver ailments, encompassing conditions such as cirrhosis and hepatocellular carcinoma. Globally, 257 million people are affected by HBV annually and 887,000 deaths are attributed to it, representing a substantial health burden. Regrettably, none of the existing therapies for chronic hepatitis B (CHB) have achieved satisfactory clinical cure rates. This issue stems from the existence of covalently closed circular DNA (cccDNA), which is difficult to eliminate from the nucleus of infected hepatocytes. HBV genetic material is composed of partially double-stranded DNA that forms complexes with viral polymerase inside an icosahedral capsid composed of a dimeric core protein. The HBV core protein, consisting of 183 to 185 amino acids, plays integral roles in multiple essential functions within the HBV replication process. In this review, we describe the effects of sulfamoyl-based carboxamide capsid assembly modulators (CAMs) on capsid assembly, which can suppress HBV replication and disrupt the production of new cccDNA. We present research on classical, first-generation sulfamoyl benzocarboxamide CAMs, elucidating their structural composition and antiviral efficacy. Additionally, we explore newly identified sulfamoyl-based CAMs, including sulfamoyl bicyclic carboxamides, sulfamoyl aromatic heterocyclic carboxamides, sulfamoyl aliphatic heterocyclic carboxamides, cyclic sulfonamides, and non-carboxamide sulfomoyl-based CAMs. We believe that certain molecules derived from sulfamoyl groups have the potential to be developed into essential components of a well-suited combination therapy, ultimately yielding superior clinical efficacy outcomes in the future.
Collapse
Affiliation(s)
- Sandesha Nayak
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Jayaraj Gowda
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Syed Azeem Abbas
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Hyejin Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Soo Bong Han
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
28
|
Allison R, Guraka A, Shawa IT, Tripathi G, Moritz W, Kermanizadeh A. Drug induced liver injury - a 2023 update. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:442-467. [PMID: 37786264 DOI: 10.1080/10937404.2023.2261848] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Drug-Induced Liver Injury (DILI) constitutes hepatic damage attributed to drug exposure. DILI may be categorized as hepatocellular, cholestatic or mixed and might also involve immune responses. When DILI occurs in dose-dependent manner, it is referred to as intrinsic, while if the injury occurs spontaneously, it is termed as idiosyncratic. This review predominately focused on idiosyncratic liver injury. The established molecular mechanisms for DILI include (1) mitochondria dysfunction, (2) increased reactive oxygen species levels, (3) presence of elevated apoptosis and necrosis, (4) and bile duct injuries associated with immune mediated pathways. However, it should be emphasized that the underlying mechanisms responsible for DILI are still unknown. Prevention strategies are critical as incidences occur frequently, and treatment options are limited once the injury has developed. The aim of this review was to utilize retrospective cohort studies from across the globe to gain insight into epidemiological patterns. This review considers (1) what is currently known regarding the mechanisms underlying DILI, (2) discusses potential risk factors and (3) implications of the coronavirus pandemic on DILI presentation and research. Future perspectives are also considered and discussed and include potential new biomarkers, causality assessment and reporting methods.
Collapse
Affiliation(s)
- Rebecca Allison
- College of Science and Technology, University of Derby, Derby, UK
| | - Asha Guraka
- College of Science and Technology, University of Derby, Derby, UK
| | - Isaac Thom Shawa
- College of Science and Technology, University of Derby, Derby, UK
| | - Gyan Tripathi
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | | | - Ali Kermanizadeh
- College of Science and Technology, University of Derby, Derby, UK
| |
Collapse
|
29
|
Pan Y, Yang Q, Xu H, Yuan Z, Xu H. Screening and optimization of a water-soluble near-infrared fluorescent probe for drug-induced liver injury monitoring. Anal Chim Acta 2023; 1276:341654. [PMID: 37573102 DOI: 10.1016/j.aca.2023.341654] [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: 03/25/2023] [Revised: 06/25/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
Peroxynitrite (ONOO-) is a potential biomarker of drug-induced liver injury (DILI) and is involved in the process of DILI. Therefore, developing a reliable detection method for ONOO- will greatly contribute to ensuring drug safety and improving treatment efficiency. Here, based on the previous work, two kinds of NIR fluorescence probes PN and SPN were developed with phenyl-hydrazine as the ONOO- recognition group, which based on two fluorophores RN and SRN that are stable to ONOO-. A sensitive NIR probe SPN with good water solubility, low detection limit and good biocompatibility was selected through in vitro spectral property screening. Further experimental results show that there is a good linear relationship between the response intensity of probe SPN to ONOO- and the concentration of ONOO-, and the detection limit can reach 19.7 nM. At the cellular level, probe SPN can achieve a good and specific response to endogenous and exogenous ONOO-. Also, the probe SPN can be used for imaging and detection of DILI in zebrafish level and small animal level, indicating that probe SPN can be used as a powerful tool for diagnosis of DILI and efficacy evaluation of therapeutic drugs.
Collapse
Affiliation(s)
- Yanping Pan
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, China; Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing, 210009, China
| | - Qiuxing Yang
- Cancer Research Center Nantong, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Hong Xu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing, 210009, China
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing, 210009, China.
| | - Hui Xu
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, China.
| |
Collapse
|
30
|
Miller RT. Risk Assessment for Hepatobiliary Toxicity Liabilities in Drug Development. Toxicol Pathol 2023; 51:432-436. [PMID: 38243687 DOI: 10.1177/01926233231223751] [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] [Indexed: 01/21/2024]
Abstract
Risk assessment of hepatobiliary toxicities represents one of the greatest challenges and, more often than not, one of the most rewarding activities in which toxicologic pathologists can partake, and often times lead. This is in part because each liver toxicity picture is a bit different, informed by a broad range and diversity of relevant data, and also in part because the heavily relied upon animal models are imperfect regarding predictivity of hepatic effects in humans. Following identification and characterization of a hepatotoxicity hazard, typically in nonclinical toxicology studies, a holistic and integrated assessment of liver-relevant endpoints is conducted that typically incorporates ADME (absorption, distribution, metabolism, and excretion) information (ideally, including extensive transporter data, exposure margins, and possibly concentration of parent/metabolite at region of injury), target expression/function, in silico prediction data, in vitro hepatocyte data, liver/circulating biomarkers, and importantly, species specificity of any of these data. Of course, a thorough understanding, developed in close partnership with clinical colleagues, of the anticipated liver disease status of intended patient populations is paramount to hepatic risk assessment. This is particularly important since the likelihood of translatable determinant hepatic events observed in nonclinical models to occur in humans has been reasonably well established.
Collapse
Affiliation(s)
- Richard T Miller
- Apex Drug Discovery and Innovation Strategies, Apex, North Carolina, USA
| |
Collapse
|
31
|
Li M, Xu H, Zhao N, Zhang L, Xia H, Zhang X, Li Q, Liao M, Pan Q, Yi Z, Chai J. Hepatocyte-targeted hyaluronic acid-polyethyleneimine conjugates for acute liver injury therapy by ROS elimination and inflammation modulation. MATERIALS & DESIGN 2023; 233:112212. [DOI: 10.1016/j.matdes.2023.112212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
|
32
|
Du H, Tong S, Kuang G, Gong X, Jiang N, Yang X, Liu H, Li N, Xie Y, Xiang Y, Guo J, Li Z, Yuan Y, Wu S, Wan J. Sesamin Protects against APAP-Induced Acute Liver Injury by Inhibiting Oxidative Stress and Inflammatory Response via Deactivation of HMGB1/TLR4/NF κB Signal in Mice. J Immunol Res 2023; 2023:1116841. [PMID: 37663051 PMCID: PMC10471453 DOI: 10.1155/2023/1116841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/16/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Acetaminophen (APAP) overdose would lead to liver toxicity and even acute liver failure in severe cases by triggering an inflammatory response and oxidative stress. Sesamin has been reported to possess anti-inflammatory and antioxidant actions in several animal disease models. In the present study, the effects and mechanisms of sesamin on APAP-induced acute liver injury (ALI) were explored. The results showed that pretreatment with sesamin significantly alleviated APAP-induced ALI, as indicated by decreased serum aminotransferase activities, hepatic pathological damages, and hepatic cellular apoptosis. But sesamin has no significant effects on the expression of cytochrome P450 2E1 (CYP2E1), APAP-cysteine adducts (APAP-CYS) production, and glutathione content in the liver of APAP-administered mice. Moreover, APAP-induced liver oxidative stress and inflammatory response also were remarkedly attenuated by sesamin, including reducing hepatic reactive oxygen species levels, promoting antioxidant generation, and inhibiting the expression of TNF-α and IL-1β, as well as decreasing inflammatory cell recruitment. Notably, sesamin inhibited serum high-mobility group box 1 (HMGB1) releases and blocked hepatic activation of Toll-like receptor 4 (TLR4)-interleukin 1 receptor-associated kinase 3-nuclear factor kappa B (NF-κB) signaling pathway in APAP-administered mice. These findings indicated that sesamin could mitigate APAP-induced ALI through suppression of oxidative stress and inflammatory response, which might be mediated by the deactivation of HMGB1/TLR4/NF-κB signaling in mice.
Collapse
Affiliation(s)
- Hui Du
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Shiwen Tong
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ge Kuang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Ningman Jiang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xian Yang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Hao Liu
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Nana Li
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yao Xie
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yang Xiang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jiashi Guo
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Zhenhan Li
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Yinglin Yuan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengwang Wu
- Department of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Russo D, Aleksunes LM, Goyak K, Qian H, Zhu H. Integrating Concentration-Dependent Toxicity Data and Toxicokinetics To Inform Hepatotoxicity Response Pathways. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:12291-12301. [PMID: 37566783 PMCID: PMC10448720 DOI: 10.1021/acs.est.3c02792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Failure of animal models to predict hepatotoxicity in humans has created a push to develop biological pathway-based alternatives, such as those that use in vitro assays. Public screening programs (e.g., ToxCast/Tox21 programs) have tested thousands of chemicals using in vitro high-throughput screening (HTS) assays. Developing pathway-based models for simple biological pathways, such as endocrine disruption, has proven successful, but development remains a challenge for complex toxicities like hepatotoxicity, due to the many biological events involved. To this goal, we aimed to develop a computational strategy for developing pathway-based models for complex toxicities. Using a database of 2171 chemicals with human hepatotoxicity classifications, we identified 157 out of 1600+ ToxCast/Tox21 HTS assays to be associated with human hepatotoxicity. Then, a computational framework was used to group these assays by biological target or mechanisms into 52 key event (KE) models of hepatotoxicity. KE model output is a KE score summarizing chemical potency against a hepatotoxicity-relevant biological target or mechanism. Grouping hepatotoxic chemicals based on the chemical structure revealed chemical classes with high KE scores plausibly informing their hepatotoxicity mechanisms. Using KE scores and supervised learning to predict in vivo hepatotoxicity, including toxicokinetic information, improved the predictive performance. This new approach can be a universal computational toxicology strategy for various chemical toxicity evaluations.
Collapse
Affiliation(s)
- Daniel
P. Russo
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| | - Lauren M. Aleksunes
- Department
of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Katy Goyak
- ExxonMobil
Biomedical Sciences, Inc., Annandale, New Jersey 08801, United States
| | - Hua Qian
- ExxonMobil
Biomedical Sciences, Inc., Annandale, New Jersey 08801, United States
| | - Hao Zhu
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
34
|
Moein M, Heinonen M, Mesens N, Chamanza R, Amuzie C, Will Y, Ceulemans H, Kaski S, Herman D. Chemistry-Based Modeling on Phenotype-Based Drug-Induced Liver Injury Annotation: From Public to Proprietary Data. Chem Res Toxicol 2023; 36:1238-1247. [PMID: 37556769 PMCID: PMC10445287 DOI: 10.1021/acs.chemrestox.2c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Indexed: 08/11/2023]
Abstract
Drug-induced liver injury (DILI) is an important safety concern and a major reason to remove a drug from the market. Advancements in recent machine learning methods have led to a wide range of in silico models for DILI predictive methods based on molecule chemical structures (fingerprints). Existing publicly available DILI data sets used for model building are based on the interpretation of drug labels or patient case reports, resulting in a typical binary clinical DILI annotation. We developed a novel phenotype-based annotation to process hepatotoxicity information extracted from repeated dose in vivo preclinical toxicology studies using INHAND annotation to provide a more informative and reliable data set for machine learning algorithms. This work resulted in a data set of 430 unique compounds covering diverse liver pathology findings which were utilized to develop multiple DILI prediction models trained on the publicly available data (TG-GATEs) using the compound's fingerprint. We demonstrate that the TG-GATEs compounds DILI labels can be predicted well and how the differences between TG-GATEs and the external test compounds (Johnson & Johnson) impact the model generalization performance.
Collapse
Affiliation(s)
- Mohammad Moein
- Department
of Computer Science, Aalto University, Konemiehentie 2, 02150 Espoo, Finland
| | - Markus Heinonen
- Department
of Computer Science, Aalto University, Konemiehentie 2, 02150 Espoo, Finland
| | - Natalie Mesens
- Predictive,
Investigative and Translational Toxicology, PSTS, Janssen Research
& Development, Pharmaceutical Companies
of Johnson & Johnson, 2340 Beerse, Belgium
| | - Ronnie Chamanza
- Pathology,
PSTS, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, 2340 Beerse, Belgium
| | - Chidozie Amuzie
- Johnson
& Johnson Innovation-JLABS, 661 University Avenue, CA014 ON Toronto, Canada
| | - Yvonne Will
- Predictive,
Investigative and Translational Toxicology, PSTS, Janssen Research
& Development, Pharmaceutical Companies
of Johnson & Johnson, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Hugo Ceulemans
- In-Silico
Discovery, Janssen Pharmaceutica, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, 2340 Beerse, Belgium
| | - Samuel Kaski
- Department
of Computer Science, Aalto University, Konemiehentie 2, 02150 Espoo, Finland
| | - Dorota Herman
- In-Silico
Discovery, Janssen Pharmaceutica, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, 2340 Beerse, Belgium
| |
Collapse
|
35
|
Uzhytchak M, Lunova M, Smolková B, Jirsa M, Dejneka A, Lunov O. Iron oxide nanoparticles trigger endoplasmic reticulum damage in steatotic hepatic cells. NANOSCALE ADVANCES 2023; 5:4250-4268. [PMID: 37560414 PMCID: PMC10408607 DOI: 10.1039/d3na00071k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/13/2023] [Indexed: 08/11/2023]
Abstract
Iron oxide nanoparticles (IONPs) are being actively researched in various biomedical applications, particularly as magnetic resonance imaging (MRI) contrast agents for diagnosing various liver pathologies like nonalcoholic fatty liver diseases, nonalcoholic steatohepatitis, and cirrhosis. Emerging evidence suggests that IONPs may exacerbate hepatic steatosis and liver injury in susceptible livers such as those with nonalcoholic fatty liver disease. However, our understanding of how IONPs may affect steatotic cells at the sub-cellular level is still fragmented. Generally, there is a lack of studies identifying the molecular mechanisms of potential toxic and/or adverse effects of IONPs on "non-heathy" in vitro models. In this study, we demonstrate that IONPs, at a dose that does not cause general toxicity in hepatic cells (Alexander and HepG2), induce significant toxicity in steatotic cells (cells loaded with non-toxic doses of palmitic acid). Mechanistically, co-treatment with PA and IONPs resulted in endoplasmic reticulum (ER) stress, accompanied by the release of cathepsin B from lysosomes to the cytosol. The release of cathepsin B, along with ER stress, led to the activation of apoptotic cell death. Our results suggest that it is necessary to consider the interaction between IONPs and the liver, especially in susceptible livers. This study provides important basic knowledge for the future optimization of IONPs as MRI contrast agents for various biomedical applications.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences Prague 18221 Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences Prague 18221 Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM) Prague 14021 Czech Republic
| | - Barbora Smolková
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences Prague 18221 Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM) Prague 14021 Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences Prague 18221 Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences Prague 18221 Czech Republic
| |
Collapse
|
36
|
Gould S, Templin MV. Off target toxicities and links with physicochemical properties of medicinal products, including antibiotics, oligonucleotides, lipid nanoparticles (with cationic and/or anionic charges). Data review suggests an emerging pattern. Toxicol Lett 2023; 384:14-29. [PMID: 37454775 DOI: 10.1016/j.toxlet.2023.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Toxicology is an essential part of any drug development plan. Circumnavigating the risk of failure because of a toxicity issue can be a challenge, and failure in late development is extremely costly. To identify potential risks, it requires more than just understanding the biological target. The toxicologist needs to consider a compound's structure, it's physicochemical properties (including the impact of the overall formulation), as well as the biological target (e.g., receptor interactions). Understanding the impact of the physicochemical properties can be used to predict potential toxicities in advance by incorporating key endpoints in early screening strategies and/or used to compare toxicity profiles across lead candidates. This review discussed the risks of off-target and/or non-specific toxicities that may be associated with the physicochemical properties of compounds, especially those carrying dominant positive or negative charges, including amphiphilic small molecules, peptides, oligonucleotides and lipids/liposomes/lipid nanoparticles. The latter of which are being seen more and more in drug development, including the recent Covid pandemic, where mRNA and lipid nanoparticle technology is playing more of a role in vaccine development. The translation between non-clinical and clinical data is also considered, questioning how a physicochemical driven toxicity may be more universal across species, which means that such toxicity may be reassuringly translatable between species and as such, this information may also be considered as a support to the 3 R's, particularly in the early screening stages of a drug development plan.
Collapse
|
37
|
Xie S, Wei S, Ma X, Wang R, He T, Zhang Z, Yang J, Wang J, Chang L, Jing M, Li H, Zhou X, Zhao Y. Genetic alterations and molecular mechanisms underlying hereditary intrahepatic cholestasis. Front Pharmacol 2023; 14:1173542. [PMID: 37324459 PMCID: PMC10264785 DOI: 10.3389/fphar.2023.1173542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Hereditary cholestatic liver disease caused by a class of autosomal gene mutations results in jaundice, which involves the abnormality of the synthesis, secretion, and other disorders of bile acids metabolism. Due to the existence of a variety of gene mutations, the clinical manifestations of children are also diverse. There is no unified standard for diagnosis and single detection method, which seriously hinders the development of clinical treatment. Therefore, the mutated genes of hereditary intrahepatic cholestasis were systematically described in this review.
Collapse
Affiliation(s)
- Shuying Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shizhang Wei
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Xiao Ma
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruilin Wang
- Department of Pharmacy, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tingting He
- Department of Pharmacy, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhao Zhang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ju Yang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiawei Wang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei Chang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Manyi Jing
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Haotian Li
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Xuelin Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yanling Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
38
|
Niu L, Cao Q, Zhang T, Zhang Y, Liang T, Wang J. Simultaneous detection of mitochondrial viscosity and peroxynitrite in livers from subjects with drug-induced fatty liver disease using a novel fluorescent probe. Talanta 2023; 260:124591. [PMID: 37141820 DOI: 10.1016/j.talanta.2023.124591] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/19/2023] [Accepted: 04/23/2023] [Indexed: 05/06/2023]
Abstract
Drug-induced fatty liver disease (DIFLD) is a basic clinicopathological example of drug-induced liver injury (DILI). Some drugs can inhibit β-oxidation in hepatocyte mitochondria, leading to steatosis in the liver. Additionally, drug-induced inhibition of β-oxidation and the electron transport chain (ETC) can lead to increased production of reactive oxygen species (ROS) such as peroxynitrite (ONOO-). Therefore, it is reasonable to suspect that compared to a healthy liver, viscosity and ONOO- levels are elevated in livers during DIFLD. A novel, smart, dual-response fluorescent probe-Mito-VO-was designed and synthesized for the simultaneous detection of viscosity and ONOO- content. This probe had a large emission shift of 293 nm and was capable of monitoring the viscosity of, and the ONOO- content in, cell and animal models alike, either individually or simultaneously. For the first time, Mito-VO was successfully used to demonstrate the elevated viscosity and the amount of ONOO- in livers from mice with DIFLD.
Collapse
Affiliation(s)
- Linqiang Niu
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, PR China
| | - Qijuan Cao
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, PR China
| | - Tian Zhang
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, PR China
| | - Yahong Zhang
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, PR China
| | - Tingting Liang
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, PR China.
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, PR China.
| |
Collapse
|
39
|
Bhat S, Ahanger IA, Kazim SN. Forthcoming Developments in Models to Study the Hepatitis B Virus Replication Cycle, Pathogenesis, and Pharmacological Advancements. ACS OMEGA 2023; 8:14273-14289. [PMID: 37125123 PMCID: PMC10134252 DOI: 10.1021/acsomega.2c07154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/08/2023] [Indexed: 05/03/2023]
Abstract
Hepatitis, liver cirrhosis, and hepatocellular carcinoma are all manifestations of chronic hepatitis B. Its pathogenesis and molecular mechanism remain mysterious. As medical science progresses, different models are being used to study the disease from the physiological and molecular levels. Animal models have played an unprecedented role in achieving in-depth knowledge of the disease while posing no risk of harming humans throughout the study. The scarcity of acceptable animal models has slowed progress in hepatitis B virus (HBV) research and preclinical testing of antiviral medicines since HBV has a narrow species tropism and exclusively infects humans and higher primates. The development of human chimeric mice was supported by a better understanding of the obstacles to interspecies transmission, which has substantially opened the way for HBV research in vivo and the evaluation of possible chronic hepatitis B therapeutics. Animal models are cumbersome to handle, not accessible, and expensive. Hence, it is herculean to investigate the HBV replication cycle in animal models. Therefore, it becomes essential to build a splendid in vitro cell culture system to demonstrate the mechanisms attained by the HBV for its multiplication and sustenance. We also addressed the advantages and caveats associated with different models in examining HBV.
Collapse
Affiliation(s)
- Sajad
Ahmad Bhat
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Ishfaq Ahmad Ahanger
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Clinical
Biochemistry University of Kashmir, Srinagar, India
| | - Syed Naqui Kazim
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Phone: +91 9953621758.
| |
Collapse
|
40
|
Kubickova B, Jacobs MN. Development of a reference and proficiency chemical list for human steatosis endpoints in vitro. Front Endocrinol (Lausanne) 2023; 14:1126880. [PMID: 37168981 PMCID: PMC10166001 DOI: 10.3389/fendo.2023.1126880] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/17/2023] [Indexed: 05/13/2023] Open
Abstract
The most prevalent liver disease in humans is non-alcoholic fatty liver disease, characterised by excessive hepatic fat accumulation, or steatosis. The western diet and a sedentary lifestyle are considered to be major influences, but chemical exposure may also play a role. Suspected environmental chemicals of concern include pesticides, plasticizers, metals, and perfluorinated compounds. Here we present a detailed literature analysis of chemicals that may (or may not) be implicated in lipid accumulation in the liver, to provide a basis for developing and optimizing human steatosis-relevant in vitro test methods. Independently collated and reviewed reference and proficiency chemicals are needed to assist in the test method development where an assay is intended to ultimately be taken forward for OECD Test Guideline development purposes. The selection criteria and considerations required for acceptance of proficiency chemical selection for OECD Test Guideline development. (i.e., structural diversity, range of activity including negatives, relevant chemical sectors, global restrictions, etc.) is described herein. Of 160 chemicals initially screened for inclusion, 36 were prioritized for detailed review. Based on the selection criteria and a weight-of-evidence basis, 18 chemicals (9 steatosis inducers, 9 negatives), including some environmental chemicals of concern, were ranked as high priority chemicals to assist in vitro human steatosis test method optimisation and proficiency testing, and inform potential subsequent test method (pre-)validation.
Collapse
Affiliation(s)
| | - Miriam N. Jacobs
- Radiation, Chemical and Environmental Hazards (RCE), Department of Toxicology, UK Health Security Agency (UKHSA), Harwell Science and Innovation Campus, Chilton, United Kingdom
| |
Collapse
|
41
|
Fu J, Qiu H, Tan CS. Microfluidic Liver-on-a-Chip for Preclinical Drug Discovery. Pharmaceutics 2023; 15:pharmaceutics15041300. [PMID: 37111785 PMCID: PMC10141038 DOI: 10.3390/pharmaceutics15041300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Drug discovery is an expensive, long, and complex process, usually with a high degree of uncertainty. In order to improve the efficiency of drug development, effective methods are demanded to screen lead molecules and eliminate toxic compounds in the preclinical pipeline. Drug metabolism is crucial in determining the efficacy and potential side effects, mainly in the liver. Recently, the liver-on-a-chip (LoC) platform based on microfluidic technology has attracted widespread attention. LoC systems can be applied to predict drug metabolism and hepatotoxicity or to investigate PK/PD (pharmacokinetics/pharmacodynamics) performance when combined with other artificial organ-on-chips. This review discusses the liver physiological microenvironment simulated by LoC, especially the cell compositions and roles. We summarize the current methods of constructing LoC and the pharmacological and toxicological application of LoC in preclinical research. In conclusion, we also discussed the limitations of LoC in drug discovery and proposed a direction for improvement, which may provide an agenda for further research.
Collapse
Affiliation(s)
- Jingyu Fu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Hailong Qiu
- Tianjin Key Laboratory of Functional Crystal Materials, Institute of Functional Crystal, Tianjin University of Technology, Tianjin 300384, China
| | - Cherie S Tan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
42
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
43
|
He X, Liang SM, Wang HQ, Tao L, Sun FF, Wang Y, Zhang C, Huang YC, Xu DX, Chen X. Mitoquinone protects against acetaminophen-induced liver injury in an FSP1-dependent and GPX4-independent manner. Toxicol Appl Pharmacol 2023; 465:116452. [PMID: 36894071 DOI: 10.1016/j.taap.2023.116452] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
Mitochondrial oxidative stress has been a crucial mediator in acetaminophen (APAP)-induced hepatotoxicity. MitoQ, an analog of coenzyme Q10, is targeted towards mitochondria and acts as a potent antioxidant. This study aimed to explore the effect of MitoQ on APAP-induced liver injury and its possible mechanisms. To investigate this, CD-1 mice and AML-12 cells were treated with APAP. Hepatic MDA and 4-HNE, two markers of lipid peroxidation (LPO), were elevated as early as 2 h after APAP. Oxidized lipids were rapidly upregulated in APAP-exposed AML-12 cells. Hepatocyte death and mitochondrial ultrastructure alterations were observed in APAP-induced acute liver injury. The in vitro experiments showed that mitochondrial membrane potentials and OXPHOS subunits were downregulated in APAP-exposed hepatocytes. MtROS and oxidized lipids were elevated in APAP-exposed hepatocytes. We discovered that APAP-induced hepatocyte death and liver injury were ameliorated by attenuation of protein nitration and LPO in MitoQ-pretreated mice. Mechanistically, knockdown of GPX4, a key enzyme for LPO defense systems, exacerbated APAP-induced oxidized lipids, but did not influence the protective effect of MitoQ on APAP-induced LPO and hepatocyte death. Whereas knockdown of FSP1, another key enzyme for LPO defense systems, had little effect on APAP-induced lipid oxidation but partially weakened the protection of MitoQ on APAP-induced LPO and hepatocyte death. These results suggest that MitoQ may alleviate APAP-evoked hepatotoxicity by eliminating protein nitration and suppressing hepatic LPO. MitoQ prevents APAP-induced liver injury partially dependent of FSP1 and independent of GPX4.
Collapse
Affiliation(s)
- Xue He
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Shi-Min Liang
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Hong-Qian Wang
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Li Tao
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Fei-Fei Sun
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Yan Wang
- Department of Toxicology, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Cheng Zhang
- Department of Toxicology, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Yi-Chao Huang
- Department of Toxicology, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - De-Xiang Xu
- Department of Toxicology, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China.
| | - Xi Chen
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
44
|
Izawa T, Travlos GS, Cortes RA, Clayton NP, Sills RC, Pandiri AR. Absence of Increased Susceptibility to Acetaminophen-Induced Liver Injury in a Diet-Induced NAFLD Mouse Model. Toxicol Pathol 2023; 51:112-125. [PMID: 37158481 PMCID: PMC10523943 DOI: 10.1177/01926233231171101] [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] [Indexed: 05/10/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease and its influence on drug-induced liver injury (DILI) is not fully understood. We investigated whether NAFLD can influence acetaminophen (APAP [N-acetyl-p-aminophenol])-induced hepatotoxicity in a diet-induced obese (DIO) mouse model of NAFLD. The male C57BL/6NTac DIO mice, fed a high-fat diet for more than 12 weeks, developed obesity, hyperinsulinemia, impaired glucose tolerance, and hepatomegaly with hepatic steatosis, similar to human NAFLD. In the acute toxicity study after a single dose of APAP (150 mg/kg), compared with control lean mice, the DIO mice had decreased serum transaminase levels and less severe hepatocellular injury. The DIO mice also had altered expression of genes related to APAP metabolism. Chronic APAP exposure for 26 weeks did not predispose the DIO mice with NAFLD to more severe hepatotoxicity compared with the lean mice. These results suggested that the C57BL/6NTac DIO mouse model appears to be more tolerant to APAP-induced hepatotoxicity than lean mice, potentially related to altered xenobiotic metabolizing capacity in the fatty liver. Further mechanistic studies with APAP and other drugs in NAFLD animal models are necessary to investigate the mechanism of altered susceptibility to intrinsic DILI in some human NAFLD patients.
Collapse
Affiliation(s)
- Takeshi Izawa
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
- Laboratory of Veterinary Pathology, Osaka Metropolitan University, Izumisano, Osaka, Japan
| | - Gregory S. Travlos
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| | - Ricardo A. Cortes
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
- Experimental Pathology Laboratories, Inc., Research Triangle Park, NC, USA
| | - Natasha P. Clayton
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| | - Robert C. Sills
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| | - Arun R. Pandiri
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| |
Collapse
|
45
|
Nabil-Adam A, Ashour ML, Shreadah MA. The hepatoprotective candidates by synergistic formula of marine and terrestrial against Acetaminophen toxicity using in-vitro, in-vivo, and in silico screening approach. Saudi J Biol Sci 2023; 30:103607. [PMID: 36941882 PMCID: PMC10023929 DOI: 10.1016/j.sjbs.2023.103607] [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: 12/02/2022] [Revised: 01/27/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023] Open
Abstract
Background One of the most regularly used hepatotoxic medicines is paracetamol (acetaminophen, N-acetyl-p-aminophenol; APAP). It causes liver failure in overdoses but is safe at therapeutic dosages. Combination therapy combining many natural compounds with a synergistic impact as hepatoprotective agents has become an essential therapeutic method against various disorders. Objective Due to the lack of literature on paracetamol's effects on hematological and hepatic status parameters in male albino mice, the main goal of this study was to compare the hepatoprotective activities of a mixture of three marine-derived polyphenolics and polysaccharides (Sargassum vulgare Bacillus oceanisediminis, and alginic acids) to Chrysanthemum extract and the mixture of them. Methods Sargassumvulgare, Bacillus Oceanisediminis, and alginate, as well as Chrysanthemum ethanol extracts, were tested for APAP-induced liver damage. Group 1 received saline solution subcutaneously, while Group 2 received 500 mg/kg body weight/day APAP intraperitoneal. Group 3 got 200 mg/day algal extract i.p. As in group 3, group 4 got an i.p. dose of 200 mg of algal extract before the APAP dose. This group was protected by Sargassum vulgare extract. Group 5: Received 200 mg/100 g/body of Bacillus oceanisediminis extracts i.p. for one week. Group 6: Received 200 mg/body of Bacillus oceanisediminis extract i.p. for one week before APAP treatment. Alginate (p200 mg/body weight/day) was given to Group 7. As in group 7, group 8 received 200 mg/body weight/day alginate extract i.p. before APAP. Group 9: Chrysanthemum extracts 200 mg/day for a week. Group 10: got an i.p. dose of Chrysanthemum extracts for one week before the APAP dose. Group 11: Four mixed extracts (Bacillus Oceanisediminis, Sargassum vulgare, Chrysanthemum, and alginate) were i.p200 mg/day for one week as a positive (+ve) control group. Group 12: Received i.p200 mg/kg combination extract for one week before APAP. Results Due to their synergistic antioxidant and anti-inflammatory actions, marine extracts and combinations of marine-derived extracts demonstrated a great effect against APAP toxicity, demonstrating hepatoprotective potential against APAP-induced liver damage. Conclusion The synergy of the three marine-derived combinations may lead to novel liver toxicity prevention agents.
Collapse
Affiliation(s)
- Asmaa Nabil-Adam
- Marine Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Egypt
| | - Mohamed L. Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abbasia, Cairo 11566, Egypt
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeaddah 21442, Saudi Arabia
| | - Mohamed A. Shreadah
- Marine Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Egypt
| |
Collapse
|
46
|
Li S, Zhuge A, Xia J, Wang S, Lv L, Wang K, Jiang H, Yan R, Yang L, Bian X, Jiang X, Wang Q, Han S, Li L. Bifidobacterium longum R0175 protects mice against APAP-induced liver injury by modulating the Nrf2 pathway. Free Radic Biol Med 2023; 203:11-23. [PMID: 37003500 DOI: 10.1016/j.freeradbiomed.2023.03.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 02/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Acetaminophen (APAP) overdose is the most common driver of drug-induced liver injury (DILI) worldwide, and the gut microbiome plays a crucial role in this process. In this study, we estimated the effect of Bifidobacterium longum R0175 on APAP-induced liver injury in mice and discovered that B. longum R0175 alleviated liver injury by diminishing inflammation, reducing oxidative stress levels, inhibiting hepatocyte death and improving APAP-induced microbiome dysbiosis. Further studies revealed that the antioxidative effects of B. longum R0175 were primarily due to activation of the Nrf2 pathway, which was supported by the Nrf2 pathway inhibitor ML385 counteracting these ameliorative effects. B. longum R0175 modified intestinal metabolites, especially the key metabolite sedanolide, which could activate the Nrf2 pathway and contribute to the protective effects against APAP-induced liver injury. Moreover, we found that sedanolide exhibited close interrelationships with specific microbial taxa, indicating that this factor may be derived from gut microbes. In conclusion, our work demonstrated that B. longum R0175 could reduce oxidative damage, inflammation and hepatocyte death by activating the Nrf2 pathway. Importantly, we identified the microbiota-derived metabolite sedanolide, which was first discovered in the mouse intestine, as a key agonist of the Nrf2 pathway and primary effector of B. longum R0175 in APAP challenge. These findings provide new perspectives for APAP overdose therapy and demonstrate the enormous potential of B. longum R0175 in alleviating acute liver injury.
Collapse
Affiliation(s)
- Shengjie Li
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Aoxiang Zhuge
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Shuting Wang
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Kaicen Wang
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Huiyong Jiang
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Ren Yan
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Liya Yang
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xiaoyuan Bian
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xianwan Jiang
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innocation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China.
| |
Collapse
|
47
|
Park JE, Ahn CH, Lee HJ, Sim DY, Park SY, Kim B, Shim BS, Lee DY, Kim SH. Antioxidant-Based Preventive Effect of Phytochemicals on Anticancer Drug-Induced Hepatotoxicity. Antioxid Redox Signal 2023; 38:1101-1121. [PMID: 36242510 DOI: 10.1089/ars.2022.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Drug-induced liver injury (DILI) or hepatotoxicity has been a hot issue to overcome on the safety and physiological function of the liver, since it is known to have biochemical, cellular, immunological, and molecular alterations in the liver mainly induced by alcohol, chemicals, drugs, heavy metals, and genetic factors. Recently efficient therapeutic and preventive strategies by some phytochemicals are of interest, targeting oxidative stress-mediated hepatotoxicity alone or in combination with anticancer drugs. Recent Advances: To assess DILI, the variety of in vitro and in vivo animal models has been developed mainly by using carbon tetrachloride, d-galactosamine, acetaminophen, and lipopolysaccharide. Also, the mechanisms on hepatotoxicity by several drugs and herbs have been explored in detail. Recent studies reveal that antioxidants including vitamins and some phytochemicals were reported to prevent against DILI. Critical Issues: Antioxidant therapy with some phytochemicals is noteworthy, since oxidative stress is critically involved in DILI via production of chemically reactive oxygen species or metabolites, impairment of mitochondrial respiratory chain, and induction of redox cycling. Future Directions: For efficient antioxidant therapy, DILI susceptibility, Human Leukocyte Antigen genetic factors, biomarkers, and pathogenesis implicated in hepatotoxicity should be further explored in association with oxidative stress-mediated signaling, while more randomized preclinical and clinical trials are required with optimal safe doses of drugs and/or phytochemicals alone or in combination for efficient clinical practice along with the development of advanced DILI diagnostic tools.
Collapse
Affiliation(s)
- Ji Eon Park
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chi-Hoon Ahn
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyo-Jung Lee
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Deok Yong Sim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su Yeon Park
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bum Sang Shim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration (RDA), Eumseong, Republic of Korea
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Abstract
The epidemic of obesity, type 2 diabetes and nonalcoholic liver disease (NAFLD) favors drug consumption, which augments the risk of adverse events including liver injury. For more than 30 years, a series of experimental and clinical investigations reported or suggested that the common pain reliever acetaminophen (APAP) could be more hepatotoxic in obesity and related metabolic diseases, at least after an overdose. Nonetheless, several investigations did not reproduce these data. This discrepancy might come from the extent of obesity and steatosis, accumulation of specific lipid species, mitochondrial dysfunction and diabetes-related parameters such as ketonemia and hyperglycemia. Among these factors, some of them seem pivotal for the induction of cytochrome P450 2E1 (CYP2E1), which favors the conversion of APAP to the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). In contrast, other factors might explain why obesity and NAFLD are not always associated with more frequent or more severe APAP-induced acute hepatotoxicity, such as increased volume of distribution in the body, higher hepatic glucuronidation and reduced CYP3A4 activity. Accordingly, the occurrence and outcome of APAP-induced liver injury in an obese individual with NAFLD would depend on a delicate balance between metabolic factors that augment the generation of NAPQI and others that can mitigate hepatotoxicity.
Collapse
|
49
|
Mann JP, Lenz D, Stamataki Z, Kelly D. Common mechanisms in pediatric acute liver failure. Trends Mol Med 2023; 29:228-240. [PMID: 36496278 DOI: 10.1016/j.molmed.2022.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Acute liver failure (ALF) is a rare but potentially fatal disease in children. The etiology is multifactorial, including infection, autoimmune, and genetic disorders, as well as indeterminate hepatitis, which has a higher requirement for liver transplantation. Activation of the innate and adaptive immune systems leads to hepatocyte-specific injury which is mitigated by T regulatory cell activation. Recovery of the native liver depends on activation of apoptotic and regenerative pathways, including the integrated stress response (ISR; e.g., PERK), p53, and HNF4α. Loss-of-function mutations in these pathways cause recurrent ALF in response to non-hepatotropic viruses. Deeper understanding of these mechanisms will lead to improved diagnosis, management, and outcomes for pediatric ALF.
Collapse
Affiliation(s)
- Jake P Mann
- Liver Unit, Birmingham Women's and Children's Hospital, and University of Birmingham, Birmingham, UK
| | - Dominic Lenz
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Zania Stamataki
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Deirdre Kelly
- Liver Unit, Birmingham Women's and Children's Hospital, and University of Birmingham, Birmingham, UK; Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
50
|
Shen XL, Guo YN, Lu MH, Ding KN, Liang SS, Mou RW, Yuan S, He YM, Tang LP. Acetaminophen-induced hepatotoxicity predominantly via inhibiting Nrf2 antioxidative pathway and activating TLR4-NF-κB-MAPK inflammatory response in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114590. [PMID: 36738614 DOI: 10.1016/j.ecoenv.2023.114590] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 06/18/2023]
Abstract
To explore the action time and molecular mechanism underlying the effect of acetaminophen (APAP) on liver injury. APAP was used to establish drug-induced liver injury (DILI) model in mice. Mice in the model group were intraperitoneally injected 300 mg/kg APAP for 6, 12, and 24 h respectively, and control group mice were given the same volume of normal saline. The mice were anesthetized through intravenous injection of sodium pentobarbital at 6, 12, and 24 h after APAP poisoning. Analysis of ALT, AST and ALP in serum, liver histopathological observation, oxidative damage and western blot were performed. The livers in APAP exposed mice were pale, smaller, with a rough texture, and poorly arranged cells. Lesions, large areas of hyperemia, inflammation, swelling, poorly cell arrangement, necrosis, and apoptosis of liver cells were obvious in the liver tissue sections. Serum ALT, AST and ALP levels were significantly enhanced at 12 h of APAP adminstration mice than that of in control group mice (P<0.05). The histopathological alterations and proinflammatory cytokines (IL-1β, TNF-α and IL-6) levels were most severe at 12 h of APAP-induced hepatotoxicity. APAP treatment induced oxidative stress by decreasing hepatic activities of superoxide dismutase (SOD) and glutathione (GSH) (P<0.05), and enhancing malondialdehyde (MDA) content (P<0.05). Moreover, APAP inhibited erythroid 2-related factor 2 (Nrf2) antioxidative pathway with decreased of Nrf2 and HO-1 proteins levels. Furthermore, APAP aggravated the activation of NLRP3 inflammasome by increasing of NLRP3, caspase-1, ASC, IL-1β and IL-18 proteins levels. Finally, APAP further significantly activated the toll-like receptor 4 (TLR4), nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways. This study demonstrated that APAP-induced hepatotoxicity by inhibiting of Nrf2 antioxidative pathway and promoting TLR4-NF-κB-MAPK inflammatory response and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xing-Ling Shen
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Yan-Na Guo
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Meng-Han Lu
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Kang-Ning Ding
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Shao-Shan Liang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Rui-Wei Mou
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Sheng Yuan
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Yong-Ming He
- School of Life Science and Engineering, Foshan University, Foshan 528225, China.
| | - Lu-Ping Tang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China.
| |
Collapse
|